You are on page 1of 809

INDIAN

PHARMACOPOEIA
2007
Volume 3

THE INDIAN PHARMACOPOEIA COMMISSION


GHAZIABAD
INDIAN PHARMACOPOEIA 2007

Volume 3
CONTENTS

Monographs on Drug Substances, Dosage Forms and Pharmaceutical Aids


Monographs to Z ....

Monographs on Vaccines and Immunosera for Human Use ....


Monographs on Herbs and Herbal Products ....
Monographs on Blood and Blood-related Products ....
Monographs on Biotechnology Products ....
Monographs on Veterinary Products ....

Index ....
INDIAN PHARMACOPOEIA 2007 GENERAL NOTICES

GENERAL NOTICES

General Statements ....


Name ....
Official and Official Articles ....
Official Standards ....
Added Substances ....
Alternative Methods ....
Meanings of Terms ....
Provisions Applicable to Monographs and Test Methods ....
Expression of Contents ....
Expression of Concentrations ....
Abbreviated Statements ....
Weights and Measures ....
Monographs ....
General Monographs ....
Production ....
Manufacture of Drug Products ....
Excipients ....
Individual Monographs ....
Titles ....
Chemical Formulae ....
Atomic and Molecular Weights ....
Definitions ....
Statement of Contents ....
Descriptions ....
Identification ....
Tests and Assay ....
Tests ....
Other tests ....
Limits ....
Quantities ....

793
GENERAL NOTICES INDIAN PHARMACOPOEIA 2007

Apparatus ....
Reagents and Solutions ....
Indicators ....
Reference Substances ....
Tests Animals ....
Calculation of Results ....
Storage ....
Storage Containers ....
Labelling ....

794
IP 2007 GENERAL NOTICES

General Notices use but not necessarily to articles that may be sold under the
same name for other purposes.
The active pharmaceutical ingredients (drug substances),
General Statements excipients (pharmaceutical aids), pharmaceutical preparations
The General Notices provide the basic guidelines for the (dosage forms) and other articles described in the monographs
interpretation and application of the standards, tests, assays, are intended for human and veterinary use (unless explicitly
and other specifications of the Indian Pharmacopoeia (IP), as restricted to one of these uses).
well as to the statements made in the monographs and other The requirements given in the monographs are not framed to
texts of the Pharmacopoeia. provide against all possible impurities, contaminants or
A monograph is to be constructed in accordance with any adulterants; they provide appropriate limitation of potential
general monograph or notice or any appendix, note or other impurities only.
explanatory material that is contained in this Pharmacopoeia A preparation must comply throughout the shelf-life assigned
and that is applicable to that monograph. All statements to it by the manufacturer; for opened or broached containers
contained in the monograph, except where a specific general the maximum period of validity for use may sometimes be
notice indicates otherwise and with the exceptions given stated in the individual monograph. Nevertheless, the
hereafter, constitute standards for the official articles. An article responsibility for assigning the period of validity shall be
is not of pharmacopoeial quality unless it complies with all of with the manufacturer.
the requirements stated. Added Substances. An official substance, as distinguished
Exceptions to the General Notices do exist, and where they from an official preparation, contains no added substances
do, the wording in the individual monograph or an appendix except when specifically permitted in the individual monograph.
takes precedence and specifically indicates directions or the Unless otherwise specified in the individual monograph, or
intent. Thus, the specific wording of standards, tests, assays elsewhere in the General Notices, suitable substances may be
and other specifications is binding wherever deviations from added to an official preparation to enhance its stability,
the General Notices exist. Likewise, where there is no specific usefulness or elegance, or to facilitate its preparation. Such
mention to the contrary, the General Notices apply. auxiliary substances shall be harmless in the amounts used,
shall not exceed the minimum quantity required to provide
Name. The full name or title of this book, including addenda
their intended effect, shall not impair the therapeutic efficacy
thereto, is Indian Pharmacopoeia 2007, abbreviated to IP 2007.
or the bioavailability or safety of the preparation and shall not
In the texts, the term “Pharmacopoeia” or “IP” without
interfere with the tests and assays prescribed for determining
qualification means the Indian Pharmacopoeia 2007 and any
compliance with the official standards. Particular care should
addenda thereto.
be taken to ensure that such substances are free from harmful
Official and Official Articles. The word ‘official’ wherever organisms. The freedom to the manufacturers to add auxiliary
used in this Pharmacopoeia or with reference thereto, is substances imposes on them the responsibility of satisfying
synonymous with ‘pharmacopoeial’, with ‘IP’ and with the licensing authorities on the purpose of the addition and
‘compendial’. The designation IP in conjunction with the the innocuity of such substances.
official title on the label of an article is an indication that the Alternative Methods. The tests and assays described are the
article purports to comply with IP standards. official methods upon which the standards of the
The following terms are used where the articles for which Pharmacopoeia are based. Alternative methods of analysis
monographs are provided are to be distinguished. may be used for control purposes, provided that the methods
used are shown to give results of equivalent accuracy and
An official substance is a single drug or a drug entity or a enable an unequivocal decision to be made as to whether
pharmaceutical aid for which the monograph title includes no compliance with the standards of the monographs would be
indication of the nature of a dosage form. achieved if the official methods were used. Automated
An official preparation is a drug product (dosage form) and is procedures utilising the same basic chemistry as the test
the finished or partially finished preparation or product of one procedures given in the monograph may also be used to
or more official substances formulated for use on the patient. determine compliance. Such alternative or automated
procedures must be validated.
An article is an item for which a monograph is provided,
whether an official substance or an official preparation. In the event of doubt or dispute, the methods of analysis of
the Pharmacopoeia are alone authoritative and only the result
Official Standards. The requirements stated in the obtained by the procedure given in this Pharmacopoeia is
monographs apply to articles that are intended for medicinal conclusive.

795
GENERAL NOTICES IP 2007

Meanings of Terms — per cent v/v (percentage, volume in volume) expressing


Alcohol. The term “alcohol” without qualification means the number of millilitres of substance in 100 millilitres of
ethanol (95 per cent). Other dilutions of ethanol are indicated final product.
by the term “alcohol” or “alcohol” followed by a statement of The expression “parts per million” refers to the weight in
the percentage by volume of ethanol (C2H6O) required. weight, unless otherwise stated.
Desiccator. A tightly-closed container of suitable size and Where the content of a substance is expressed in terms of the
design that maintains an atmosphere of low moisture content chemical formula for that substance an upper limit exceeding
by means of silica gel or phosphorus pentoxide or other 100 per cent may be stated. Such an upper limit applies to the
suitable desiccant. result of the assay calculated in terms of the equivalent content
of the specified chemical formula. For example, the statement
Drying and ignition to constant weight. Two consecutive
‘contains not less than 99.0 per cent and not more than 101.0
weighings after the drying or igniting operations do not differ
per cent of C7H6O2 implies that the result of the assay is not
by more than 0.5 mg, the second weighing following an
less than 99.0 per cent and not more than 101.0 per cent,
additional period of drying or of ignition respectively
calculated in terms of the equivalent content of C7H6O2.
appropriate to the nature and quantity of the residue.
Where the result of an assay or test is required to be calculated
Ethanol. The term “ethanol” without qualification means
with reference to the dried, anhydrous, ignited substance, or
anhydrous ethanol or absolute alcohol.
the substance free from solvent, the determination of loss on
Filtration. Unless otherwise stated, filtration is the passing of drying, water content, loss on ignition, content of the specified
a liquid through a suitable filter paper or equivalent device solvent, respectively is carried out by the method prescribed
until the filtrate is clear. in the relevant test in the monograph.
Freshly prepared. Made not more than 24 hours before it is Expression of Concentrations. The following expressions in
issued for use. addition to the ones given under Expression of Content are
also used:
Label. Any printed packing material, including package inserts
that provide information on the article. — per cent w/v (percentage, weight in volume) expressing
the number of grams of substance in 100 millilitres of
Negligible. A quantity not exceeding 0.50 mg. product
Solution. Where the name of the solvent is not stated, — per cent v/w (percentage, volume in weight) expressing
“solution” implies a solution in water. The water used complies the number of millilitres of substance in 100 grams of
with the requirements of the monograph on Purified Water. product.
The term ‘distilled water’ indicates Purified Water prepared by
distillation. Usually, the strength of solutions of solids in liquids is
expressed as percentage weight in volume, of liquids in liquids
Temperature. The symbol º used without qualification as percentage volume in volume, of solids in semi-solid bases
indicates the use of the Celsius thermometric scale. (e.g. creams) and of gases in liquids as percentage weight in
Water. If the term is used without qualification it means Purified weight.
Water of the Pharmacopoeia. The term ‘distilled water’ When the concentration of a solution is expressed as parts of
indicates Purified Water prepared by distillation. dissolved substance in parts of solution, it means parts by
weight (g) of a solid in parts by volume (ml) of the final solution;
Water-bath. A bath of boiling water unless water at another
as parts by weight (g) of a gas in parts by weight (g) of the
temperature is indicated. Other methods of heating may be
final solution.
used provided the required temperature is approximately
maintained but not exceeded. When the concentration of a solution is expressed in molarity
designated by the symbol M preceded by a number, it denotes
Provisions Applicable To Monographs and Test Methods the number of moles of the stated solute contained in sufficient
Purified Water (unless otherwise stated) to produce 1 litre of
Expression of Content. Where the content of a substance is solution.
defined, the expression “per cent” is used according to
circumstances with one of two meanings: Abbreviated Statements. Incomplete sentences are employed
in parts of the monographs for directness and brevity (for
— per cent w/w (percentage, weight in weight) expressing example, Iodine Value. Not more than ……; Relative Density.
the number of grams of substance in 100 grams of final …….to……..) Where the tests are abbreviated, it is to be
product, understood that the test method referred to in brackets

796
IP 2007 GENERAL NOTICES

provides the method to be followed and that the values Excipients. Any substance added in preparing an official
specified are the applicable limits. preparation shall be innocuous, shall have no adverse influence
in the therapeutic efficacy of the active ingredients and shall
Weights and Measures. The metric system of weights and
not interfere with the tests and assays of the Pharmacopoeia.
measures is employed in the Pharmacopoeia. All measures are
Care should be taken to ensure that such substances are free
required to be graduated at 25º and all measurements in tests
from harmful organisms.
and assays, unless otherwise stated, are to be made at that
temperature. Graduated glass apparatus used in analytical Individual Monographs
operations shall comply with the requirements stated in
Chapter 2.1.6 Drug products that are the subject of an individual monograph
are also required to comply with the tests given in the general
monographs.
Monographs
Titles. The main title for a drug substance is the International
General Monographs Non-proprietary Name (INN) approved by the World Health
Organization. Subsidiary names and synonyms have also been
General monographs on dosage forms include requirements given in some cases; where included, they have the same
of general application and apply to all preparations within the significance as the main title.
scope of the Introduction section of the general monograph,
except where a preamble limits the application. The The main titles of drug products are the ones commonly
requirements are not necessarily comprehensive for a given recognised in practice. Synonyms drawn from the full non-
specific preparation; additional requirements may sometimes proprietary name of the active ingredient or ingredients have
be given in the individual monograph for it. also been given. Where, however, a product contains one or
the other of different salts of an active molecule, the main title
Production. Statements given under the heading Production is based on the full name of the active ingredient. For example,
relate to particular aspects of the manufacturing process and Chloroquine Phosphate Tablets and Chloroquine
are not necessarily comprehensive. However, they are SulphateTablets.
mandatory instructions to manufacturers. They may relate,
for example, to source materials, to the manufacturing process Chemical Formulae. When the chemical structure of an official
and its validation and control, to any in-process testing that substance is known or generally accepted, the graphic and
is to be carried out by the manufacturer on the final product molecular formulae are normally given at the beginning of the
either on selected batches or on each batch prior to release. monograph for information. This information refers to the
All this cannot be verified on a sample of the final product by chemically pure substance and is not to be regarded as an
an independent analyst. It is for the licensing authority to indication of the purity of the official material. Elsewhere, in
verify that the instructions have been followed. statement of purity and strength and in descriptions of
processes of assay, it will be evident from the context that the
The absence of a section on Production does not imply that formulae denote the chemically pure substances.
attention to features such as those given above is not required.
An article described in a monograph of the Pharmacopoeia is Where the absolute stereochemical configuration is specified,
to be manufactured in accordance with the principles of good the International Union of Pure and Applied Chemistry
manufacturing practice and in accordance with the (IUPAC) R/S and E/Z systems of designation have been used.
requirements of the Drugs and Cosmetics Rules, 1945. The If the substance is an enantiomer of unknown absolute
general principles applicable to the manufacture and quality stereochemistry, the sign of the optical rotation, as determined
assurance of drugs and preparations meant for human use in the solvent and under the conditions specified in the
apply equally to veterinary products as well. monograph, has been attached to the systematic name. An
indication of sign of rotation has also been given where this is
Manufacture of Drug Products. The opening definitive incorporated in a trivial name that appears on an IUPAC
statement in certain monographs for drug products is given in preferred list.
terms of the active ingredient(s) only. Any ingredient(s) other
than those included in the statement, must comply with the Atomic and Molecular Weights. The atomic weight or
general notice on Excipients and the product must conform to molecular weight is shown , as and when appropriate at the
the Pharmacopoeial requirements. top right hand corner of the monograph. The atomic and
molecular weights and graphic formulae do not constitute
Official preparations are prepared only from ingredients that analytical standards for the substances described.
comply with the requirements of the pharmacopoeial
monographs for those individual ingredients for which Definition. The opening statement of a monograph is one
monographs are provided. that constitutes an official definition of the substance,

797
GENERAL NOTICES IP 2007

preparation or other article that is the subject of the are not framed to take into account all possible impurities. It is
monograph. In certain monographs for pharmaceutical not to be presumed, for example, that an impurity that is not
preparations the statement is given in terms of the principal detectable by means of the prescribed tests is tolerated.
ingredient(s). Material found to contain such an impurity is not of
In monographs on vegetable drugs, the definition indicates pharmacopoeial quality if the nature or amount of the impurity
whether the subject of the monograph is, for example, the found is incompatible with good pharmaceutical practice.
whole drug or the drug in powdered form. Pharmacopoeial methods and limits should be used merely as
Certain pharmaceutical substances and other articles are compliance requirements and not as requirements to guarantee
defined by reference to a particular method of manufacture. A total quality assurance. Tests and assays are prescribed for
statement that a substance or article is prepared or obtained the minimum sample available on which the attributes of the
by a certain method constitutes part of the official definition article should be measured. Assurance of quality must be
and implies that other methods are not permitted. A statement ensured by the manufacturer by the use of statistically valid
that a substance may be prepared or obtained by a certain sampling and testing programmes.
method, however, indicates that this is one possible method Tests. Unless otherwise stated, the assays and tests are carried
and does not imply that other methods are not permissible. out at a temperature between 20º and 30º.
Statement of content. The limits of content stated are those Where it is directed that an analytical operation is to be carried
determined by the method described under Assay. out ‘in subdued light’, precautions should be taken to avoid
Description. The statements under the heading Description exposure to direct sunlight or other strong light. Where a
are not to be interpreted in a strict sense and are not to be procedure is directed to be performed ‘protected from light’
regarded as official requirements. precautions should be taken to exclude actinic light by the
Solubility. Statements on solubility are given in Chapter 2.4.26 use of low-actinic glassware, working in a dark room or similar
and are intended as information on the approximate solubility procedures.
at a temperature between 15º and 30º, unless otherwise stated, For preparations other than those of fixed strength, the
and are not to be considered as official requirements. However, quantity to be taken for a test or an assay is usually expressed
a test for solubility stated in a monograph constitutes part of in terms of the active ingredient. This means that the quantity
the standards for the substance that is the subject of that of the active ingredient expected to be present and the quantity
monograph. of the preparation to be taken are calculated from the strength
stated on the label.
Test Methods
Other Tests. In the monographs on dosage forms and certain
References to general methods of testing are indicated by test preparations, under the sub-heading ‘Other tests’ it is stated
method numbers in brackets immediately after the heading of that the article complies with the tests stated under the general
the test or at the end of the text. monograph of the relevant dosage form or preparation. Details
Identification. The tests given under the heading Identification of such tests are provided in the general monographs.
are not necessarily sufficient to establish absolute proof of Limits. The limits given are based on data obtained in normal
identity. They provide a means of verifying that the identity analytical practice. They take into account normal analytical
of the material under examination is in accordance with the errors, of acceptable variations in manufacture and of
label on the container. deterioration to an extent that is acceptable. No further
In certain monographs alternative series of identification tests tolerances are to be applied to the limits for determining whether
are given; compliance with either one or the other set of tests or not the article under examination complies with the
is adequate to verify the identity of the article. requirements of the monograph.
When tests for infrared absorption are applied to material Quantities. Unless otherwise stated, the quantities to be taken
extracted from formulated preparations, strict concordance for assays, limit tests and other tests are of the substance
with the specified reference spectrum may not always be under examination.
possible, but nevertheless a close resemblance between the In tests with numerical limits and assays, the quantity stated
spectrum of the extracted material and the specified reference to be taken for testing is approximate. The amount actually
spectrum should be achieved. used, which may deviate by not more than 10 per cent from
that stated, is accurately weighed or measured and the result
Tests and Assays
of analysis is calculated from this exact quantity. In tests where
The tests and assays are the official methods upon which the the limit is not numerical but usually depends upon
standards of the Pharmacopoeia depend. The requirements comparison with the behaviour of a reference in the same

798
IP 2007 GENERAL NOTICES

conditions, the stated quantity is taken for testing. Reagents Indian Pharmacopoeia Commission (IPC). They are the official
are used in the prescribed amounts. standards to be used in cases of arbitration. Secondary
Quantities are weighed or measured with an accuracy Standards (Working Standards) may be used for routine
commensurate with the indicated degree of precision. For analysis, provided they are standardized at regular intervals
weighings, the precision is plus or minus 5 units after the last against the Reference Substances
figure stated. For example, 0.25 g is to be interpreted as 0.245 Biological Reference Substances, also abbreviated to IPRS
g to 0.255 g. For the measurement of volumes, if the figure and Standard Preparations of antibiotics are issued by
after the decimal point is a zero or ends in a zero, e.g. 10.0 ml 0r agencies authorised by the IPC. They are standardized against
0.50 ml, the volume is measured using a pipette, a volumetric the International Standards and Reference Preparations
flask or a burette, as appropriate; in other cases, a graduated established by the World Health Organization (WHO). The
measuring cylinder or a graduated pipette may be used. potency of these preparations is expressed in International
Volumes stated in microlitres are measured using a micropipette Units.
or microsyringe.
Reference spectra are published by the IPC and they are
The term ‘transfer’ is used generally to indicate a quantitative accompanied by information concerning the conditions used
operation. for sample preparation and recording of the spectra.
Apparatus. Measuring and weighing devices and other Test animals. Unless otherwise directed, animals used in a
apparatus are described in the chapter entitled ‘Apparatus for test or an assay shall be healthy and are drawn from a uniform
Tests and Assays’. A specification for a definite size or type stock, and have not previously been treated with any material
of container or apparatus in a test or assay is given merely as that will interfere with the test or the assay.
a recommendation. Calculation of results. In determining compliance with a
Unless otherwise stated, comparative tests are carried out numerical limit in assay or test, the result should be calculated
using identical tubes of colourless, transparent, neutral glass to one decimal place more than the significant figures stated
with a flat base, commonly known as Nessler cylinders. and then rounded up or down as follows: if the last figure
calculated is 5 to 9, the preceding figure is increased by 1; if it
Reagents and Solutions. The reagents required for the tests
is 4 or less, the preceding figure is left unchanged.
and assays of the Pharmacopoeia are defined in the various
chapters showing their nature, degree of purity and the Storage. Statements under the side-heading Storage constitute
strengths of the solutions to be made from them. The non-mandatory advice. The articles of the Pharmacopoeia are
requirements set out are not intended to imply that the materials to be stored under conditions that prevent contamination and,
are suitable for use in medicine; regents not covered by as far as possible, deterioration. Precautions that should be
monographs in the pharmacopoeia shall not be claimed to be taken in relation to the effects of the atmosphere, moisture,
of IP quality. heat and light are indicated, where appropriate, in the individual
monograph.
The term ‘analytical reagent grade of commerce’ implies that
the chemical is of a high degree of purity wherein the limits of Specific directions are given in some monographs with respect
various impurities are known. Where it is directed to use a to the temperatures at which Pharmacopoeial articles should
‘general laboratory reagent grade of commerce’ it is intended be stored, where it is considered that usage at a lower or
that a chemically pure grade material, not necessarily required higher temperature may produce undesirable results. The
to be tested for limiting or absence of certain impurities, is to storage conditions are defined by the following terms:
be used. — Store in a dry, well-ventilated place at a temperature not
Indicators. Where the use of an indicator solution is mentioned exceeding 30º
in an assay or test, approximately 0.1 ml of the solution shall — Store in a refrigerator (2º to 8º). Do not freeze
be added, unless otherwise directed.
— Store in a freezer (-2º to -18º)
Reference Substances. Certain monographs require the use — Store in a deep freezer (Below -18º)
of a chemical reference substance or a biological reference
preparation or a reference spectrum These are authentic Storage conditions not related to temperature are indicated in
specimens chosen and verified on the basis of their suitability the following terms:
for intended use as prescribed in the Pharmacopoeia and are — Store protected from light
not necessarily suitable in other circumstances. — Store protected from light and moisture
IP Reference Substances, abbreviated to IPRS (and referred Where no specific storage directions or limitations are given
to as RS in the individual monographs) are issued by the in the monograph or by the manufacturer, it is to be understood

799
GENERAL NOTICES IP 2007

that the storage conditions include protection from moisture, of being tightly closed, and re-closed after use.
freezing and excessive heat (any temperature above 40º).
In certain cases, special requirements of pack have been
Storage Containers. The requirements, guidance and indicated in some monographs under Storage, using
information on containers for pharmaceutical use are given in expressions that have been defined in chapter 6.1.
the chapter entitled Containers (6.1) Labelling. The labelling of drugs and pharmaceuticals is
In general, an article should be packed in a well-closed governed by the Drugs and Cosmetics Rules, 1945. The
container i.e. one that protects the contents from statements that are given in the monographs under the side-
contamination by extraneous solids, liquids or vapours and heading ‘Labelling’ are not comprehensive. Only those that
from loss of the article under normal conditions of handling are necessary to demonstrate compliance or otherwise with
and storage. the monograph have been given and they are mandatory. For
example, in the monograph on Betamethasone Sodium Tablets
Where, additionally, loss or deterioration of the article from
the labelling statement is “The label states the strength in
effervescence, deliquescence or evaporation under normal
terms of the equivalent amount of betamethasone”. Any other
conditions of storage is likely, the container must be capable
statements are included as recommendations.

800
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

DRUG SUBSTANCES, DOSAGE FORMS


AND
PHARMACEUTICAL AIDS

N to Z ..................................................................................................................

801
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

N
Nalidixic Acid ....
Nalidixic Acid Tablets ....
Nalorphine Hydrochloride ....
Nalorphine Injection ....
Nandrolone Decanoate ....
Nandrolone Decanoate Injection ....
Nandrolone Phenylpropionate ....
Nandrolone Phenylpropionate Injection ....
Naphazoline Nitrate ....
Nelfinavir Mesylate ....
Nelfinavir Mesylate Oral Powder ....
Nelfinavir Tablets ....
Neomycin Sulphate ....
Neomycin Eye Drops ....
Neomycin Eye Ointment ....
Neostigmine Bromide ....
Neostigmine Tablets ....
Neostigmine Methylsulphate ....
Neostigmine Injection ....
Nevirapine ....
Nevirapine Oral Suspension ....
Nevirapine Tablets ....
Niclosamide ....
Niclosamide Tablets ....
Nicotinamide ....
Nicotinamide Tablets ....
Nicotinic Acid ....
Nicotinic Tablets ....
Nicoumalone ....
Nicoumalone Tablets ....

803
MONOGRAPHS INDIAN PHARMACOPOEIA 2007 2007

Nifedipine ....
Nifedipine Capsules ....
Nifedipine Sustained release-Tablets ....
Nifedipine Tablets ....
Nikethamide ....
Nikethamide Injection ....
Nitrazepam ....
Nitrazepam Tablets ....
Nitrofurantoin ....
Nitrofurantoin Tablets ....
Nitrofurazone ....
Nitrous Oxide ....
Noradrenaline Bitartrate ....
Noradrenaline Bitartrate Injection ....
Norethisterone ....
Norethisterone Tablets ....
Norfloxacin ....
Norfloxacin Eye Drops ....
Norfloxacin Tablets ....
Norgestrel ....
Norgestrel And Ethinyloestradiol Tablets ....
Nortriptyline Hydrochloride ....
Nortriptyline Tablets ....
Noscapine ....
Noscapine Linctus ....
Novobiocin Sodium ....
Nystatin ....
Nystatin Ointment ....
Nystatin Pessaries ....
Nystatin Tablets ....

804
IP 2007 NALIDIXIC ACID TABLETS

Nalidixic Acid Reference solution (b). A 0.0008 per cent w/v solution of the
substance under examination in dichloromethane.
CH3 Reference solution (c). A 0.1 per cent w/v solution of nalidixic
acid RS in dichloromethane.
H3C N N Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
COOH Any secondary spot in the chromatogram obtained with test
O solution (a) is not more intense than the spot in the
chromatogram obtained with reference solution (a) and not
more than one such spot is more intense than the spot in the
C12H12N2O3 Mol. Wt. 232.2
chromatogram obtained with reference solution (b).
Nalidixic Acid is 1-ethyl-1,4-dihydro-7-methyl-4-oxo-1,8-
naphthyridine-3-carboxylic acid. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Nalidixic Acid contains not less than 99.0 per cent and not
more than 101.0 per cent of C12H12N2O3, calculated on the Sulphated ash (2.3.18) Not more than 0.1 per cent.
dried basis. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Description. A white to slightly yellow, crystalline powder. on 1.0 g by drying in an oven at 105°.
Assay. Weigh accurately about 0.15 g, dissolve in 10 ml of
Identification dichloromethane, add 30 ml of 2-propanol and 10 ml of carbon
dioxide-free water and titrate with 0.1 M ethanolic sodium
Test A may be omitted if tests B, C and D are carried out. Tests
hydroxide, determining the end-point potentiometrically
B, C and D may be omitted if test A is carried out.
(2.4.25) and using a glass electrode as the indicator electrode
A. Determine by infrared absorption spectrophotometry (2.4.6). and a silver-silver chloride reference electrode with a sleeve
Compare the spectrum with that obtained with nalidixic acid diaphragm or a capillary tip filled with a saturated solution of
RS or with the reference spectrum of nalidixic acid. lithium chloride in ethanol. Throughout the titration keep
B. When examined in the range 230 nm to 360 nm (2.4.7), a the temperature of the solution at 15° to 20° and pass a current
0.0005 per cent w/v solution in 0.1 M sodium hydroxide shows of nitrogen through the solution.
absorption maxima at about 258 nm and 334 nm; ratio of the 1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
absorbance at about 258 nm to that at about 334 nm, 2.2 to 2.4. 0.02322 g of C12H12N2O3.
C. In the test for Related substances, the principal spot in the Storage. Store protected from light and moisture.
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution (c).
D. Dissolve 0.1 g in 2 ml of hydrochloric acid and add 0.5 ml
of a 10 per cent w/v solution of 2-naphthol in ethanol (95 per Nalidixic Acid Tablets
cent); an orange-red colour develops.
Nalidixic Acid Tablets contain not less than 95.0 per cent and
Tests not more than 105.0 per cent of the stated amount of nalidixic
acid, C12H12N2O3.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel HF254. Identification
Mobile phase. A mixture of 70 volumes of ethanol (95 per To a quantity of the powdered tablets containing 1 g of Nalidixic
cent), 20 volumes of dichloromethane and 10 volumes of 5 M Acid add 50 ml of chloroform, shake for 15 minutes, filter and
ammonia. evaporate the filtrate to dryness. The residue, after drying at
Test solution (a). Dissolve 0.2 g of the substance under 105°, complies with the following tests.
examination in 10 ml of dichloromethane. A. Determine by infrared absorption spectrophotometry (2.4.6).
Test solution (b). A 0.1 per cent w/v solution of the substance Compare the spectrum with that obtained with nalidixic acid
under examination in dichloromethane. RS or with the reference spectrum of nalidixic acid.
Reference solution (a). A 0.002 per cent w/v solution of the B. When examined in the range 230 nm to 360 nm (2.4.7), a
substance under examination in dichloromethane. 0.0005 per cent w/v solution in 0.1 M sodium hydroxide shows

805
NALORPHINE HYDROCHLORIDE IP 2007

absorption maxima at about 258 nm and 334 nm; ratio of the Nalorphine Hydrochloride contains not less than 97.0 per cent
absorbance at about 258 nm to that at about 334 nm, 2.2 to 2.4. and not more than 103.0 per cent of C19H21NO3,HCl, calculated
on the dried basis.
Tests
Description. A white or almost white, crystalline powder;
Related substances. Determine by thin-layer chromatography odourless. It slowly darkens on exposure to air and light.
(2.4.17), coating the plate with silica gel HF254.
Identification
Mobile phase. A mixture of 70 volumes of ethanol (95 per
cent), 20 volumes of dichloromethane and 10 volumes of Test A may be omitted if tests B, C, D and E are carried out.
5 M ammonia. Tests C and D may be omitted if tests A, B and E are carried
out.
Test solution. Shake a quantity of the powdered tablets
containing 0.1 g of Nalidixic Acid with 50 ml of chloroform for A. Determine by infrared absorption spectrophotometry (2.4.6).
15 minutes, filter, evaporate the filtrate to dryness and dissolve Compare the spectrum with that obtained with nalorphine
the residue in 5 ml of chloroform. hydrochloride RS.
Reference solution. Dilute 1 volume of the test solution to B. When examined in the range 230 nm to 360 nm (2.4.7), a
200 volumes with chloroform. 0.01 per cent w/v solution in 0.1 M sodium hydroxide shows
Apply to the plate 10 µl of each solution. After development, an absorption maximum only at about 298 nm; absorbance at
dry the plate in air and examine in ultraviolet light at 254 nm. about 298 nm, about 0.6.
Any secondary spot in the chromatogram obtained with the C. To 10 ml of a 2 per cent w/v solution add 0.05 ml of dilute
test solution is not more intense than the spot in the ammonia solution; a white precipitate soluble in sodium
chromatogram obtained with the reference solution. hydroxide solution is produced.
Other Tests. Complies with the tests stated under Tablets. D. Dissolve 2 mg in 2 ml of water, add 0.15 ml of potassium
Assay. Weigh and powder 20 tablets. Weigh accurately a ferricyanide solution containing, in each ml, 0.05 ml of ferric
quantity of the powder containing about 0.1 g of Nalidixic chloride solution; a deep bluish green colour is produced
Acid, add 150 ml of 0.1 M sodium hydroxide, shake for 3 immediately.
minutes, dilute to 200.0 ml with 0.1 M sodium hydroxide, mix E. Gives reaction A of chlorides (2.3.1).
and allow to stand for 15 minutes. Dilute 2.0 ml of the solution
to 100.0 ml with water and measure the absorbance of the Tests
resulting solution at the maximum at about 334 nm (2.4.7),
using 0.1 M sodium hydroxide as the blank. Calculate the Melting range (2.4.21). 260° to 263°.
content of C12H12N2O3 taking 494 as the specific absorbance Acidity. Dissolve 0.2 g in 10 ml of freshly boiled and cooled
at 334 nm. water and titrate with 0.02 M sodium hydroxide using methyl
Storage. Store protected from light and moisture. red solution as indicator; not more than 0.2 ml of 0.02 M
sodium hydroxide is required to change the colour of the
solution.
Specific optical rotation (2.4.22). –122° to –125°, determined
Nalorphine Hydrochloride in a 2.0 per cent w/v solution.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
HO
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 100° at a pressure not exceeding
O 0.7 kPa for 2 hours.
N , HCl
Assay. Weigh accurately about 25 mg and dissolve in sufficient
H CH2
water to produce 250 ml. Measure the absorbance of the
HO resulting solution at the maximum at about 285 nm (2.4.7).
Calculate the content of C19H21NO3,HCl from the absorbance
C19H21NO3,HCl Mol. Wt. 347.8 obtained by repeating the operation with nalorphine
Nalorphine Hydrochloride is 17-allyl-7,8-didehydro-4,5α- hydrochloride RS in place of the substance under
epoxymorphinan-3,6α-diol hydrochloride. examination.

806
IP 2007 NANDROLONE DECANOATE

Storage. Store protected from light and moisture. Nandrolone Decanoate

O
Nalorphine Injection
H3C O CH2(CH2)7CH3
Nalorphine Hydrochloride Injection
Nalorphine Injection is a sterile solution of Nalorphine H H
Hydrochloride in Water for Injections containing suitable
H H
buffering agents.
O
Nalorphine Injection contains not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of nalorphine
hydrochloride, C19H21NO3,HCl. C28H44O3 Mol. Wt. 428.7
Nandrolone Decanoate is 3-oxo-4-estren-17β-yl decanoate.
Identification Nandrolone Decanoate contains not less than 97.0 per cent
A. To a volume containing 50 mg of Nalorphine Hydrochloride and not more than 103.0 per cent of C28H44O3, calculated on
add dilute ammonia solution until the solution is alkaline and the dried basis.
extract with 25 ml of a mixture of 1 volume of ethanol (95 per Description. A white to creamy-off white, crystalline powder;
cent) and 3 volumes of chloroform and evaporate the extract odour, faint and characteristic.
to dryness. Dry the residue at a pressure not exceeding 2 kPa.
The residue complies with the following test. Identification
Determine by infrared absorption spectrophotometry (2.4.6). A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with nalorphine Compare the spectrum with that obtained with nandrolone
hydrochloride RS. decanoate RS or with the reference spectrum of nandrolone
B. To a volume containing 0.1 g of Nalorphine Hydrochloride decanoate.
add 0.05 ml of dilute ammonia solution; a white precipitate B. When examined in the range 230 nm to 360 nm (2.4.7), a
soluble in sodium hydroxide solution is produced. 0.001 per cent w/v solution in ethanol (95 per cent) shows an
absorption maximum only at about 239 nm; absorbance at
C. Gives reaction A of chlorides (2.3.1).
about 239 nm, about 0.41.
Tests C. Dissolve 25 mg in 1 ml of methanol, add 2 ml of
semicarbazide acetate solution, heat under a reflux condenser
pH (2.4.24). 6.0 to 7.5. for 30 minutes and cool; the precipitate, after recrystallisation
Other Tests. Complies with the tests stated under Parenteral from ethanol (95 per cent), melts at about 175° (2.4.21).
Preparations (Injections).
Tests
Assay. Transfer an accurately measured volume containing
about 10 mg of Nalorphine Hydrochloride to a separating Specific optical rotation (2.4.22). +32.0 ° to +36.0°, determined
funnel, add 1 ml of dilute hydrochloric acid and dilute to in a 2.0 per cent w/v solution in dioxan.
10 ml with water. Extract with five successive quantities, each Related substances. Determine by thin-layer chromatography
of 5 ml, of chloroform, allowing the layers to separate before (2.4.17), coating the plate with silica gel GF254.
drawing off each chloroform extract and discard the chloroform
Mobile phase. A mixture of 70 volumes of heptane and
extracts. Transfer the aqueous layer to a 100-ml volumetric
30 volumes of acetone.
flask with the aid of small quantities of water and dilute to
volume with water. Measure the absorbance of the resulting Test solution. Dissolve 0.1 g of the substance under
solution at the maximum at about 285 nm (2.4.7). Calculate examination in 10 ml of chloroform.
the content of C19H21NO3,HCl from the absorbance obtained Reference solution (a). A 0.005 per cent w/v solution of the
by repeating the operation with nalorphine hydrochloride substance under examination in chloroform.
RS.
Reference solution (b). A 0.01 per cent w/v solution of
Storage. Store protected from light. nandrolone RS in chloroform.

807
NANDROLONE DECANOATE INJECTION IP 2007

Apply to the plate 5 µl of each solution. After development, Tests


dry the plate in air and examine in ultraviolet light at 254 nm. In
the chromatogram obtained with the test solution any spot Other Tests. Complies with the tests stated under Parenteral
corresponding to nandrolone is not more intense than the Preparations (Injections).
spot in the chromatogram obtained with reference solution Assay. To an accurately measured volume containing about
(b) and any other secondary spot is not more intense than the 0.1 g of Nandrolone Decanoate add sufficient chloroform to
spot in the chromatogram obtained with reference solution produce 100.0 ml. Dilute 3.0 ml of the solution to 50.0 ml with
(a). chloroform. To 5.0 ml of this solution add 10 ml of isoniazid
Sulphated ash (2.3.18). Not more than 0.1 per cent. solution and sufficient methanol to produce 20.0 ml. Allow to
stand for 45 minutes and measure the absorbance of the
Loss on drying (2.4.19). Not more than 0.5 per cent, determined resulting solution at the maximum at about 380 nm (2.4.7),
on 1.0 g by drying over phosphorus pentoxide at a pressure using as the blank 5 ml of chloroform treated in the same
not exceeding 0.7 kPa for 4 hours. manner. Calculate the content of C28H44O3 from the absorbance
Assay. Weigh accurately about 10 mg and dissolve in sufficient obtained by repeating the operation using a suitable quantity
ethanol (95 per cent) to produce 100.0 ml. Dilute 5.0 ml to of nandrolone RS.
50.0 ml with ethanol (95 per cent) and measure the absorbance 1 mg of C18H26O2 is equivalent to 1.562 mg of C28H44O3.
of the resulting solution at the maximum at about 239 nm (2.4.7).
Calculate the content of C28H44O3 taking 407 as the specific Storage. Store protected from light.
absorbance at 239 nm.
Storage. Store protected from light and moisture.
Nandrolone Phenylpropionate
Nandrolone Phenpropionate
Nandrolone Decanoate Injection O
Nandrolone Decanoate Injection is a sterile solution of
Nandrolone Decanoate in Ethyl Oleate or other suitable ester, H3C O
in a suitable fixed oil or in any mixture of these.
H H
Nandrolone Decanoate Injection contains not less than 90.0
per cent and not more than 110.0 per cent of the stated amount H H
of nandrolone decanoate, C28H44O3. O
Identification C27H34O3 Mol.Wt. 406.6
Determine by thin-layer chromatography (2.4.17), coating the Nandrolone Phenylpropionate is 3-oxo-4-estren-17β-yl 3-
plate with silica gel GF254. phenylpropionate.
Mobile phase. A mixture of 70 volumes of heptane and Nandrolone Phenylpropionate contains not less than 97.0 per
30 volumes of acetone. cent and not more than 103.0 per cent of C27H34O3, calculated
Test solution. Dilute a suitable volume of the injection with on the dried basis.
carbon tetrachloride to give a solution containing 0.5 per- Description. A white to creamy-white, crystalline powder;
cent w/v solution of Nandrolone Decanoate. odour, characteristic.
Reference solution. A 0.5 per cent w/v solution of nandrolone
Identification
decanoate RS in carbon tetrachloride.
Apply to the plate 5 µl of each solution. After development, A. Determine by infrared absorption spectrophotometry (2.4.6).
dry the plate in air until the odour of solvent is no longer Compare the spectrum with that obtained with nandrolone
detectable, spray with a 10 per cent v/v solution of sulphuric phenylpropionate RS or with the reference spectrum of
acid in ethanol (95 per cent), heat at 105° for 30 minutes and nandrolone phenylpropionate.
examine in ultraviolet light at 365 nm. The principal spot in the B. When examined in the range 230 nm to 360 nm (2.4.7), a
chromatogram obtained with the test solution corresponds to 0.001 per cent w/v solution in ethanol (95 per cent) shows an
that in the chromatogram obtained with the reference solution. absorption maximum only at about 240 nm; absorbance at
Ignore any subsidiary spots due to the vehicle. about 240 nm, about 0.43.

808
IP 2007 NANDROLONE PHENYLPROPIONATE INJECTION

C. Dissolve 25 mg in 1 ml of methanol, add 2 ml of Identification


semicarbazide acetate solution, heat under a reflux condenser
for 30 minutes and cool; the precipitate, after recrystallisation Dissolve a volume of the injection containing 50 mg of
from ethanol (95 per cent) melts at about 182° (2.4.21). Nandrolone Phenylpropionate in 8 ml of light petroleum
(40° to 60°) and extract with three 8-ml quantities of a mixture
Tests of 7 volumes of glacial acetic acid and 3 volumes of water.
Wash the combined extracts with 10 ml of light petroleum
Specific optical rotation (2.4.22). +48.0 ° to +51.0°, determined (40° to 60°), dilute with water until the solution becomes
in a 1.0 per cent w/v solution in dioxan. turbid, allow to stand for 2 hours in ice and filter. The precipitate,
Related substances. Determine by thin-layer chromatography after washing with water and drying over phosphorus
(2.4.17), coating the plate with silica gel GF254. pentoxide at a pressure not exceeding 0.7 kPa, complies with
the following test.
Mobile phase. A mixture of 70 volumes of heptane and
30 volumes of acetone. Determine by thin-layer chromatography (2.4.17), using a
silica gel GF254 precoated plate the surface of which has
Test solution. Dissolve 0.1 g of the substance under
been modified by chemically-bonded octadecylsilyl groups.
examination in 100 ml of chloroform.
Mobile phase. A mixture of 20 volumes of water, 40 volumes
Reference solution (a). A 0.005 per cent w/v solution of the of acetonitrile and 60 volumes of propan-2-ol.
substance under examination in chloroform.
Test solution. A 0.5 per cent w/v solution of the dried
Reference solution (b). A 0.01 per cent w/v solution of
precipitate in chloroform.
nandrolone RS in chloroform.
Reference solution (a). A 0.5 per cent w/v solution of
Apply to the plate 5 µl of each solution. After development,
nandrolone phenylpropionate RS in chloroform.
dry the plate in air and examine in ultraviolet light at 254 nm. In
the chromatogram obtained with the test solution any spot Reference solution (b). A mixture of equal volumes of the test
corresponding to nandrolone is not more intense than the solution and the reference solution.
spot in the chromatogram obtained with reference solution Apply to the plate 5 µl of each solution. After development,
(b) and any other secondary spot is not more intense than the dry the plate in air until the solvent has evaporated and heat it
spot in the chromatogram obtained with reference solution at 100° for 10 minutes. Allow to cool and examine in ultraviolet
(a). light at 254 nm. The principal spot in the chromatogram
Sulphated ash (2.3.18). Not more than 0.1 per cent. obtained with the test solution corresponds to that in the
chromatogram obtained with reference solution (a). The
Loss on drying (2.4.19) Not more than 0.5 per cent, determined
principal spot in the chromatogram obtained with reference
on 1.0 g by drying over phosphorus pentoxide at a pressure
solution (b) appears as a single spot.
not exceeding 0.7 kPa for 4 hours.
Assay. Weigh accurately about 10 mg, dissolve in sufficient Tests
ethanol to produce 100.0 ml, dilute 5.0 ml to 50.0 ml with ethanol
Other tests. Complies with the tests stated under Parenteral
and measure the absorbance of the resulting solution at the
Preparations (Injections).
maximum at about 240 nm (2.4.7). Calculate the content of
C27H34O3 taking 430 as the specific absorbance at 240 nm. Assay. To an accurately measured volume containing about
0.1 g of Nandrolone Phenylpropionate add sufficient
Storage. Store protected from light.
chloroform to produce 100.0 ml. Dilute 3.0 ml of this solution
to 50.0 ml with chloroform. To 5.0 ml of the resulting solution
add 10 ml of isoniazid solution and sufficient methanol to
produce 20.0 ml. Allow to stand for 45 minutes and measure
Nandrolone Phenylpropionate the absorbance of the solution at the maximum at about
Injection 380 nm (2.4.7), using as blank 5 ml of chloroform treated in the
same manner. Calculate the content of C27H34O3 from the
Nandrolone Phenylpropionate Injection is a sterile solution
absorbance obtained from a 0.006 per cent w/v solution of
of Nandrolone Phenylpropionate in Ethyl Oleate or other
nandrolone phenylpropionate RS treated in the same manner.
suitable ester, in a suitable fixed oil or in a mixture of these.
Storage. Store protected from light.
Nandrolone Phenylpropionate Injection contains not less than
92.5 per cent and not more than 107.5 per cent of the stated Labelling. The label states that the preparation is for
amount of nandrolone phenylpropionate, C27H34O3. intramuscular injection only.

809
NAPHAZOLINE NITRATE IP 2007

Naphazoline Nitrate Test solution. Dissolve 0.2 g of the substance under


examination in 10 ml of methanol.
N Reference solution. A solution containing 2 per cent w/v of
naphazoline nitrate RS and 0.01 per cent w/v of
N
naphthylacetylethylenediamine hydrochloride RS.
H
, HNO3 Apply to the plate 10 µl of each solution. After development,
dry the plate at 105° for 5 minutes, spray with a 0.5 per cent
w/v solution of ninhydrin in methanol and heat at 105° for
C4HI4N2,HNO3 Mol. Wt. 273.3 10 minutes. Any spot corresponding to naphthylacetyl-
ethylenediamine hydrochloride in the chromatogram obtained
Naphazoline Nitrate is 2-(1-napthylmethyl)-2-imidazoline
with the test solution is not more intense than the
nitrate.
corresponding spot in the chromatogram obtained with the
Naphazoline Nitrate contains not less than 99.0 per cent and reference solution. The test is not valid unless the
not more than 101.0 per cent of C4HI4N2,HNO3 calculated on chromatogram obtained with the reference solution shows
the dried basis. two clearly separated spots.
Description. A white or almost white. crystalline powder. Chlorides (2.3.12). 15.0 ml of 1.0 per cent w/v solution in carbon
dioxide-free water complies with the limit test for chlorides
Identification (375 ppm).
Test A may be omitted if tests B, C and D are carried out. Tests Sulphated ash (2.3.18). Not more than 0.1 per cent.
B and C may be omitted if tests A and D are carried out.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
A. Determine by infrared absorption spectrophotometry (2.4.6). on 1.0 g by drying in an oven at 105° for 3 hours.
Compare the spectrum with that obtained with naphazoline
Assay. Weigh accurately about 0.2 g, dissolve in 30 ml of
nitrate RS.
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
B. When examined in the range 230 nm to 360 nm (2.4.7), a acid, determining the end-point potentiometrically (2.4.25).
0.002 per cent w/v solution in 0.01 M hydrochloric acid shows Carry out a blank titration.
absorption maxima at about 270 nm, 280 nm, 287 nm and
1 ml of 0.1 M perchloric acid is equivalent to 0.02733 g of
291 nm; absorbances at these maxima are about 0.43, 0.50, 0.35
C4HI4N2,HNO3.
and 0.34 respectively.
Storage. Store protected from light.
C. Dissolve about 0.5 mg in 1 m1 of methanol, add 0.5 ml of a
freshly prepared 5 per cent w/v solution of sodium
nitroprusside and 0.5 ml of a 2 per cent w/v solution of sodium
hydroxide, allow to stand for 10 minutes and add 1 ml of a
8 per cent w/v solution of sodium bicarbonate; a violet colour Nelfinavir Mesylate
is produced.
D. Dissolve about 10 mg in 5 ml of water, add 0.2 g of magnesium
oxide, shake mechanically for 30 minutes. add 10 ml of
chloroform and shake vigorously. Allow to stand, separate H
CH3
S O N
the chloroform layer, filter and evaporate the aqueous layer to CH3 O CH3
dryness. The residue gives reaction A for nitrates (2.3.1). HO CH3
N N , CH3SO3H
H H
Tests OH
H
Appearance of solution. A 1.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1) and colourless (2.4.1).
pH (2.4.24). 5.0 to 6.5, determined in a 1.0 per cent w/v solution. C32H45N3O4S,CH4O3S Mol. Wt. 663.9
Naphthylacetylethylenediamine. Determine by thin-layer Nelfinavir Mesylate is (3S,4aS,8aS)-N-(tert-butyldecahydro-
chromatography (2.4.17), coating the plate with silica gel G. 2-[(2R,3R)-3-(3-hydroxy-o-toluamido)-hydroxy-4-
Mobile phase. A mixture of 100 volumes of methanol and (phenylthio)butyl]isoquinoline-3-carboxamide methyl
1.5 volumes of strong ammonia solution. sulphonate.

810
IP 2007 NELFINAVIR MESTYLATE ORAL POWDER

Nelfinavir Mesylate contains not less than 98.0 per cent and 1 ml of 0.1 M sodium hydroxide is equivalent to 0.00961 g of
not more than 101.0 per cent of C32H45N3O 4S,CH4O3S, CH3SO3H.
calculated on the anhydrous basis.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Description. A white or almost white powder. heavy metals, Method B (20 ppm).
Identification Sulphated ash (2.3.18). Not more than 0.1 per cent.

A. Determine by infrared absorption spectrophotometry (2.4.6). Water (2.3.43). Not more than 3.0 per cent, determined on
Compare the spectrum with that obtained with nelfinavir 0.5 g.
mesylate RS or with the reference spectrum of nelfinavir Assay. Determine by liquid chromatography (2.4.14).
mesylate.
Test solution. A 0.01 per cent w/v solution of the substance
B. In the Assay, the principal peak in the chromatogram under examination in the mobile phase.
obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. Reference solution. A 0.01 per cent w/v solution of nelfinavir
mesylate RS in the mobile phase.
Tests
Chromatographic system
Specific optical rotation (2.4.22). –105° to –120°, determined – a stainless steel column 25 cm x 4.6 mm, packed with
in a 1.0 per cent w/v solution in methanol. octadecylsilane bonded to porous silica (5 µm),
Related substances. Determine by liquid chromatography – mobile phase: a filtered and degassed mixture of
(2.4.14), using the chromatographic system described in the 45 volumes of acetonitrile, 20 volumes of methanol
Assay. and 35 volumes of a buffer prepared by dissolving 4.0 g
of sodium dihydrogen phosphate in 1000 ml of water,
Test solution. A 0.1 per cent w/v solution of the substance
to which 1 ml of dimethylamine solution and 1 g of
under examination in the mobile phase.
sodium octanesulphonate are added and mixed to
Reference solution (a). A 0.001 per cent w/v solution of the dissolve,
substance under examination in the mobile phase. – flow rate. 1 ml per minute,
Reference solution (b). A 0.01 per cent w/v solution of – spectrophotometer set at 215 nm,
methanesulphonic acid in the mobile phase. – a 20 µl loop injector.
Inject reference solution (a). The test is not valid unless the Inject the reference solution. The test is not valid unless the
column efficiency determined from the nelfinavir peak is not column efficiency determined from the nelfinavir peak is not
less than 4000 theoretical plates and the tailing factor is not less than 5000 theoretical plates, the tailing factor is not more
more than 2.0. than 2.0 and the relative standard deviation for replicate
Separately inject reference solution (b) and record the injections is not more than 2.0 per cent.
chromatograms. Separately inject the test solution and Separately inject the test solution and the reference solution
continue the chromatography for at least three times the and measure the responses for the principal peak. Calculate
retention time of the principal peak. In the chromatogram the content of C32H45N3O4S,CH4O3S.
obtained with the test solution, the area of any peak other
than the principal peak is not greater than half of the area of Storage. Store protected from light.
the principal peak in the chromatogram obtained with reference
solution (a) (0.5 per cent) and the sum of the areas of all such
peaks is not greater than the area of the principal peak in the
chromatogram obtained with reference solution (a) (1.0 per Nelfinavir Mesylate Oral Powder
cent). Ignore any peak due to methanesulphonic acid
corresponding to the retention time of the principal peak in Nelfinavir Mesylate Oral Powder contains not less than
the chromatogram obtained with reference solution (b). 90.0 per cent and not more than 110.0 per cent of the stated
amount of nelfinavir, C32H45N3O4S.
Methanesulphonic acid. 13.5 per cent to 15.5 per cent w/w,
calculated on the anhydrous basis, determined by the following Identification
method. Weigh accurately about 0.6 g, dissolve in 50 ml of
dimethylformamide and titrate with 0.1 M sodium hydroxide, In the Assay, the principal peak in the chromatogram obtained
determining the end-point potentiometrically (2.4.25). Carry with the test solution corresponds to the peak in the
out a blank titration. chromatogram obtained with the reference solution.

811
NELFINAVIR TABLETS IP 2007

Tests (1.0 per cent) and the sum of areas of all the secondary peaks
is not more than twice the area of the peak in the chromatogram
Dissolution (2.5.2). obtained with the reference solution (b) (2.0 per cent).
Apparatus. No 1
Water (2.3.43). Not more than 12.0 per cent, determined on 0.5 g.
Medium. 900 ml of 0.1 M hydrochloric acid.
Speed and time. 75 rpm and 45 minutes. Assay. Determine by liquid chromatography (2.4.14).
Solvent mixture. 30 volumes of water and 70 volumes of
Withdraw a suitable volume of the medium and filter.
methanol.
Determine by liquid chromatography (2.4.14).
Test solution. Weigh accurately a quantity of the powder
Test solution. Use the filtrate and, if necessary, dilute with the containing 50 mg of Nelfinavir Mestlate, disperse in 50 ml of
dissolution medium. 0.1 M hydrochloric acid, dilute to 250.0 ml with the solvent
Reference solution. A 0.065 per cent w/v solution of nelfinavir mixture and filter.
mesylate RS in methanol. Dilute 10 ml of the solution to 100 ml Reference solution. Dissolve 10 mg of nelfinavir mesylate RS
with the dissolution medium. in 10 ml of 0.1 M hydrochloric acid and dilute to 50.0 ml with
Use the chromatographic system described under Assay. the solvent mixture.
Inject the test solution and the reference solution. Chromatographic system
– a stainless steel column 15 cm x 4.6 mm, packed with
D. Not less than 75 per cent of the stated amount of octadecylsilane bonded to porous silica (5 µm),
C32H45N3O4S. – column temperature 40º,
Related substances. Determine by liquid chromatography – mobile phase: a mixture of 35 volumes of a buffer solution
(2.4.14). prepared by dissolving 4 g of sodium dihydrogen
Test solution. Weigh accurately a quantity of the oral powder phosphate dihydrate and 1g of 1-octane sulphonic
containing 50 mg of Nelfinavir Mesylate, disperse in 10 ml of acid sodium salt into 1000 ml of water, adding 1ml of
methanol, dilute to 50 ml with the mobile phase and filter. dimethylamine and filtering, 45 volumes acetonitrile
and 20 volumes of methanol,
Reference solution (a). Dissolve 10 mg of nelfinavir mesylate – flow rate. 2 ml per minute,
RS in 2 ml of methanol and dilute to 10 ml with the mobile – spectrophotometer set at 220 nm,
phase. – a 10 µl loop injector.
Reference solution (b). Dilute 1 ml of reference solution (a) to Inject the reference solution. The test is not valid unless the
100 ml with the mobile phase. tailing factor is not more than 2.0, the column efficiency in not
Chromatographic system less than 2000 theoretical plates and the relative standard
– a stainless steel column 15 cm x 4.6 mm, packed with deviation for replicate injections is not more than 2.0 per cent.
octadecylsilane bonded to porous silica (5 µm), Inject the test solution and the reference solution.
– column temperature 45º,
– mobile phase: a mixture of 28 volumes of a buffer solution Calculate the content of C32H45N3O4S in the oral powder.
prepared by dissolving 4.88 g of anhydrous sodium Storage. Store protected from moisture, at a temperature not
dihydrogen phosphate in 1000 ml of water, adjusting exceeding 30º.
the pH to 3.4 with phosphoric acid and filtering, Labelling. The label states the strength in terms of the
27 volumes of acetonitrile, 20 volumes of methanol equivalent amount of nelfinavir.
and 25 volumes of water. Adjust the pH to 4.8 with 0.1
M sodium hydroxide or orthophosphoric acid.
– flow rate. 1 ml per minute,
– spectrophotometer set at 220 nm,
Nelfinavir Tablets
– a 10 µl loop injector. Nelfinavir Mesylate Tablets
Inject the reference solution (a). The test is not valid unless Nelfinavir Tablets contain not less than 90.0 per cent and not
the tailing factor is not more than 2.0 and the column efficiency more than 110.0 per cent of the stated amount of nelfinavir
in not less than 4000 theoretical plates. mesylate, C32H45N3O4S,CH4O3S.
Inject the test solution and reference solution (b). In the
Identification
chromatogram obtained with the test solution, the area of any
secondary peak is not more than the area of the peak in the A. Shake a quantity of the powdered tablets containing about
chromatogram obtained with the reference solution (b) 0.1 g of Nelfinavir Mesylate with 80 ml of methanol for

812
IP 2007 NEOMYCIN SULPHATE

10 minutes, add sufficient methanol to produce 100 ml, mix Inject separately the diluent (10 ml of methanol diluted to
and filter. Dilute 5 ml of the filtrate to 100 ml with methanol. 50 ml with the mobile phase) and the test solution and continue
When examined in the range 200 nm to 300 nm the resulting the chromatography for 4 times the retention time of the
solution shows an absorption maximum only at about 254 nm principal peak. Examine the diluent chromatogram for any
(2.4.7). extraneous peaks and ignore the corresponding peaks
observed in the chromatogram obtained with the test solution.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the Any secondary peak observed in the chromatogram obtained
chromatogram obtained with the reference solution. with the test solution should not be more than 1.0 per cent
and the sum of the areas of all the secondary peaks should
Tests not be more than 2.0 per cent when calculated by percentage
Dissolution (2.5.2). area normalisation. Inhibit integration of peak due to
Apparatus. No 1 methanesulphonic acid.
Medium. 900 ml of 0.01 M hydrochloric acid. Other tests. Complies with the tests stated under Tablets.
Speed and time. 50 rpm and 30 minutes. Assay. Determine by liquid chromatography (2.4.14).
Withdraw a suitable volume of the medium and filter promptly Test solution. Weigh accurately a quantity of the powdered
through a membrane filter disc with an average pore diameter tablets containing about 200 mg of Nelfinavir Mesylate, add
not greater than 1.0 µm. Reject the first few ml of the filtrate about 20 ml of methanol, mix with the aid of ultrasound for
and dilute a suitable volume of the filtrate with the same 10 minutes and dilute to 100.0 ml with the mobile phase. Filter
solvent. Measure the absorbance of the resulting solution at through a membrane filter disc with an average pore diameter
the maximum at about 250 nm (2.4.7). Calculate the content of not greater than 1.0 µm, rejecting the first few ml of the filtrate.
C32H45N3O4S,CH4O3S from the absorbance of a solution of Further dilute 5.0 ml of the filtrate to 100.0 ml with the mobile
known concentration of nelfinavir mesylate RS. phase.
D. Not less than 75 per cent of the stated amount of Reference solution. Weigh accurately about 50 mg of
C32H45N3O4S, CH4O3S. nelfinavir mesylate RS, add about 10 ml of methanol, mix with
Related substances. Determine by liquid chromatography the aid of ultrasound to dissolve and dilute to 50.0 ml with the
(2.4.14). mobile phase. Dilute 5.0 ml of this solution to 50.0 ml with the
Test solution. Weigh accurately a quantity of the powdered mobile phase.
tablets containing about 100 mg of Nelfinavir Mesylate, add Use the chromatographic system described in the test for
about 20 ml of methanol, mix with the aid of ultrasound for Related substances.
10 minutes and dilute to 100 ml with the mobile phase. Inject the reference solution. The test is not valid unless the
Reference solution. Weigh accurately about 10 mg of column efficiency determined from the nelfinavir mesylate peak
nelfinavir mesylate RS, add about 10 ml of methanol, shake is not less than 5000 theoretical plates, the tailing factor is not
for 10 minutes and dilute to 50 ml with the mobile phase. more than 2.0 and the relative standard deviation for replicate
Chromatographic system injections is not more than 2.0 per cent.
– a stainless steel column 25 cm x 4.6 mm, packed with Inject separately the test solution and the reference solution
octadecylsilane bonded to porous silica particles or and measure the responses for the major peak. Calculate the
ceramic microparticles (5 µm), content of C32H45N3O4S,CH4O3S in the tablets.
– mobile phase: a filtered and degassed mixture of
Storage. Store protected from light.
45 volumes of acetonitrile, 20 volumes of methanol
and 35 volumes of a buffer prepared by dissolving 4.0 g
of sodium dihydrogen phosphate in 1000 ml of water,
to which are added 1 ml of dimethylamine solution and Neomycin Sulphate
1 g of sodium octanesulphonate and mixing to dissolve, Neomycin Sulphate is a mixture of the sulphates of substances
– flow rate. 1 ml per minute, obtained by the growth of certain selected strains of
– spectrophotometer set at 215 nm, Streptomyces fradiae.
– a 20 µl loop injector.
Neomycin Sulphate has a potency of not less than 600 Units
Inject the reference solution. The test is not valid unless the
per mg, calculated on the dried basis.
column efficiency determined from the nelfinavir mesylate peak
is not less than 4000 theoretical plates and the tailing factor is Description. A white or yellowish-white powder; odourless
not more than 2.0. or almost odourless; hygroscopic.

813
NEOMYCIN EYE DROPS IP 2007

Identification Mobile phase. A mixture of 80 volumes of a 20 per cent w/v


solution of sodium chloride and 20 volumes of methanol.
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel H. Test solution. Dissolve 40 mg of the substance under
examination in water and dilute to 5 ml with the same solvent.
Mobile phase. A freshly prepared 3.85 per cent w/v solution
of ammonium acetate. Reference solution (a). Dissolve 30 mg of framycetin sulphate
RS in water and dilute to 25 ml with the same solvent.
Test solution. Dissolve 0.2 g of the substance under
Reference solution (b). Dilute 5 ml of reference solution (a) to
examination in 10 ml of water.
25 ml with water.
Reference solution. A 2.0 per cent w/v solution of neomycin
Reference solution (c). Dissolve 40 mg of neomycin sulphate
sulphate RS in water.
RS in water and dilute to 5 ml with the same solvent.
Apply to the plate 1 µl of each solution. After development,
Apply to the plate as 5-mm bands 5 µl of each solution. Dry
dry the plate in air for 10 minutes, heat at 100° for 1 hour and
the bands; allow the mobile phase to rise at least 12 cm. Dry
spray with a 0.1 per cent w/v solution of ninhydrin in
the plate at 100° to 105° for 10 minutes. Spray the plate with
1-butanol saturated with water. Heat again at 100° for
ethanolic ninhydrin solution and heat at 100° to 105° for
5 minutes. The principal spot in the chromatogram obtained
10 minutes. In the chromatogram obtained with the test
with the test solution corresponds to that in the chromatogram
solution the principal band corresponds to the principal band
obtained with the reference solution.
in the chromatogram obtained with reference solution (c) and
B. Dissolve about 10 mg in 5 ml of water, add 0.1 ml of pyridine the band due to neomycin C with an Rf value slightly less than
and 2 ml of a 0.1 per cent w/v solution of ninhydrin and heat that of the principal band is not more intense than the band
on a water-bath at a temperature of about 70° for 10 minutes; obtained with reference solution (a) (15 per cent) but is more
a deep violet colour is produced. intense than the band in the chromatogram obtained with
C. A 5 per cent w/v solution gives the reactions of sulphates reference solution (b) (3 per cent). The test is not valid unless
(2.3.1). in the chromatogram obtained with reference solution (c) a
band appears with an Rf value slightly less than that of the
Tests principal band.
pH (2.4.24). 5.0 to 7.5, determined in a 1.0 per cent w/v solution. Sulphated ash (2.3.18). Not more than 1.0 per cent.
Specific optical rotation (2.4.22).+53.5° to +59.0°, determined Loss on drying (2.4.19). Not more than 8.0 per cent, determined
in a 10.0 per cent w/v solution. on 0.5 g by drying in an oven at 60° over phosphorus pentoxide
at a pressure not exceeding 0.7 kPa for 3 hours.
Neamine. Determine by thin-layer chromatography (2.4.17),
coating the plate with silica gel H. Assay. Determine by the microbiological assay of antibiotics,
Method A (2.2.10).
Mbile phase. A mixture of 30 volumes of methanol, 20 volumes
of strong ammonia solution and 10 volumes of Storage. Store protected from light and moisture.
dichloromethane. Labelling. The label states the strength in terms of Units of
Test solution. Dissolve 0.25 g of the substance under neomycin per mg.
examination in 10 ml of water.
Reference solution. A 0.05 per cent w/v solution of neamine
RS in water. Neomycin Eye Drops
Apply to the plate as 5-mm bands 5 µl of each solution. Dry Neomycin Sulphate Eye Drops
the bands; allow the mobile phase to rise at least 8 cm. Dry the
Neomycin Sulphate Eye Drops are a sterile solution of
plate in a current of warm air, heat at 110° for 10 minutes, spray
Neomycin Sulphate in Purified Water.
the plate with ninhydrin and stannous chloride reagent and
heat at 110° for 15 minutes. Spray the plate again with the Neomycin Sulphate Eye Drops contain not less than 90.0 per
same reagent and heat at 110° for 15 minutes. Any band cent and not more than 115.0 per cent w/v of the stated amount
corresponding to neamine in the chromatogram obtained with of neomycin sulphate.
the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution. Identification
Neomycin C. Determine by thin-layer chromatography (2.4.17), Determine by thin-layer chromatography (2.4.17), coating the
coating the plate with silica gel of a suitable grade. plate with silica gel.

814
IP 2007 NEOMYCIN EYE OINTMENT

Mobile phase. A mixture of 60 volumes of methanol, Chromatographic system


40 volumes of strong ammonia solution and 20 volumes of – a stainless steel column 20 cm x 4.6 mm, packed with
chloroform. porous silica particles (5 µm) (such as Nucleosil 100-5),
Test solution. Dilute if necessary a volume of the eye drops to – mobile phase: a mixture of 97 ml of tetrahydrofuran,
produce a solution containing 0.5 per cent w/v of Neomycin 1.0 ml of water and 0.5 ml of glacial acetic acid diluted
Sulphate in water. with sufficient of a 2.0 per cent v/v solution of ethanol
in ethanol-free chloroform to produce 250 ml,
Reference solution (a). A 0.5 per cent w/v solution of – flow rate. 1.6 ml per minute,
neomycin sulphate RS in water. – spectrophotometer set at 350 nm,
Reference solution (b). A mixture of equal volumes of the eye – a 10 µl loop injector.
drops and reference solution (a). If necessary the tetrahydrofuran and water content of the
Apply to the plate 1 µl of each solution. After development, mobile phase may be adjusted so that the chromatogram
dry the plate in air, spray with a 1 per cent w/v solution of obtained with the reference solution shows resolution similar
ninhydrin in 1-butanol and heat at 105° for 2 minutes. The to that in the specimen chromatogram supplied with framycetin
principal red spot in the chromatogram obtained with the test sulphate RS. The mobile phase should be passed through the
solution corresponds to that in the chromatogram obtained column for several hours before the solutions are injected.
with reference solution (a) and the principal red spot in the Continue the chromatography for 1.4 times the retention time
chromatogram obtained with reference solution (b) appears of the peak due to neomycin B.
as a single spot. The column efficiency, determined using the peak due to
Neomycin B in the chromatogram obtained with the test
Tests solution, should be not less than 13,000 theoretical plates.
Neamine. Determine by thin-layer chromatography (2.4.17), In the chromatogram obtained with the test solution the area
coating the plate with silica gel H. of the peak corresponding to neomycin C is not less than
Mobile phase. A mixture of 30 volumes of methanol, 3.0 per cent and not more than 15.0 per cent of sum of the areas
20 volumes of strong ammonia solution and 10 volumes of of the peaks corresponding to Neomycin B and Neomycin C.
dichloromethane. Other Tests. Complies with the tests stated under Eye Drops.
Test solution. A volume of the eye drops containing 5 µg Assay. Measure accurately a quantity containing 5 mg of
(3.5 Units). Neomycin Sulphate and dilute to 50.0 ml with sterile phosphate
Reference solution. The same volume of water containing buffer pH 8.0 and mix. Dilute 10.0 ml of the resulting solution
0.1 µg of neamine RS. to 100.0 ml with the same solvent.

Apply to the plate each solution. After development, dry the Determine by the microbiological assay of antibiotics, Method
plate in a current of warm air, heat at 110° for 10 minutes, spray A (2.2.10)
the plate with ninhydrin and stannous chloride reagent and The upper fiducial limit of error is not less than 90.0 per cent
heat at 110° for 15 minutes. Spray the plate again with the and the lower fiducial limit of error is not more than 115.0 per
same reagent and heat at 110° for 15 minutes. Any spot cent of the stated number of Units per ml.
corresponding to neamine in the chromatogram obtained with
Storage. Store protected from light.
the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution. Labelling. The strength is stated in terms of percentage w/v
as well as the number of Units per ml.
Neomycin C. Determine by liquid chromatography (2.4.14).
Test solution. Dilute the eye drops with 0.02 M borax to
contain 1 mg (700 Units) per ml. To 0.5 ml of the diluted solution
add 1.5 ml of a freshly prepared 2 per cent w/v solution of Neomycin Eye Ointment
1-fluoro-2,4-dinitrobenzene in methanol, dilute to 25 ml with
Neomycin Sulphate Eye Ointment
the mobile phase, allow to stand and use the clear lower layer.
Neomycin Sulphate Eye Ointment is a sterile preparation
Reference solution. Add 1.5 ml of the 1-fluoro-2,4-
containing Neomycin Sulphate in a suitable basis.
dinitrobenzene solution to 0.5 ml of a 0.1 per cent w/v solution
of neomycin sulphate RS in 0.02 M borax, heat in a water- Neomycin Sulphate Eye Ointment contains not less than
bath at 60° for 1 hour and cool; dilute the solution to 25 ml with 90.0 per cent and not more than 115.0 per cent of the stated
the mobile phase, allow to stand and use the clear lower layer. amount of neomycin sulphate.

815
NEOMYCIN EYE OINTMENT IP 2007

Identification cent w/v solution of 1-fluoro-2,4-dinitrobenzene in methanol,


heat on a water-bath at 60° for 1 hour and cool. Dilute the
Determine by thin-layer chromatography (2.4.17), coating the resulting solution to 25 ml with the mobile phase, allow to
plate with silica gel. stand and use the clear lower layer.
Mobile phase. A mixture of 60 volumes of methanol, 40 volumes Reference solution. Add 1.5 ml of the 1-fluoro-2,4-
of strong ammonia solution and 20 volumes of chloroform. dinitrobenzene solution to 0.5 ml of a 0.1 per cent w/v solution
Test solution. Disperse a quantity of the eye ointment of neomycin sulphate RS in 0.02 M borax and proceed as for
containing 20 mg of Neomycin Sulphate in 20 ml of chloroform, the test solution.
extract with 5 ml of water and use the aqueous extract. Chromatographic system
Reference solution (a). A 0.4 per cent w/v solution of – a stainless steel column 20 cm x 4.6 mm, packed with
neomycin sulphate RS in water. porous silica particles (5 µm),
– mobile phase: 97 ml of tetrahydrofuran, 1.0 ml of water
Reference solution (b). A mixture of equal volumes of test and 0.5 ml of glacial acetic acid with sufficient of a
solution and reference solution (a). 2.0 per cent v/v solution of ethanol in ethanol-free
Apply to the plate 1 µl of each solution. After development, chloroform to produce 250 ml,
dry the plate in air, spray with a 1 per cent w/v solution of – flow rate. 1.6 ml per minute,
ninhydrin in 1-butanol and heat at 105° for 2 minutes. The – spectrophotometer set at 350 nm,
principal red spot in the chromatogram obtained with the test – a 10 µl loop injector.
solution corresponds to that in the chromatogram obtained If necessary the tetrahydrofuran and water content of the
with reference solution (a) and the principal red spot in the mobile phase may be adjusted so that the chromatogram
chromatogram obtained with reference solution (b) appears obtained with reference solution shows resolution similar to
as a single spot. that in the specimen chromatogram supplied with framycetin
sulphate RS. The mobile phase should be passed through the
Tests column for several hours before the solutions are injected.
Neamine. Determine by thin-layer chromatography (2.4.17), Continue the chromatography for 1.4 times the retention time
coating the plate with silica gel H. of the peak due to neomycin B.

Mobile phase. A mixture of 30 volumes of methanol, The column efficiency, determined using the peak due to
20 volumes of strong ammonia solution and 10 volumes of Neomycin B in the chromatogram obtained with the test
dichloromethane. solution, should be not less than 13,000 theoretical plates.
In the chromatogram obtained with the test solution the area
Test solution. Disperse a quantity of the eye ointment
of the peak corresponding to neomycin C is not less than
containing 20 mg of Neomycin Sulphate in 20 ml of chloroform,
3.0 per cent and not more than 15.0 per cent of the sum of the
shake gently with 8 ml of water, allow the layers to separate
areas of the peaks corresponding to Neomycin B and Neomycin
and use the aqueous layer.
C.
Reference solution. A 0.005 per cent w/v solution of neamine
Other Tests. Complies with the tests stated under Eye
RS in water.
Ointments.
Apply to the plate 2 µl of each solution. After development,
Assay. Weigh accurately a quantity containing 5 mg of
dry the plate in a current of warm air, heat at 110° for 10 minutes,
Neomycin Sulphate, dissolve in 25 ml of chloroform, extract
spray with ninhydrin and stannous chloride reagent and
with four quantities, each of 20 ml, of sterile phosphate buffer
heat at 110° for 15 minutes. Spray the plate again with the
pH 8.0, combine the extracts and add sufficient of the buffer
same reagent and heat at 110° for 15 minutes. Any spot
solution to produce 100.0 ml.
corresponding to neamine in the chromatogram obtained with
the test solution is not more intense than the spot in the Carry out the microbiological assay of antibiotics, Method A
chromatogram obtained with the reference solution. (2.2.10).
Neomycin C. Determine by liquid chromatography (2.4.17) The upper fiducial limit of error is not less than 90.0 per cent
and the lower fiducial limit of error is not more than 115.0 per
Test solution. Disperse a quantity of the eye ointment cent of the stated number of Units per g.
containing 5 mg of Neomycin Sulphate in 20 ml of light
petroleum (120° to 160°), add 5 ml of 0.02 M borax, shake, Storage. Store protected from light.
separate the aqueous layer and centrifuge. To 0.5 ml of the Labelling. The strength is stated in terms of percentage w/v
separated aqueous layer add 1.5 ml of a freshly prepared 2 per as well as the number of Units per ml.

816
IP 2007 NEOSTIGMINE TABLETS

Neostigmine Bromide measured immediately after preparation, not more than


0.25 (2.4.7).

CH3 Sulphates (2.3.17). 0.75 g complies with the limit test for
sulphates (200 ppm).
H3C N CH3
Sulphated ash (2.3.18). Not more than 0.1 per cent.
O Br Loss on drying (2.4.19). Not more than 1.0 per cent, determined
CH3 on 1.0 g by drying in an oven at 105°.
O N
CH3 Assay. Weigh accurately about 0.5 g, dissolve in 20 ml of
anhydrous glacial acetic acid, add 5 ml of acetic anhydride.
Titrate with 0.1 M perchloric acid, using crystal violet
C12H19BrN2O2 Mol. Wt. 303.2
solution as indicator. Carry out a blank titration.
Neostigmine Bromide is 3-(dimethylcarbamoyloxy)
1 ml of 0.1 M perchloric acid is equivalent to 0.03032 g of
trimethylanilinium bromide.
C12H19BrN2O2.
Neostigmine Bromide contains not less than 98.0 per cent and
Storage. Store protected from light and moisture.
not more than 101.0 per cent of C12H19BrN2O2, calculated on
the dried basis.
Description. Colourless crystals or a white, crystalline powder;
odourless; hygroscopic.
Neostigmine Tablets
Neostigmine Bromide Tablets
Identification
Neostigmine Bromide Tablets contain not less than 92.5 per
Test A may be omitted if tests B, C, D and E are carried out. cent and not more than 107.5 per cent of the stated amount of
Tests B, C and D may be omitted if tests A and E are carried neostigmine bromide, C12H19BrN2O2.
out.
A. Determine by infrared absorption spectrophotometry (2.4.6). Identification
Compare the spectrum with that obtained with neostigmine Triturate a quantity of the powdered tablets containing about
bromide RS. 0.3 g of Neostigmine Bromide with three quantities, each of
B. When examined in the range 230 nm to 360 nm (2.4.7), a 5 ml of ether and discard the ether. Macerate the residue with
0.02 per cent w/v solution in 0.5 M sulphuric acid shows several quantities, each of 10 ml of ethanol (95 per cent),
absorption maxima at about 260 nm and 266 nm. filtering after each maceration. Evaporate the combined filtrates
on a water-bath and dry the residue at 105° for 1 hour. The
C. Warm about 50 mg with 1 ml of dilute sodium hydroxide
residue melts at about 167°, with decomposition. The residue
solution; an odour of dimethylamine develops slowly.
complies with the following tests.
D. Warm about 50 mg with 0.4 g of potassium hydroxide and
A. Warm about 50 mg with 0.4 g of potassium hydroxide and
2 ml of ethanol (95 per cent) on a water-bath for 3 minutes,
2 ml of ethanol (95 per cent) on a water-bath for 3 minutes,
replacing the evaporated ethanol. Cool, add 2 ml of dilute
replacing the evaporated ethanol. Cool, add 2 ml of dilute
diazobenzenesulphonic acid solution; an orange-red colour
diazobenzenesulphonic acid solution; an orange-red colour
is produced.
is produced.
E. Gives the reactions of bromides (2.3.1). B. Gives the reactions of bromides (2.3.1).
Tests Tests
Appearance of solution. A 0.5 per cent w/v solution is clear
Other Tests. Complies with the tests stated under Tablets.
(2.4.1), and colourless (2.4.1).
Assay. Weigh and powder 20 tablets. Weigh accurately a
Acidity. Dissolve 0.2 g in 20 ml of carbon dioxide-free water
quantity of the powder containing about 0.15 g of Neostigmine
and titrate to pH 7.0 with 0.02 M sodium hydroxide (carbonate-
Bromide, transfer to a semi-micro ammonia-distillation
free); not more than 0.1 ml is required.
apparatus, add 20 ml of a 50 per cent w/v solution of sodium
3-Hydroxytrimethylanilinium bromide. Dissolve 50 mg in a hydroxide and 0.5 ml of a 2 per cent w/v solution of 2-octanol
mixture of 1 ml of sodium carbonate solution and 9 ml of in liquid paraffin. Pass a current of steam through the mixture,
water. Absorbance of the resulting solution at about 294 nm, collect the distillate in 50 ml of 0.01 M sulphuric acid until the

817
NEOSTIGMINE METHYLSULPHATE IP 2007

volume is about 200 ml and titrate the excess of acid with phthalein solution; the solution is colourless. Add
0.02 M sodium hydroxide using methyl red solution as 0.3 ml of 0.01 M sodium hydroxide; the solution becomes red.
indicator. Repeat the operation without the substance under Add 0.4 ml of 0.01 M hydrochloric acid; the solution becomes
examination. The difference between the titrations represents colourless. Add 0.1 ml of methyl red solution; the solution
the amount of sulphuric acid required to neutralise the becomes red or yellowish-red.
dimethylamine produced. 3-Hydroxytrimethylanilinium methyl sulphate. Dissolve
1 ml of 0.01 M sulphuric acid is equivalent to 0.006064 g of 50 mg in a mixture of 1 ml of sodium carbonate solution and
C12H19BrN2O2. 9 ml of water. Absorbance of the resulting solution at about
Storage. Store protected from light and moisture. 294 nm, measured immediately after preparation, not more than
0.20 (2.4.7).
Chlorides (2.3.12). 1.0 g complies with the limit test for
Neostigmine Methylsulphate chlorides (250 ppm).
C13H22N2O6S Mol. Wt. 334.4 Sulphates (2.3.17). 0.75 g complies with the limit test for
sulphates (200 ppm).
Neostigmine Methylsulphate is 3-(dimethylcarbamoyloxy)-
trimethylanilinium methyl sulphate. Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105°.
Neostigmine Methylsulphate contains not less than 98.5 per
cent and not more than 101.0 per cent of C13H22N2O6S, Sulphated ash (2.3.18). Not more than 0.1 per cent.
calculated on the dried basis. Assay. Weigh accurately about 0.3 g and dissolve in 150 ml of
Description. Colourless crystals or a white, crystalline powder; water. Add 100 ml of 2 M sodium hydroxide, distill and collect
hygroscopic. the distillate in 50 ml of a 4 per cent w/v solution of boric acid
until a total volume of 250 ml is reached. Titrate the distillate
Identification with 0.1 M hydrochloric acid using 0.25 ml of methyl red-
methylene blue solution as indicator. Repeat the operation
Test A may be omitted if tests B, C and D are carried out. Tests
without the substance under examination. The difference
B, C and D may be omitted if test A is carried out.
between the titrations represents the amount of hydrochloric
A. Determine by infrared absorption spectrophotometry (2.4.6). acid required.
Compare the spectrum with that obtained with neostigmine
1 ml of 0.1 M hydrochloric acid is equivalent to 0.03344 g of
methylsulphate RS or with the reference spectrum of
C13H22N2O6S.
neostigmine methylsulphate.
Storage. Store protected from light and moisture.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.05 per cent w/v solution in 0.5 M sulphuric acid shows
absorption maxima, at about 261 nm and 267 nm. The ratio of
the absorbance at the maximum at about 267 nm to that at the Neostigmine Injection
maximum at 261 nm is 0.84 to 0.87.
Neostigmine Methylsulphate Injection
C. Dissolve 0.1 g in 5 ml of distilled water and add 1 ml of a
6 per cent w/v solution of barium chloride; no precipitate is Neostigmine Injection is a sterile solution of Neostigmine
produced. Add 2 ml of hydrochloric acid and heat in a water- Methylsulphate in Water for Injections.
bath for 10 minutes; a white precipitate is produced. Neostigmine Injection contains not less than 90.0 per cent
D. Warm about 50 mg with 0.4 g of potassium hydroxide and and not more than 110.0 per cent of the stated amount of
2 ml of ethanol (95 per cent) on a water-bath for 3 minutes, neostigmine methylsulphate, C13H22N2O6S.
replacing the evaporated ethanol. Cool, add 2 ml of dilute
Identification
diazobenzenesulphonic acid solution; an orange-red colour
is produced. A. Dilute, if necessary, a volume of the injection containing
2.5 mg of Neostigmine Methylsulphate to 5 ml with water,
Tests shake with three quantities, each of 10 ml, of ether and discard
Appearance of solution. A 5.0 per cent w/v solution in distilled the ether extracts.
water is clear (2.4.1), and colourless (2.4.1). When examined in the range 230 nm to 360 nm (2.4.7), a 2 cm
Acidity or alkalinity. To 4.0 ml of a 5.0 per cent w/v solution in layer of the resulting solution shows absorption maxima at
distilled water add 6.0 ml of water and 0.1 ml of phenol- about 260 nm and 267 nm.

818
IP 2007 NEVIRAPINE

B. Determine by thin-layer chromatography (2.4.17), coating – spectrophotometer set at 215 nm,


the plate with silica gel G. – a 10 µl loop injector.
Mobile phase. A mixture of 50 volumes of chloroform, In the chromatogram obtained with reference solution (b) the
35 volumes of methanol, 10 volumes of formic acid and principal peak has a retention time of about 6.8 minutes
5 volumes of water. (neostigmine methylsulphate) and there is a peak with a
relative retention time of about 0.5 ((3-hydroxy)
Test solution. Dilute the injection under examination, if
trimethylanilinium methylsulphate). In the chromatogram
necessary, with water to produce a solution containing
obtained with the test solution, the area of any secondary
0.05 per cent w/v of Neostigmine Methylsulphate.
peak with a retention time corresponding to that of the peak
Reference solution (a). A 0.05 per cent w/v solution of due to (3-hydroxy)trimethylanilinium methylsulphate in the
neostigmine methylsulphate RS in water. chromatogram obtained with reference solution (b) is not
Reference solution (b). A mixture of equal volumes of the test greater than the area of the principal peak in the chromatogram
solution and reference solution (a). obtained with reference solution (a) (1 per cent).

Apply to the plate 10 µl of each solution. After development, Other Tests. Complies with the tests stated under Parenteral
dry the plate in air, spray with dilute potassium Preparations (Injections).
iodobismuthate solution. The principal spot in the Assay. Dilute an accurately measured volume containing about
chromatogram obtained with the test solution corresponds to 25 mg of Neostigmine Methylsulphate to 50.0 ml with water.
that in the chromatogram obtained with reference solution (a). Measure the absorbance of the resulting solution at the
The principal spot in the chromatogram obtained with maximum at about 260 nm (2.4.7). Calculate the content of
reference solution (b) appears as a single, compact spot. C13H22N2O6S taking 14.35 as the specific absorbance at
C. To 1 ml add 0.5 ml of sodium hydroxide solution and 260 nm.
evaporate to dryness on a water-bath. Heat quickly in an oil- Storage. Store protected from light.
bath to about 250° and maintain at this temperature for about
30 seconds. Cool, dissolve the residue in 1 ml of water, cool in
ice water and add 1 ml of diazobenzenesulphonic acid
solution; an orange-red colour is produced. Nevirapine
Tests
O
pH. (2.4.24) 4.5 to 6.5. H 3C
HN
3-Hydroxy trimethylanilinium methyl sulphate. Determine
by liquid chromatography (2.4.14).
N N N
Test solution. Dilute the injection if necessary, with water to
contain a 0.05 per cent w/v solution of Neostigmine
Methylsulphate.
Reference solution (a). Dilute 1 volume of the test solution to C15H14N4O Mol. Wt. 266.3
100 volumes with water. Nevirapine is 11-cyclopropyl-5,11-dihydro-4-methy-6H-
Reference solution (b). Add 0.05 ml of 5 M sodium hydroxide dipyrido[3,2-b:2′,3′-e][1,4]diazepin-6-one.
to 1 ml of the test solution and allow to stand for 5 minutes. Nevirapine contains not less than 98.0 per cent and not more
Add 0.1 ml of 5 M hydrochloric acid and use immediately. than 102.0 per cent of C15H14N4O, calculated on the dried basis.
Chromatographic system Description. A white or almost white crystalline powder.
– a stainless steel column 25 cm × 4.6 mm packed with
octadecylsilane chemically bonded to porous silica Identification
particles (5 µm) (such as Lichrosphere 60 RP-select B),
A. Determine by infrared absorption spectrophotometry (2.4.6).
– mobile phase: 0.0015 M solution of sodium
Compare the spectrum with that obtained with nevirapine RS
heptanesulphonate in a mixture of 15 volumes of
or with the reference spectrum of nevirapine.
acetonitrile and 85 volumes of 0.05 M potassium
dihydrogen orthophosphate adjusted to pH 3.0 with B. In the Assay, the principal peak in the chromatogram
orthophosphoric acid, obtained with the test solution corresponds to that in the
– flow rate. of 1.1 ml per minute, chromatogram obtained with the reference solution.

819
NEVIRAPINE TABLETS IP 2007

Tests and 60 volumes of a buffer prepared by dissolving


12.0 g of sodium dihydrogen phosphate in about 800 ml
Related substances. Determine by liquid chromatography of water, adjusting the pH to 3.0 with orthophosphoric
(2.4.14). acid and diluting to 1000.0 ml with water,
Test solution. Dissolve 0.1 g of the substance under – flow rate. 1.2 ml per minute,
examination in 100 ml of methanol. – spectrophotometer set at 230 nm,
– a 20 µl loop injector.
Reference solution. Dilute 1 ml of the test solution to 100 ml
with methanol. Inject the reference solution. The test is not valid unless the
Chromatographic system column efficiency determined from the nevirapine peak is not
– a stainless steel column 25 cm x 4.6 mm, packed with less than 3000 theoretical plates, the tailing factor is not more
octadecylsilane bonded to porous silica or ceramic than 2.0 and the relative standard deviation for the replicate
microparticles (5 µm), injections is not more than 2.0 per cent.
– mobile phase: a filtered and degassed mixture of Separately inject the test solution and the reference solution
20 volumes of methanol, 20 volumes of acetonitrile and measure the responses for the principal peak. Calculate
and 60 volumes of a buffer prepared by dissolving the content of C15H14N4O.
12.0 g of sodium dihydrogen phosphate in about 800 ml
Storage. Store protected from moisture.
of water, adjusting the pH to 3.0 with phosphoric acid
and diluting to 1000.0 ml with water,
– flow rate. 1.2 ml per minute,
– spectrophotometer set at 230 nm, Nevirapine Tablets
– a 20 µl loop injector.
Nevirapine Tablets contain not less than 90.0 per cent and not
Inject the reference solution. The test is not valid unless the more than 110.0 per cent of the stated amount of nevirapine,
column efficiency determined from the nevirapine peak is not C15H14N4O.
less than 5000 theoretical plates and the tailing factor is not
more than 2.0. Identification
Separately inject the test solution and the reference solution. A. When examined in the range 250 nm to 450 nm (2.4.7) a
In the chromatogram obtained with the test solution, the area 0.001 per cent w/v solution in the mobile phase described
of any peak other than the principal peak is not greater than under Assay, shows an absorption maximum at about 230 nm.
half of the area of the principal peak in the chromatogram
B. In the Assay, the principal peak in the chromatogram
obtained with the reference solution (0.5 per cent) and the
obtained with the test solution corresponds to the peak in the
sum of the areas of all such peaks is not greater than the area
chromatogram obtained with the reference solution.
of the principal peak in the chromatogram obtained with the
reference solution (1.0 per cent). Dissolution (2.5.2).
Heavy metals (2.3.13). 1.0 g complies with the limit test for Apparatus. No 1.
heavy metals, Method B (20 ppm). Medium. 900 ml of 0.1 M hydrochloric acid.
Sulphated ash (2.3.18). Not more than 0.2 per cent. Speed and time. 50 rpm and 45 minutes.

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Withdraw a suitable volume of the medium and filter through
on 1.0 g by drying at 105° for 3 hours. a membrane filter disc with an average pore diameter not greater
than 1.0 µm, rejecting the first few ml of the filtrate. Measure
Assay: Determine by liquid chromatography (2.4.14). the absorbance of the resulting solution, at the maximum at
Test solution. A 0.005 per cent w/v solution of the substance about 313 nm (2.4.7).
under examination in methanol.
Calculate the content of C15H14N4O from the absorbance
Reference solution. A 0.005 per cent w/v solution of obtained from a solution of known concentration of nevirapine
nevirapine RS in methanol. RS in 0.1 M hydrochloric acid.
Chromatographic system D. Not less than 70 per cent of the stated amount of C15H14N4O.
– a stainless steel column 25 cm x 4.6 mm, packed with
Related substances. Determine by liquid chromatography
octadecylsilane bonded to porous silica or ceramic
(2.4.14).
microparticles (5 µm),
– mobile phase: a filtered and degassed mixture of Test solution. Shake a quantity of the powdered tablets with a
20 volumes of methanol, 20 volumes of acetonitrile suitable quantity of the mobile phase to obtain a mixture

820
IP 2007 NEVIRAPINE ORAL SUSPENSION

containing 0.05 per cent w/v of Nevirapine and filter through – flow rate. 1.2 ml per minute,
a membrane filter disc with an average diameter not exceeding – spectrophotometer set at 230 nm,
1.0 µm, rejecting the first few ml of the filtrate. – a 20 µl loop injector.
Reference solution. A 0.05 per cent w/v solution of nevirapine Inject the reference solution. The test is not valid unless the
RS in the mobile phase. column efficiency determined from the nevirapine peak is not
less than 7500 theoretical plates, the tailing factor is not more
Chromatographic system
than 1.5 and the relative standard deviation for replicate
– a stainless steel column 25 cm x 4.6 mm, packed with
injections is not more than 2.0 per cent.
octadecylsilane bonded to porous silica or ceramic
microparticles (5 µm), Separately inject the test solution and the reference solution
– mobile phase: a filtered and degassed mixture of and measure the responses for the principal peak. Calculate
20 volumes of methanol, 20 volumes of acetonitrile the content of C15H14N4O in the tablets.
and 60 volumes of a buffer prepared by dissolving
Storage. Store protected from moisture at a temperature not
12.0 g of sodium dihydrogen phosphate in about 800 ml
exceeding 30°.
of water, adjusting the pH to 3.0 with orthophosphoric
acid and diluting to 1000.0 ml with water,
– flow rate. 1.2 ml per minute,
– spectrophotometer set at 230 nm, Nevirapine Oral Suspension
– a 20 µl loop injector.
Nevirapine Oral Suspension is a suspension of Nevirapine in
Inject the reference solution. The test is not valid unless the a suitable flavoured vehicle.
column efficiency determined from the nevirapine peak is not
less than 7500 theoretical plates and the tailing factor is not Nevirapine Oral Suspension contains not less than 90.0 per
more than 1.5 and the relative standard deviation for replicate cent and not more than 110.0 per cent of the stated amount of
injections is not more than 2 per cent. nevirapine, C15H14N4O.

Inject the test solution and continue the chromatography for Identification
at least five times the retention time of the principal peak.
A. Determine by thin-layer chromatography (2.4.17), coating
Determine the amount of related substances by the area
the plate with silica gel GF254.
normalisation method. Any individual impurity is not more
than 1.0 per cent and the sum of all impurities is not more than Mobile phase. A mixture of 40 volumes of 1-butanol,
2.0 per cent. 30 volumes of heptane, 30 volumes of acetone and 10 volumes
of strong ammonia solution.
Other tests. Comply with the tests stated under Tablets.
Test solution. Dilute the preparation under examination with
Assay. Determine by liquid chromatography (2.4.14).
methanol to obtain a solution containing 1 mg of Nevirapine
Test solution. Weigh and powder 20 tablets. Shake a quantity per ml.
of powder containing about 100 mg of Nevirapine with
Reference solution. A 0.1 per cent w/v solution of nevirapine
sufficient of the mobile phase to obtain a mixture containing
RS in a mixture of 75 volumes of methanol and 25 volumes of
0.05 per cent w/v of Nevirapine. Mix and filter through a
water.
membrane filter disc with an average pore diameter not greater
than 1.0 µm, rejecting the first few ml of the filtrate. Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Reference solution. A 0.05 per cent w/v solution of nevirapine The principal spot in the chromatogram obtained with the test
RS in the mobile phase. solution corresponds to that in the chromatogram obtained
Chromatographic system with the reference solution.
– a stainless steel column 25 cm x 4.6 mm, packed with
B. In the Assay, the principal peak in the chromatogram
octadecylsilane bonded to porous silica or ceramic
obtained with the test solution corresponds to the peak in the
microparticles (5 µm),
chromatogram obtained with the reference solution.
– mobile phase: a filtered and degassed mixture of
20 volumes of methanol, 20 volumes of acetonitrile Tests
and 60 volumes of a buffer prepared by dissolving
12.0 g of sodium dihydrogen phosphate in about 800 ml pH (2.4.24). 5.0 to 7.0.
of water, adjusting the pH to 3.0 with phosphoric acid Related substances. Determine by liquid chromatography
and diluting to 1000.0 ml with water, (2.4.14).

821
NICLOSAMIDE IP 2007

Test solution. To an accurately measured volume of the Reference solution. Weigh accurately about 50 mg of
preparation under examination containing about 25 mg of nevirapine RS, add about 20 ml of methanol, mix with the aid
Nevirapine add about 10 ml of methanol, mix with the aid of of ultrasound to dissolve and dilute to 100.0 ml with water.
ultrasound for 10 minutes, dilute to 50 ml with water, mix and Dilute 10.0 ml of this solution to 25.0 ml with water.
filter. Use the chromatographic system described under the test for
Reference solution. Weigh accurately about 25 mg of Related substances.
nevirapine RS, add about 10 ml of methanol, mix with the aid
Inject the reference solution. The test is not valid unless the
of ultrasound to dissolve and dilute to 50 ml with water.
relative standard deviation for replicate injections is not more
Chromatographic system than 2.0 per cent.
– a stainless steel column 25 cm x 4.6 mm, packed with
octylsilyl silica gel for chromatography (5 µm)(such as Inject separately the test solution and the reference solution
Hypersil C8), and measure the responses for the principal peak.
– mobile phase: filtered and degassed gradient mixtures Determine the weight per ml of the oral suspension (2.4.29)
of methanol and 0.1 M ammonium acetate, and calculate the content of C15H14N4O weight in volume.
– flow rate. 1 ml per minute,
Storage. Store protected from light.
– a linear gradient programme using the conditions given
below,
– spectrophotometer set at 270 nm,
– a 20 µl loop injector.
Niclosamide
Time 0.1 M ammonium acetate Methanol
(in min.) (per cent v/v) (per cent v/v) Anhydrous Niclosamide
0 95 5
5 95 5 Cl NO2
25 20 80 OH O
30 20 80 N
31 95 5 H
40 95 5
Inject the reference solution. The test is not valid unless the Cl
column efficiency determined from the nevirapine peak is not
less than 3000 theoretical plates and the tailing factor is not C13H8Cl2N2O4 Mol. Wt. 327.1
more than 2.0. Niclosamide is 2′,5-dichloro-4′-nitrosalicylanilide.
Inject separately the diluent (dilute 10 ml of methanol to 50 ml
Niclosamide contains not less than 98.0 per cent and not more
with water) and the test solution. Examine the diluent
than 101.0 per cent of C13H8Cl2N2O4, calculated on the dried
chromatogram for any extraneous peaks and ignore the
basis.
corresponding peaks observed in the chromatogram obtained
with the test solution. Ignore any peaks due to preservatives Description. A yellowish white to yellowish, fine crystals or
also. powder; odourless.
Any secondary peak observed in the chromatogram obtained
Identification
with the test solution should not be more than 1.0 per cent
and the sum of the areas of all the secondary peaks should Test A may be omitted if tests B and C are carried out. Tests B
not be more than 2.0 per cent when calculated by percentage and C may be omitted if test A is carried out.
area normalisation.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Other tests. Comply with the tests stated under Oral Liquids. Compare the spectrum with that obtained with niclosamide
Assay. Determine by liquid chromatography (2.4.14) RS or with the reference spectrum of niclosamide.
Test solution. Weigh accurately a quantity of the preparation B. Heat 50 mg with 5 ml of 1 M hydrochloric acid and 0.1 g of
under examination containing 25 mg of Nevirapine, add about zinc powder in a water-bath for 10 minutes, cool and filter. To
10 ml of methanol, mix with the aid of ultrasound for 10 minutes, the filtrate add 1 ml of a 0.5 per cent w/v solution of sodium
dilute to 50.0 ml with water, mix and filter. Further dilute 10.0 ml nitrite and allow to stand for 3 minutes. Add 2 ml of a 2 per
of the filtrate to 25.0 ml with water. cent w/v solution of ammonium sulphamate, shake, allow to

822
IP 2007 NICLOSAMIDE TABLETS

stand for 3 minutes and add 2 ml of a 0.5 per cent w/v solution Niclosamide Tablets
of N- (1-naphthyl) ethylenediamine dihydrochloride; a violet
colour is produced. Niclosamide Tablets contain not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of
C. Heat the substance under examination on a copper wire in niclosamide, C 13 H 8 Cl 2 N 2O 4 . The tablets may contain
a non-luminous flame; a green colour is imparted to the flame. sweetening and flavouring agents.
Tests Identification
Chlorides (2.3.12). To 2.0 g add a mixture of 40 ml of water and Heat a quantity of the powdered tablets containing 0.5 g of
1.2 ml of 5 M acetic acid, boil for 2 minutes, cool and filter; Niclosamide with 25 ml of hot ethanol (95 per cent), filter
10 ml of the filtrate diluted to 15 ml with water complies with while hot and evaporate the filtrate to dryness on a water-
the limit test for chlorides (500 ppm). bath. The residue complies with the following tests.
2-Chloro-4-nitroaniline. Not more than 100 ppm, determined A. Determine by infrared absorption spectrophotometry (2.4.6).
by the following method. Boil 0.25 g with 5 ml of methanol, Compare the spectrum with that obtained with niclosamide
cool, add 45 ml of 1 M hydrochloric acid, heat to boiling, RS or with the reference spectrum of niclosamide.
cool, filter and dilute the filtrate to 50.0 ml with 1 M
hydrochloric acid. To 10.0 ml of this solution add 0.5 ml of a B. Heat 50 mg with 5 ml of 1 M hydrochloric acid and 0.1 g of
0.5 per cent w/v solution of sodium nitrite and allow to stand zinc powder in a water-bath for 10 minutes, cool and filter. To
for 3 minutes. Add 1.0 ml of a 2 per cent w/v solution of the filtrate add 1 ml of a 0.5 per cent w/v solution of sodium
ammonium sulphamate, shake, allow to stand for 3 minutes nitrite and allow to stand for 3 minutes. Add 2 ml of a 2 per
and add 1.0 ml of a 0.5 per cent w/v solution of N- (1-naphthyl) cent w/v solution of ammonium sulphamate, shake, allow to
ethylenediamine dihydrochloride. Any pinkish violet colour stand for 3 minutes and add 2 ml of a 0.5 per cent w/v solution
produced is not more intense than that obtained in a solution of N- (1-naphthyl) ethylenediamine dihydrochloride; a violet
prepared at the same time and in the same manner using colour is produced.
10.0 ml of a solution prepared by diluting 2.0 ml of a
0.00050 per cent w/v solution of 2-chloro-4-nitroaniline in Tests
methanol to 20 ml with 1 M hydrochloric acid and beginning 2-Chloro-4-nitroaniline. Not more than 100 ppm, Boil a
at the words “add 0.5 ml of a 0.5 per cent w/v solution of quantity of the powdered tablets containing 0.1 g of
sodium nitrite.....”. Niclosamide with 20 ml of methanol and 20 ml of a solution in
5-Chlorosalicylic acid. Not more than 60 ppm, determined by methanol containing 10 µg of 2-chloro-4-nitroaniline, cool,
the following method. Boil 1.0 g with 15 ml of water for add 45 ml of 1 M hydrochloric acid, heat to boiling, cool, filter
2 minutes, cool, filter through a membrane filter (pore size and dilute the filtrate to 50.0 ml with 1 M hydrochloric acid.
0.45 µm), wash the filter and dilute the combined filtrate and To 10.0 ml of this solution add 0.5 ml of a 0.5 per cent w/v
washings to 20 ml with water (solution A). Dissolve 30 mg of solution of sodium nitrite and allow to stand for 3 minutes.
5-chlorosalicylic acid in 20 ml of methanol and add sufficient Add 1.0 ml of a 2 per cent w/v solution of ammonium
water to produce 100.0 ml. Dilute 1.0 ml of this solution to sulphamate, shake, allow to stand for 3 minutes and add
100.0 ml with water (solution B). To 10.0 ml of each of solutions 1.0 ml of a 0.5 per cent w/v solution of N- (1-naphthyl)
A and B add separately 0.1 ml of ferric chloride solution; any ethylenediamine dihydrochloride. Any pinkish violet colour
violet colour produced in solution A is not more intense than produced is not more intense than that obtained in a solution
that obtained in solution B. prepared at the same time and in the same manner using
10.0 ml of a solution prepared by diluting 2.0 ml of a 0.0005 per
Sulphated ash (2.3.18). Not more than 0.1 per cent.
cent w/v solution of 2-chloro-4-nitroaniline in methanol to
Loss on drying (2.4.19). Not more than 0.5 per cent, determined 20.0 ml with 1 M hydrochloric acid and beginning at the
on 1.0 g by drying in an oven at 105° for 4 hours. words “add 0.5 ml of a 0.5 per cent w/v solution of sodium
nitrite.....”.
Assay. Weigh accurately about 0.3 g, dissolve in 80 ml of a
mixture of equal volumes of acetone and methanol. Titrate 5-Chlorosalicylic acid. Boil a quantity of the powdered tablets
with 0.1 M tetrabutylammonium hydroxide, determining the containing 0.5 g of Niclosamide with 10 ml of water for
end-point potentiometrically (2.4.25). Carry out a blank titration. 2 minutes, cool, filter and to the filtrate add 0.2 ml of ferric
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to chloride solution; no red or violet colour is produced.
0.03271 g of C13H8Cl2N2O4. Disintegration. The test does not apply.
Storage. Store protected from light and moisture. Other Tests. Comply with the tests stated under Tablets.

823
NICLOTINAMIDE IP 2007

Assay. Weigh and powder 20 tablets. Weigh accurately a Mobile phase. A mixture of 48 volumes of chloroform,
quantity of the powdered tablets containing about 0.3 g of 45 volumes of ethanol and 4 volumes of water.
Niclosamide dissolved in 60 ml of dimethylformamide. Titrate Test solution. Dissolve 0.8 g of the substance under
with 0.1 M tetrabutylammonium hydroxide, determining the examination in 10 ml of ethanol (50 per cent).
end-point potentiometrically (2.4.25). Carry out a blank titration.
Reference solution. A 0.02 per cent w/v solution of the
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to substance under examination in ethanol (50 per cent).
0.03271 g of C13H8Cl2N2O4.
Apply to the plate 5 µl of each solution. Allow the mobile
Storage. Store protected from light and moisture. phase to rise 10 cm. Dry the plate in air and examine in ultraviolet
Labelling. The label states that the tablets should be chewed light at 254 nm. Any secondary spot in the chromatogram
thoroughly before swallowing. obtained with the test solution is not more intense than the
spot in the chromatogram obtained with the reference solution.
Heavy metals (2.3.13). Dissolve 0.67 g in 10 ml of water, 7.5 ml
Nicotinamide of 1 M hydrochloric acid and sufficient water to produce
25 ml. The solution complies with the limit test for heavy metals,
Niacinamide Method A (30 ppm).
N Chlorides (2.3.12). 1.0 g complies with the limit test for
chlorides (250 ppm).
NH2
Sulphates (2.3.17). 1.2 g complies with the limit test for
O sulphates (125 ppm).
C6H6N2O Mol. Wt. 122.1 Sulphated ash (2.3.18). Not more than 0.1 per cent.
Nicotinamide is pyridine-3-carboxamide. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying over phosphorus pentoxide at a pressure
Nicotinamide contains not less than 99.0 per cent and not
of 1.5 to 2.7 kPa for 18 hours.
more than 101.0 per cent of C6H6N2O, calculated on the dried
basis. Assay. Weigh accurately about 0.25 g, dissolve in 20 ml of
anhydrous glacial acetic acid, heating slightly if necessary.
Description. Colourless crystals or a white, crystalline powder;
Add 5 ml of acetic anhydride. Titrate with 0.1 M perchloric
odour, faint and characteristic.
acid, using crystal violet solution as indicator. Carry out a
Identification blank titration.

Test A may be omitted if tests B, C and D are carried out. Tests 1 ml of 0.1 M perchloric acid is equivalent to 0.01221 g of
B, C and D may be omitted if test A is carried out. C6H6N2O.
A. Determine by infrared absorption spectrophotometry (2.4.6). Storage. Store protected from moisture.
Compare the spectrum with that obtained with nicotinamide
RS or with the reference spectrum of nicotinamide.
B. Heat about 5 mg in a dry tube; pyridine is evolved.
Nicotinamide Tablets
C. Boil 0.1 g with 1 ml of dilute sodium hydroxide solution; Niacinamide Tablets
ammonia is evolved. Nicotinamide Tablets contain not less than 92.5 per cent and
D. To 2 ml of a 0.1 per cent w/v solution add 6 ml of cyanogen not more than 107.5 per cent of the stated amount of
bromide solution and 1 ml of a 2.5 per cent w/v solution of nicotinamide, C6H6N2O.
aniline; a golden yellow colour develops.
Identification
Tests
Shake a quantity of the powdered tablets containing 0.2 g of
Appearance of solution. A 5.0 per cent w/v solution in carbon Nicotinamide with 50 ml of ethanol for 15 minutes, filter and
dioxide-free water is clear (2.4.1), and not more intensely evaporate the filtrate to dryness on a water-bath. The residue
coloured than reference solution BYS7 (2.4.1). complies with the following tests.
pH (2.4.24). 6.0 to 7.5, determined in a 5.0 per cent w/v solution. A. Determine by infrared absorption spectrophotometry (2.4.6).
Related substances. Determine by thin-layer chromatography Compare the spectrum with that obtained with nicotinamide
(2.4.17), coating the plate with silica gel GF254. RS or with the reference spectrum of nicotinamide.

824
IP 2007 NICLOTINIC ACID

B. Boil 0.1 g with 1 ml of dilute sodium hydroxide solution; Nicotinic Acid contains not less than 99.5 per cent and not
ammonia is evolved. more than 100.5 per cent of C6H5NO2, calculated on the dried
C. To 2 ml of a 0.1 per cent w/v solution add 6 ml of cyanogen basis.
bromide solution and 1 ml of a 2.5 per cent w/v solution of Description. A white or creamy-white, crystalline powder.
aniline; a golden yellow colour develops.
D. When examined in the range 230 nm to 360 nm (2.4.7), the
Identification
solution obtained in the Assay shows an absorption maximum Test A may be omitted if tests B, C and D are carried out. Tests
only at about 262 nm and two shoulders at about 258 nm and B, C and D may be omitted if test A is carried out.
269 nm.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Tests Compare the spectrum with that obtained with nicotinic acid
RS or with the reference spectrum of nicotinic acid.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. B. Heat a small quantity with twice its weight of soda lime;
pyridine is evolved.
Mobile phase. A mixture of 48 volumes of chloroform,
45 volumes of ethanol and 4 volumes of water. C. Dissolve about 50 mg in 20 ml of water, neutralise to litmus
paper with 0.1 M sodium hydroxide, add 3 ml of copper
Test solution. Shake a quantity of the powdered tablets sulphate solution; a blue precipitate is gradually produced.
containing 0.1 g of Nicotinamide with 15 ml of ethanol for
15 minutes, filter, evaporate to dryness on a water-bath and D. To 2 ml of a 0.1 per cent w/v solution add 6 ml of cyanogen
dissolve the residue as completely as possible in 1 ml of bromide solution and 1 ml of a 2.5 per cent w/v solution of
ethanol. aniline; a golden yellow colour is produced.

Reference solution. Dilute 1 volume of the test solution to Tests


400 volumes with ethanol
Related substances. Determine by thin-layer chromatography
Apply to the plate 5 µl of each solution. Allow the mobile
(2.4.17), coating the plate with silica gel GF254.
phase to rise 10 cm. Dry the plate in air and examine in ultraviolet
light at 254 nm. Any secondary spot in the chromatogram Mobile phase. A mixture of 85 volumes of 1-propanol,
obtained with the test solution is not more intense than the 10 volumes of anhydrous formic acid and 5 volumes of water.
spot in the chromatogram obtained with the reference solution. Test solution. Dissolve 0.2 g of the substance under
Other Tests. Comply with the tests stated under Tablets. examination in 10 ml of water. Warm slightly, if necessary.
Assay. Weigh and powder 20 tablets. Weigh accurately a Reference solution. A 0.01 per cent w/v solution of the
quantity of the powder containing about 50 mg of substance under examination in water.
Nicotinamide, shake with 50 ml of ethanol (95 per cent) for 15 Apply to the plate 5 µl of each solution. After development,
minutes and dilute to 100.0 ml with ethanol (95 per cent). Mix, dry the plate at 105° for 10 minutes and examine in ultraviolet
filter, dilute 5.0 ml of the filtrate to 100.0 ml with ethanol (95 light at 254 nm. Any secondary spot in the chromatogram
per cent) and measure the absorbance of the resulting solution obtained with the test solution is not more intense than the
at the maximum at about 262 nm (2.4.7). Calculate the content spot in the chromatogram obtained with the reference solution.
of C6H6N2O taking 241 as the specific absorbance at 262 nm.
Heavy metals (2.3.13). Mix 1.0 g with 1.5 ml of dilute
Storage. Store protected from light and moisture. hydrochloric acid and sufficient water to produce 25 ml, heat
gently and cool to room temperature. The resulting solution
complies with the limit test for heavy metals, Method B
Nicotinic Acid (20 ppm).
Chlorides (2.3.12). 1.0 g complies with the limit test for
Niacin
chlorides (250 ppm).
N Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
COOH on 1.0 g by drying in an oven at 105° for 1 hour.
C6H5NO2 Mol. Wt. 123.1
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of
Nicotinic Acid is pyridine-3-carboxylic acid. carbon dioxide-free water and titrate with 0.1 M sodium

825
NICLOTINIC TABLETS IP 2007

hydroxide using phenol red solution as indicator. Repeat the stand for 15 minutes, swirling occasionally, and then shake
operation without the substance under examination. The for 10 minutes. Filter through a plug of cotton and wash the
difference between the titrations represents the amount of filter with ethanol (95 per cent). Add 50 ml of carbon dioxide-
sodium hydroxide required. free water and titrate with 0.1 M sodium hydroxide using
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01231 g of phenol red solution as indicator.
C6H5NO2. 1 ml of 0.1 M sodium hydroxide is equivalent to 0.01231 g of
C6H5NO2.
Storage. Store protected from light and moisture.
Storage. Store protected from light and moisture.

Nicotinic Tablets
Niacin Acid Tablets Nicoumalone
Nicotinic Acid Tablets contain not less than 92.5 per cent and Acenocoumarol
not more than 107.5 per cent of the stated amount of nicotinic
acid, C6H5NO2. O O NO2

Identification
A. Determine by thin-layer chromatography (2.4.17), coating OH CH3
the plate with silica gel GF254.
O
Mobile phase. A mixture of 48 volumes of chloroform,
45 volumes of ethanol (95 per cent) and 8 volumes of water. C19H15NO6 Mol. Wt. 353.3
Test solution. Shake a quantity of the powdered tablets Nicoumalone is (RS)-4-hydroxy-3-[1-(4-nitrophenyl)-3-
containing 50 mg of Nicotinic Acid with 50 ml of hot ethanol oxobutyl]coumarin.
(95 per cent), filter and allow the filtrate to cool. Nicoumalone contains not less than 98.5 per cent and not
Reference solution. A 0.1 per cent w/v solution of nicotinic more than 100.5 per cent of C19H15NO6, calculated on the dried
acid RS in ethanol (95 per cent). basis.
Apply to the plate 5 µl of each solution. After development, Description. A white to brownish-white powder; odourless or
dry the plate in air and examine in ultraviolet light at 254 nm. almost odourless.
The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained Identification
with the reference solution. A. Determine by infrared absorption spectrophotometry (2.4.6).
B. Triturate a quantity of the powdered tablets containing Compare the spectrum with that obtained with nicoumalone
50 mg of Nicotinic Acid with 10 ml of water and filter. To 2 ml RS or with the reference spectrum of nicoumalone.
of the filtrate add 6 ml of cyanogen bromide solution and 1 ml B. When examined in the range 230 nm to 360 nm (2.4.7), a
of a 2.5 per cent w/v solution of aniline; a golden yellow 0.001 per cent w/v solution in a mixture of 9 volumes of
precipitate is produced. methanol and 1 volume of 1 M hydrochloric acid shows
C. Shake a quantity of the powdered tablets containing 0.1 g absorption maxima at about 283 nm and 306 nm; absorbances
of Nicotinic Acid with ethanol (95 per cent), filter and at the maxima, about 0.65 and about 0.52, respectively.
evaporate the filtrate to dryness. Add to the residue 10 mg of C. Heat 50 mg with 2.5 ml of glacial acetic acid, 0.5 ml of
citric acid and 0.15 ml of acetic anhydride and heat on a hydrochloric acid and 0.2 g of zinc powder on a water-bath
water-bath; a reddish-violet colour is produced. for 5 minutes, cool and filter. To the filtrate add 0.05 ml of
Tests sodium nitrite solution and add the mixture to 10 ml of a 1 per
cent w/v solution of 2-naphthol containing 3 ml of 5 M sodium
Other Tests. Comply with the tests stated under Tablets. hydroxide; a bright red precipitate is produced.
Assay. Weigh and powder 20 tablets. Weigh accurately a Tests
quantity of the powder containing about 0.25 g of Nicotinic
Acid, add 40 ml of hot ethanol (95 per cent), previously Appearance of solution. A 2.0 per cent w/v solution in acetone
neutralised to phenolphthalein solution, and shake. Allow to is clear (2.4.1).

826
IP 2007 NICOUMALONE TABLETS

B. A 2.0 per cent w/v solution in 0.1 M sodium hydroxide is On the residue determine by infrared absorption
clear (2.4.1), and yellow. spectrophotometry (2.4.6). Compare the spectrum with that
Related substances. Determine by thin-layer chromatography obtained with nicoumalone RS or with the reference spectrum
(2.4.17), coating the plate with silica gel GF254. of nicoumalone.
B. When examined in the range 230 nm to 360 nm (2.4.7), the
Mobile phase. A mixture of 50 volumes of chloroform,
final solution obtained in the Assay shows absorption maxima
50 volumes of cyclohexane and 20 volumes of glacial acetic
at about 283 nm and 306 nm.
acid.
C. Heat 50 mg of the residue obtained in test A, with 2.5 ml of
Test solution. Dissolve 0.2 g of the substance under
glacial acetic acid, 0.5 ml of hydrochloric acid and 0.2 g of
examination in 10 ml of acetone.
zinc powder on a water-bath for 5 minutes, cool and filter. To
Reference solution. A 0.002 per cent w/v solution of the the filtrate add 0.05 ml of sodium nitrite solution and add the
substance under examination in acetone. mixture to 10 ml of a 1 per cent w/v solution of 2-naphthol
Apply to the plate 20 µl of each solution. After development, containing 3 ml of 5 M sodium hydroxide; a bright red
dry the plate in air and immediately examine in ultraviolet light precipitate is produced.
at 254 nm. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the Tests
chromatogram obtained with the reference solution. Related substances. Determine by thin-layer chromatography
Sulphated ash (2.3.18). Not more than 0.1 per cent. (2.4.17), coating the plate with silica gel GF254.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Mobile phase. A mixture of 50 volumes of chloroform,
on 1.0 g by drying in an oven at 105°. 50 volumes of cyclohexane and 20 volumes of glacial acetic
acid.
Assay. Weigh accurately about 0.75 g, dissolve in 50 ml of
acetone and titrate with 0.1 M sodium hydroxide using Test solution. Shake a quantity of the powdered tablets
bromothymol blue solution as indicator. Repeat the operation containing 20 mg of Nicoumalone with 5 ml of acetone,
without the substance under examination. The difference centrifuge and use the supernatant liquid.
between the titrations represents the amount of sodium Reference solution. Dilute 1 volume of the test solution to
hydroxide required. 200 volumes with acetone.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03533 g of Apply to the plate 20 µl of each solution. After development,
C19H15NO6. dry the plate in air and immediately examine in ultraviolet light
Storage. Store protected from light. at 254 nm. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Uniformity of content. Comply with the test stated under
Nicoumalone Tablets Tablets.
Acenocoumarol Tablets Finely crush one tablet, add 30 ml of methanol, stir the mixture
Nicoumalone Tablets contain not less than 90.0 per cent and for 30 minutes and filter through sintered glass, washing the
not more than 110.0 per cent of the stated amount of residue with three quantities, each of 15 ml, of methanol. To
nicoumalone, C19H15NO6. the combined filtrate and washings add 10 ml of 1 M
hydrochloric acid and sufficient methanol to produce
Identification 100.0 ml. If necessary, dilute further with a solvent prepared
by diluting 1 volume of 1 M hydrochloric acid to 10 volumes
A. Heat a quantity of the powdered tablets containing 50 mg with methanol to produce a solution containing about
of Nicoumalone with 30 ml of acetone under a reflux condenser 0.001 per cent w/v solution of Nicoumalone. Measure the
for 5 minutes, filter and wash the residue with two quantities, absorbance of the resulting solution at the maximum at about
each of 10 ml, of acetone. Evaporate the combined filtrate and 306 nm (2.4.7). Calculate the content of C19H15NO6 taking 521
washings to 5 ml, add water dropwise until the solution as the specific absorbance at 306 nm.
becomes turbid, heat on a water-bath until the solution is
Other Tests. Comply with the tests stated under Tablets.
clear and allow to stand. Filter, wash the crystals with a mixture
of equal volumes of acetone and water and dry at 100° at a Assay. Weigh and powder 20 tablets. Weigh accurately a
pressure of 2 kPa for 30 minutes. quantity of the powder containing about 10 mg of

827
NIFEDIPINE IP 2007

Nicoumalone, add 30 ml of methanol, stir the mixture for the peak due to nifedipine in the chromatogram obtained with
30 minutes and filter through sintered-glass, washing the the reference solution.
residue with three quantities, each of 15 ml, of methanol. To C. Determine by thin-layer chromatography (2.4.17), coating
the combined filtrate and washings add 10 ml of 1 M the plate with silica gel GF254.
hydrochloric acid and sufficient methanol to produce
100.0 ml. Dilute 5.0 ml of this solution to 50.0 ml with a solvent Mobile phase. A mixture of 60 volumes of cyclohexane and
prepared by diluting 1 volume of 1 M hydrochloric acid to 40 volumes of ethyl acetate.
10 volumes with methanol and measure the absorbance of the Test solution. Dissolve 0.1 g of the substance under
resulting solution at the maximum at about 306 nm (2.4.7). examination in 100 ml of methanol.
Calculate the content of C19H15NO6 taking 521 as the specific
absorbance at 306 nm. Reference solution. A 0.1 per cent w/v solution of nifedipine
RS in methanol.
Storage. Store protected from light and moisture.
Apply to the plate 5 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Nifedipine with the reference solution.
D. To 25 mg add 10 ml of a mixture of 5 volumes of ethanol
H (95 per cent), 3.5 volumes of water and 1.5 volumes of
H3C N CH3 hydrochloric acid and dissolve with gentle heating. Add
0.5 g of granulated zinc and allow to stand for 5 minutes,
H3CO OCH3
swirling occasionally. Filter, add 5 ml of a 1 per cent w/v solution
O O of sodium nitrite to the filtrate and allow to stand for 2 minutes.
NO2 Add 2 ml of a 5 per cent w/v solution of ammonium sulphamate,
shake vigorously with care and add 2 ml of a 0.5 per cent w/v
solution of N-(1- naphthyl) ethylenediamine dihydrochloride;
an intense red colour develops which persists for more than
C17H18N2O6 Mol. Wt. 346.3
5 minutes.
Nifedipine is dimethyl 1,4-dihydro-2,6-dimethyl-4-(2-
nitrophenyl)pyridine-3,5-dicarboxylate. Tests
Nifedipine contains not less than 98.0 per cent and not more Related substances. Determine by liquid chromatography
than 102.0 per cent of C17H18N2O6, calculated on the dried (2.4.14)
basis.
Test solution. Dissolve 0.2 g of the substance under
Description. A yellow, crystalline powder; readily affected by examination in 20 ml of methanol and dilute to 50 ml with the
exposure to light. mobile phase.
NOTE — Nifedipine, when exposed to daylight and certain Reference solution (a). Dissolve an accurately weighed
wavelengths of artificial light, readily converts to a quantity of nifedipine RS in sufficient methanol to produce a
nitrosophenyl derivative. Exposure to ultraviolet light leads 1.0 per cent w/v solution and dilute quantitatively with the
to the formation of a nitrophenyl derivative. Perform the mobile phase to obtain a 0.4 per cent w/v solution.
tests and assay in the dark or under long-wavelength light
Reference solution (b). A 0.04 per cent w/v solution of
(greater than 420 nm). Use low-actinic glassware.
dimethyl-2,6-dimethyl-4-(2-nitrophenyl)pyridine-3,5-
Identification dicaboxylate RS (nitrophenylpyridine analogue) in methanol.
Reference solution (c). A 0.04 per cent w/v solution of
Test A may be omitted if tests B, C and D are carried out. Tests
dimethyl-2,6-dimethyl-4-(2-nitrosophenyl)pyridine-3,5-
B, C and D may be omitted if test A is carried out.
dicarboxylate RS (nitroso- phenylpyridine analogue) in
A. Determine by infrared absorption spectrophotometry (2.4.6). methanol.
Compare the spectrum with that obtained with nifedipine RS
Reference solution (d). Mix 1 volume of each of reference
or with the reference spectrum of nifedipine.
solutions (b) and (c) and 0.1 volume of the test solution, dilute
B. In the test for Related substances, the principal peak in the to 10 volumes with the mobile phase and then dilute 2 volumes
chromatogram obtained with the test solution corresponds to of the resulting solution to 10 volumes with the mobile phase.

828
IP 2007 NIFEDIPINE CAPSULES

Chromatographic system Nifedipine Capsules


– a stainless steel column 15 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm), Nifedipine Capsules contain not less than 90.0 per cent and
– mobile phase: 55 volumes of water, 36 volumes of not more than 110.0 per cent of the stated amount of nifedipine,
methanol and 9 volumes of acetonitrile, C17H18N2O6.
– flow rate. 1 ml per minute, NOTE — Nifedipine, when exposed to daylight and certain
– spectrophotometer set at 235 nm, wavelengths of artificial light, readily converts to a
– a 20 µl loop injector. nitrosophenyl derivative. Exposure to ultraviolet light leads
Inject reference solution (d). The peaks appear in the order to the formation of a nitrophenyl derivative. Perform the
nitrophenylpyridine analogue, nitrosophenylpyridine tests and assay in the dark or under long-wavelength light
analogue and nifedipine, which has a retention time of about (greater than 420 nm). Use low-actinic glassware.
15.5 minutes. The test is not valid unless, in the chromatogram
obtained with reference solution (d), (a) the resolution factor
Identification
between the peaks due to the nitrophenylpyridine analogue Determine by thin-layer chromatography (2.4.17), coating the
and the nitrosophenylpyridine analogue is greater than 1.5, plate with silica gel GF254.
(b) the resolution between the peaks due to the
Mobile phase. A mixture of equal volumes of ethyl acetate
nitrosophenylpyridine analogue and nifedipine is greater than
and cyclohexane.
1.5, and (c) the height of the peak due to the nitrophenyl-
pyridine analogue is at least 20 per cent of the full-scale Test solution. Transfer a quantity of the contents of the capsules
deflection. containing 30 mg of Nifedipine into a centrifuge tube containing
0.1 M sodium hydroxide, add 25 ml of dichloromethane,
Inject the test solution and reference solutions (a) and (d) and stopper the tube and shake gently for 1 hour. Centrifuge for
record the chromatograms for twice the retention time of 10 minutes at 2000 to 2500 rpm. Remove the supernatant
nifedipine. In the chromatogram obtained with the test solution aqueous layer by aspiration with a syringe and transfer 5 ml of
no secondary peak other than any peaks corresponding to the clarified lower layer to a suitable vial.
the nitrophenylpyridine analogue and the nitrosophenylpyridine
analogue has an area greater than that of the peak due to Reference solution (a). A 0.12 per cent w/v solution of
nifedipine in the chromatogram obtained with reference solution nifedipine RS in dichloromethane.
(d) and the areas of any peaks corresponding to the Reference solution (b). A mixture of equal volumes of test
nitrophenylpyridine analogue and the nitrosophenylpyridine solution and reference solution (a).
analogue are not greater than the areas of the corresponding
Apply to the plate 500 µl of each solution as bands 20 mm by
peaks in the chromatogram obtained with reference solution
3 mm. After development, dry the plate in air until the solvent
(d). The total amount of related substances is not greater than
is not detectable and immediately examine in ultraviolet light
0.3 per cent. Ignore any peak with an area less than 10 per cent
at 254 nm. The principal band, appearing as a dark blue band,
of the area of the peak due to nifedipine in the chromatogram
in the chromatogram obtained with the test solution
obtained with reference solution (d).
corresponds to that in the chromatogram obtained with the
Heavy metals (2.3.13). 2.0 g complies with limit test for heavy reference solution. Spray with a solution prepared in the
metals, Method B (10 ppm). following manner. Dissolve 3 g of bismuth subnitrate and 30 g
Sulphated ash (2.3.18). Not more than 0.1 per cent. of potassium iodide in 10 ml of 3 M hydrochloric acid and
dilute with water to 100 ml; dilute 10 ml to 100 ml with 0.3 M
Loss on drying (2.4.19). Not more than 0.5 per cent, determined hydrochloric acid. In the chromatogram obtained with test
on 1.0 g by drying in an oven at 105° for 2 hours. solution the principal band, appearing as a compact light
Assay. Weigh accurately about 0.13 g, dissolve in a mixture of orange band against a yellow background, corresponds to
25 ml of 2-methyl-2-propanol and 25 ml of 1 M perchloric that in the chromatogram obtained with reference solution (a).
acid and titrate with 0.1 M ceric ammonium sulphate, using The band obtained with reference solution (b) appears as a
0.1 ml of ferroin solution as indicator until the pink colour is single band under both visualisation procedures.
discharged, titrating slowly towards the end-point. Carry out Tests
a blank titration.
Uniformity of content. Comply with the test stated under
1 ml of 0.1 M ceric ammonium sulphate is equivalent to Capsules.
0.01732 g of C17H18N2O6.
Transfer the contents of a capsule quantitatively to a 200-ml
Storage. Store protected from light. volumetric flask with the aid of methanol, dilute to volume

829
NIFEDIPINE SUSTAINED-RELEASE TABLETS IP 2007

with methanol and mix. Complete the Assay beginning at the with the reference solution. Spray with a solution prepared in
words “Measure the absorbance....” and calculate the content the following manner. Dissolve 3 g of bismuth subnitrate and
of C17H18N2O6 in the capsule. 30 g of potassium iodide in 10 ml of 3 M hydrochloric acid
Other Tests. Comply with the tests stated under Capsules. and dilute to 100 ml with water; dilute 10 ml of this solution to
100 ml with 0.3 M hydrochloric acid. In the chromatogram
Assay. Transfer the contents of 5 capsules containing about obtained with the test solution the principal band, appearing
50 mg of Nifedipine quantitatively to a 200-ml volumetric flask as a compact light orange band against a yellow background,
with the aid of small quantities of methanol. Dilute to volume corresponds to that in the chromatogram obtained with
with methanol and mix. To 20.0 ml add sufficient methanol to reference solution.
produce 100.0 ml and mix. Measure the absorbance of the
resulting solution at the maximum at about 350 nm (2.4.7). Tests
Calculate the content of C17H18N2O6 in the capsules from the
absorbance obtained by repeating the operation with a Dissolution (2.5.2)
0.005 per cent w/v solution of nifedipine RS in methanol. A. Apparatus No. 1
Storage. Store protected from light. Medium. 900 ml of 0.1 M hydrochloric acid
Speed and time. 150 rpm and 120 minutes.
Withdraw a suitable volume of the medium and filter. Measure
the absorbance of the filtrate, suitably diluted with the
Nifedipine Sustained-release Tablets dissolution medium, if necessary, at the maximum at about 340
nm (2.4.7).
Nifedipine Sustained-release Tablets contain not less than
90.0 per cent and not more than 110.0 per cent of the stated Calculate the content of C17H18N2O6 in the medium from the
amount of nifedipine, C17H18N2O6. absorbance obtained from a solution of known concentration
of nifedipine RS in the same medium.
NOTE - Nifedipine, when exposed to daylight and certain
wavelengths of artificial light, readily converts to a D. Not less than 25 per cent and not more than 45 per cent of
nitrosophenyl derivative. Exposure to ultraviolet light leads the stated amount of C17H18N2O6.
to the formation of a nitrophenyl derivative. Perform the
B. Apparatus No. 1
tests and the assay in the dark or under long-wavelength
light (greater than 420 nm). Use low-actinic glassware. Medium. 900 ml of phosphate buffer pH 6.8
Speed and time. 150 rpm and 6 hours.
Identification
Withdraw a suitable volume of the medium and filter. Measure
Determine by thin-layer chromatography (2.4.17), coating the the absorbance of the filtrate, suitably diluted with the
plate with silica gel GF254. dissolution medium, if necessary, at the maximum at about 340
Mobile phase. A mixture of equal volumes of ethyl acetate nm (2.4.7).
and cyclohexane. Calculate the content of C17H18N2O6 in the medium from the
Test solution. Transfer a quantity of the contents of the capsules absorbance obtained from a solution of known concentration
containing 30 mg of Nifedipine into a centrifuge tube containing of nifedipine RS in the same medium.
0.1 M sodium hydroxide, add 25 ml of dichloromethane,
D. Not less than 60 per cent of the stated amount of
stopper the tube and shake gently for 1 hour. Centrifuge for 10
C17H18N2O6.
minutes at 2000 rpm to 2500 rpm. Remove the supernatant
aqueous layer by aspiration with a syringe and use 5 ml of the Other tests. Comply with the tests stated under Tablets.
clarified lower layer.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Reference solution. A 0.12 per cent w/v solution of nifedipine quantity of the powder containing about 25 mg of Nifedipine,
RS in dichloromethane. disperse in methanol, shake and dilute to 100.0 ml with
Apply to the plate 500 µl of each solution as bands 20 mm by methanol, filter. Dilute 20.0 ml of the filtrate to 100.0 ml with
3 mm. After development, dry the plate in air until the odour of methanol. Measure the absorbance of the resulting solution
the solvent is not detectable and immediately examine in at the maximum at about 350 nm (2.4.7). Calculate the content
ultraviolet light at 254 nm. The principal band, appearing as a of C17H18N2O6 from the absorbance obtained with a 0.005 per
dark blue band, in the chromatogram obtained with the test cent w/v solution of nifedipine RS in methanol.
solution corresponds to that in the chromatogram obtained Storage. Store protected from light and moisture.

830
IP 2007 NIKETHAMIDE

Nifedipine Tablets Assay beginning at the words “Measure the absorbance....”


and calculate the content of C17H18N2O6 in the tablet.
Nifedipine Tablets contain not less than 90.0 per cent and not
more than 110.0 per cent of the stated amount of nifedipine, Other Tests. Comply with the tests stated under Tablets.
C17H18N2O6. The tablets may be coated. Assay. Weigh and powder 20 tablets. Weigh accurately a
NOTE — Nifedipine, when exposed to daylight and certain quantity of the powder containing 50 mg of Nifedipine into a
wavelengths of artificial light, readily converts to a 200-ml volumetric flask. Dissolve with the aid of 50 ml of
nitrosophenyl derivative. Exposure to ultraviolet light leads methanol. Dilute to volume with methanol, mix and filter. Dilute
to the formation of a nitrophenyl derivative. Perform the 20 ml of the filtrate to 100 ml with methanol and mix. Measure
tests and assay in the dark or under long-wavelength light the absorbance of the resulting solution at the maximum at
(greater than 420 nm). Use low-actinic glassware. about 350 nm (2.4.7). Calculate the content of C17H18N2O6 from
the absorbance obtained by repeating the operation with a
Identification 0.005 per cent w/v solution of nifedipine RS in methanol.
Determine by thin-layer chromatography (2.4.17), coating the Storage. Store protected from light and moisture.
plate with silica gel GF254.
Mobile phase. A mixture of equal volumes of ethyl acetate
and cyclohexane.
Test solution. Transfer a quantity of the powdered tablets
Nikethamide
containing 30 mg of Nifedipine to a centrifuge tube containing
20 ml of 0.1 M sodium hydroxide, add 25 ml of dichloromethane, N CH3
stopper the tube and shake gently for 1 hour. Centrifuge for 10
minutes at 2000 to 2500 rpm. Remove the supernatant aqueous N CH3
layer by aspiration with a syringe and transfer 5.0 ml of the
clarified lower layer to a suitable vial. O

Reference solution (a). A 0.12 per cent w/v solution of


C10H14N2O Mol. Wt. 178.2
nifedipine RS in dichloromethane.
Nikethamide is N,N-diethylpyridine-3-carboxamide.
Reference solution (b). A mixture of equal volumes of the test
solution and reference solution (a). Nikethamide contains not less than 99.0 per cent and not more
than 101.0 per cent of C10H14N2O, calculated on the anhydrous
Apply to the plate 500 µl of each solution as bands 20 mm by
basis.
3 mm. After development, dry the plate in air until the solvent
is not detectable and immediately examine in ultraviolet light Description. A colourless or slightly yellowish, oily liquid or
at 254 nm. The principal band, appearing as a dark blue band, crystalline mass; odour, slight and characteristic.
in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with Identification
reference solution (a). Spray with a solution prepared in the
Test A may be omitted if tests B, C and D are carried out. Tests
following manner. Dissolve 3 g of bismuth subnitrate and 30 g
C and D may be omitted if tests A and B are carried out.
of potassium iodide in 10 ml of 3 M hydrochloric acid and
dilute with water to 100 ml; dilute 10 ml to 100 ml with 0.3 M A. Determine by infrared absorption spectrophotometry (2.4.6).
hydrochloric acid. In the chromatogram obtained with the Compare the spectrum with that obtained with nikethamide
test solution the principal band, appearing as a compact light RS or with the reference spectrum of nikethamide.
orange band against a yellow background, corresponds to
B. When examined in the range 230 nm to 360 nm (2.4.7), a
that in the chromatogram obtained with reference solution (a).
0.002 per cent w/v solution shows an absorption maximum
The band obtained with reference solution (b) appears as a
only at about 263 nm; absorbance at about 263 nm, about 0.57.
single band under both visualisation procedures.
C. Heat 0.1 g with 1 ml of 2 M sodium hydroxide; diethylamine,
Tests recognisable by its odour, is evolved progressively; the fumes
Uniformity of content. Comply with the test stated under turn red litmus paper blue.
Tablets D. To 2 ml of a 0.1 per cent w/v solution add 2 ml of cyanogen
Shake one tablet with methanol in a 200-ml volumetric flask, bromide solution and 3 ml of a 2.5 per cent w/v solution of
dilute to volume with methanol, mix and filter. Complete the aniline and mix; a yellow colour is produced.

831
NIKETHAMIDE INJETION IP 2007

Tests Identification
Appearance of solution. The substance, in liquid form or A. Make 1 ml alkaline with 5 M sodium hydroxide, extract with
liquefied by gentle heating, is clear (2.4.1), and not more 5 ml of dichloromethane and evaporate the solvent.
intensely coloured than reference solution YS5 (2.4.1).
On the residue determine by infrared absorption
pH (2.4.24). 6.0 to 7.8, determined in a 25.0 per cent w/v solution. spectrophotometry (2.4.6). Compare the spectrum with that
Congealing temperature (2.4.10). 23° to 24.5°. obtained with nikethamide RS or with the reference spectrum
of nikethamide.
Refractive index (2.4.27). 1.522 to1.526.
B. Gives a voluminous precipitate with alkaline potassium
Related substances. Determine by thin-layer chromatography mercuri-iodide solution and a greyish-brown flocculent
(2.4.17), coating the plate with silica gel GF254. precipitate with tannic acid solution. Gives no precipitate
Mobile phase. A mixture of 75 volumes of chloroform and with iodine solution or with potassium mercuri-iodide
25 volumes of 1-propanol. solution.
Test solution. Dissolve 0.4 g of the substance under C. Heat 1 ml with 0.2 g of sodium hydroxide; diethylamine,
examination in 10 ml of methanol. recognisable by its odour, is evolved.
Reference solution (a). A 0.04 per cent w/v solution of
ethylnicotinamide RS in methanol. Tests
Reference solution (b). A 0.004 per cent w/v solution of pH (2.4.24). 6.0 to 8.0.
ethylnicotinamide RS in methanol.
Related substances. Determine by thin-layer chromatography
Apply to the plate 10 µl of each solution. After development, (2.4.17), coating the plate with silica gel GF254.
dry the plate in air and examine in ultraviolet light at 254 nm.
Mobile phase. A mixture of 75 volumes of chloroform and
Any spot corresponding to ethylnicotinamide in the
25 volumes of 1-propanol.
chromatogram obtained with the test solution is not more
intense than the spot in the chromatogram obtained with Test solution. Dilute 1 ml of the injection to 5 ml with methanol.
reference solution (a) and any other secondary spot is not
Reference solution (a). A 0.05 per cent w/v solution of
more intense than the spot in the chromatogram obtained
ethylnicotinamide RS in methanol.
with reference solution (b).
Heavy metals (2.3.13). 2.0 g complies with limit test for heavy Reference solution (b). A 0.005 per cent w/v solution of
metals, Method B (10 ppm). ethylnicotinamide RS in methanol.

Sulphated ash (2.3.18). Not more than 0.1 per cent. Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Water (2.3.43). Not more than 0.3 per cent, determined on Any spot corresponding to ethylnicotinamide in the
2.0 g. chromatogram obtained with the test solution is not more
Assay. Weigh accurately about 0.15 g, dissolve in 20 ml of intense than the spot in the chromatogram obtained with
anhydrous glacial acetic acid and 5 ml of acetic anhydride. reference solution (a) and any other secondary spot is not
Titrate with 0.1 M perchloric acid, determining the end-point more intense than the spot in the chromatogram obtained
potentiometrically (2.4.25). Carry out a blank titration. with reference solution (b).
1 ml of 0.1 M perchloric acid is equivalent to 0.01782 g of Other tests. Complies with the tests stated under Parenteral
C10H14N2O. Preparations (Injections).
Storge. Store protected from light. Assay. Dilute 5.0 ml to 500.0 ml with water. To 5.0 ml of the
solution add 5 ml of 1 M hydrochloric acid and sufficient
water to produce 500.0 ml. Measure the absorbance of the
Nikethamide Injection resulting solution at the maximum at about 263 nm (2.4.7).
Calculate the content of C10H14N2O taking 282 as the specific
Nikethamide Injection is a sterile solution containing 25 per absorbance at 263 nm.
cent w/v solution of Nikethamide in Water for Injections.
Storage. Store protected from light, in single dose containers.
Nikethamide Injection contains not less than 24.0 per cent
and not more than 26.0 per cent w/v solution of nikethamide,
C10H14N2O.

832
IP 2007 NITRAZEPAM TABLETS

Nitrazepam Test solution. Dissolve 0.2 g of the substance under


examination in 10 ml of acetone.
O Reference solution. A 0.002 per cent w/v solution of the
HN substance under examination in acetone.
Apply to the plate 10 µl of each solution. Allow the mobile
N phase to rise 12 cm. Dry the plate in air and examine in ultraviolet
O2N light at 254 nm. Any secondary spot in the chromatogram
obtained with the test solution is not more intense than the
spot in the chromatogram obtained with the reference solution.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
C15H11N3O3 Mol. Wt. 281.3 heavy metals, Method B (20 ppm).
Nitrazepam is 1,3-dihydro-7-nitro-5-phenyl-2H-1,4- Sulphated ash (2.3.18). Not more than 0.1 per cent.
benzodiazepin-2-one. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Nitrazepam contains not less than 99.0 per cent and not more on 1.0 g by drying in an oven at 105° for 4 hours.
than 101.0 per cent C15H11N3O3, calculated on the dried basis. Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of
Description. A yellow, crystalline powder; odourless or almost acetic anhydride. Titrate with 0.1 M perchloric acid,
odourless. determining the end-point potentiometrically (2.4.25). Carry
out a blank titration.
Identification
1 ml of 0.1 M perchloric acid is equivalent to 0.02813 g of
Test A may be omitted if tests B, C and D are carried out. Tests C15H11N3O3.
B, C and D may be omitted if test A is carried out. Storage. Store protected from light and moisture.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with nitrazepam RS
or with the reference spectrum of nitrazepam. Nitrazepam Tablets
B. Carry out the following procedure in subdued light.
Nitrazepam Tablets contain not less than 90.0 per cent and not
When examined in the range 230 nm to 360 nm (2.4.7), a freshly more than 110.0 per cent of the stated amount of nitrazepam,
prepared 0.0005 per cent w/v solution in a 0.5 per cent w/v C15H11N3O3.
solution of sulphuric acid in methanol shows an absorption
maximum only at about 280 nm; absorbance at about 280 nm, Identification
about 0.45. Carry out the following procedure in subdued light.
C. Dissolve 10 mg in 1 ml of methanol, warming if necessary, A. When examined in the range 230 nm to 360 nm (2.4.7), the
and add 0.05 ml of 2 M sodium hydroxide; an intense yellow final solution obtained in the Assay shows an absorption
colour is produced. maximum only at about 280 nm.
D. Dissolve 20 mg in a mixture of 5 ml of hydrochloric acid B. Determine by thin-layer chromatography (2.4.17), coating
and 10 ml of water, boil for 5 minutes, cool and add 2 ml of a the plate with silica gel G.
0.1 per cent w/v solution of sodium nitrite. Allow to stand for
1 minute, add 1 ml of a 0.5 per cent w/v solution of sulphamic Mobile phase. A mixture of 100 volumes of chloroform and
acid, mix, allow to stand for 1 minute, add 1 ml of a 0.1 per cent 10 volumes of methanol.
w/v solution of N-(1-naphthyl)ethylenediamine Test solution. Shake a quantity of the powdered tablets with
dihydrochloride; a red colour is produced. sufficient methanol to produce a solution containing 5 mg of
Nitrazepam per ml, allow to settle and decant the supernatant
Tests liquid.
Related substances and decomposition products. Determine Reference solution. A 0.5 per cent w/v solution of nitrazepam
by thin-layer chromatography (2.4.17), coating the plate with RS in methanol.
silica gel GF254.
Apply to the plate 2 µl of each solution. After development,
Mobile phase. A mixture of 85 volumes of nitromethane and dry the plate in air, spray it with ethanolic sulphuric acid
15 volumes of ethyl acetate. (10 per cent v/v), heat at 105° for 10 minutes and examine in

833
NITROFURANTOIN IP 2007

ultraviolet light at 365 nm. The principal spot in the light. Add 70 ml of a 0.5 per cent v/v solution of hydrochloric
chromatogram obtained with the test solution corresponds to acid in methanol, shake for 15 minutes protected from light,
that in the chromatogram obtained with the reference solution. add sufficient of the hydrochloric acid solution to produce
C. To a quantity of the powdered tablets containing 5 mg of 100.0 ml and filter. Dilute 10.0 ml of the filtrate to 100.0 ml with
Nitrazepam add 5 ml of hydrochloric acid and 10 ml of water, the same solvent and measure the absorbance of the resulting
heat on a water-bath for 15 minutes, filter and cool. To the solution immediately at the maximum at about 280 nm (2.4.7).
clear filtrate add 1 ml of a 0.1 per cent w/v solution of sodium Calculate the content of C15H11N3O3 taking 910 as the specific
nitrite, allow to stand for 3 minutes and add 1 ml of a 0.5 per absorbance at 280 nm.
cent w/v solution of sulphamic acid. Allow to stand for Storage. Store protected from light and moisture.
3 minutes and add 1 ml of a 0.1 per cent w/v solution of N-(1-
naphthyl) ethylenediamine dihydrochloride; a red colour is
produced.
Nitrofurantoin
Tests
O
Related substances and decomposition products. Determine H
by thin-layer chromatography (2.4.17), coating the plate with O 2N O C N N NH
silica gel GF254.
Mobile phase. A mixture of 40 volumes of nitromethane, O
40 volumes of toluene and 20 volumes of chloroform. C8H6N4O5 Mol. Wt. 238.2 (anhydrous)
Test solution. Shake a quantity of the powdered tablets C8H6N4O5,H2O Mol. Wt. 256.2 (hydrous)
containing 40 mg of Nitrazepam with 25 ml of chloroform,
Nitrofurantoin is 1-(5-nitrofurfurylideneamino)imidazolidine-
filter, carefully evaporate the filtrate to dryness and dissolve
2,4-dione. It is anhydrous or contains one molecule of water
the residue in 2 ml of chloroform.
of hydration.
Reference solution. Dilute 1 volume of the test solution to
200 volumes with chloroform. Nitrofurantoin contains not less than 98.0 per cent and not
more than 102.0 per cent of C8H6N4O5, calculated on the dried
Apply to the plate 5 µl of each solution. Allow the mobile basis.
phase to rise 12 cm. Dry the plate in air and examine in ultraviolet
light at 254 nm. Any secondary spot in the chromatogram Description. Lemon yellow crystals or a crystalline powder;
obtained with the test solution is not more intense than the odourless or almost odourless.
spot in the chromatogram obtained with the reference solution.
Identification
Uniformity of content. Comply with the test stated under
Tablets. Carry out the following test in subdued light.

NOTE — Carry out the following procedure in subdued light. A. When examined in the range 230 nm to 400 nm (2.4.7), the
final solution obtained in the Assay shows absorption maxima
Powder 1 tablet, add 5 ml of water, mix and allow to stand for
at about 266 nm and 367 nm; the ratio of the absorbance at the
15 minutes protected from light. Add 70 ml of a 0.5 per cent
maximum at about 367 nm to that at the maximum at about
v/v solution of hydrochloric acid in methanol, shake for
266 nm is 1.36 to 1.42.
15 minutes protected from light, add sufficient of the
hydrochloric acid solution to produce 100.0 ml and filter. B. To 1 ml of a 0.1 per cent w/v solution in dimethylformamide
Dilute 10.0 ml of the filtrate with sufficient of the hydrochloric add 0.1 ml of 0.5 M ethanolic potassium hydroxide; a brown
acid solution to produce a solution containing 0.0005 per colour develops.
cent w/v solution of Nitrazepam. Measure the absorbance of
the resulting solution immediately at the maximum at about
Tests
280 nm (2.4.7). Calculate the content of C15H11N3O3 in the tablet Related substances. Determine by thin-layer chromatography
taking 910 as the specific absorbance at 280 nm. (2.4.17), coating the plate with silica gel HF254.
Other Tests. Comply with the tests stated under Tablets. Mobile phase. A mixture of 90 volumes of nitromethane and
Assay. Carry out the following procedure in subdued light. 10 volumes of methanol.
Weigh and powder 20 tablets. Weigh accurately a quantity of Test solution. Dissolve 0.25 g of the substance under
the powder containing about 5 mg of Nitrazepam, add 5 ml of examination in minimum volume of dimethylformamide and
water, mix and allow to stand for 15 minutes protected from dilute to 10 ml with acetone.

834
IP 2007 NITROFURAZONE

Reference solution. Dilute 1 volume of the test solution to Reference solution. Dilute 1 volume of the test solution to
100 volumes with acetone. 100 volumes with acetone.
Apply to the plate 10 µl of each solution. After development, Apply to the plate 10 µl of each solution. After development,
dry the plate in air, heat at 105° for 5 minutes and examine in dry the plate in air, heat at 105° for 5 minutes and examine in
ultraviolet light at 254 nm. Spray with phenylhydrazine ultraviolet light at 254 nm. Spray with phenylhydrazine
hydrochloride solution and heat the plate at 105° for further hydrochloride solution and heat the plate at 105° for further
10 minutes. Any secondary spot in the chromatogram obtained 10 minutes. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the with the test solution, is not more intense than the spot in the
chromatogram obtained with the reference solution by both chromatogram obtained with the reference solution by both
methods of visualisation. methods of visualisation.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Other Tests. Comply with the tests stated under Tablets.
Loss on drying (2.4.19). 5.0 to 7.1 per cent (hydrous form) and Assay. Carry out the following procedure in subdued light.
not more than 1.0 per cent (anhydrous form), determined on
1.0 g by drying in an oven at 105°. Weigh and powder 20 tablets. Weigh accurately a quantity of
the powder containing about 0.15 g of Nitrofurantoin, add
Assay. Carry out the following procedure in subdued light. 50.0 ml of dimethylformamide, shake for 5 minutes, add
Weigh accurately about 75 mg and dissolve in 25.0 ml of sufficient water to produce 1000.0 ml and mix. Dilute 5.0 ml to
dimethylformamide, add sufficient water to produce 500.0 ml 100.0 ml with a solution containing 1.8 per cent w/v solution
and mix. Dilute 5.0 ml to 100.0 ml with a solution containing of sodium acetate and 0.14 per cent v/v of glacial acetic
1.8 per cent w/v solution of sodium acetate and 0.14 per cent acid. Measure the absorbance of the resulting solution at the
v/v of glacial acetic acid. Measure the absorbance of the maximum at about 367 nm (2.4.7), using as the blank the sodium
resulting solution at the maximum at about 367 nm (2.4.7), acetate-acetic acid solution. Calculate the content of C8H6N4O5
using as the blank the sodium acetate-acetic acid solution. taking 765 as the specific absorbance at 367 nm.
Calculate the content of C8H6N4O5 taking 765 as the specific
Storage. Store protected from light and moisture.
absorbance at 367 nm.
Storage. Store protected from light and moisture.
Labelling. The label states whether the material is anhydrous
or hydrous.
Nitrofurazone
Nitofural

Nitrofurantoin Tablets H
H N NH2
Nitrofurantoin Tablets contain not less than 90.0 per cent and O2N O C N
not more than 110.0 per cent of the stated amount of O
nitrofurantoin, C8H6N4O5.
C6H6N4O4 Mol. Wt. 198.1
Identification
Nitrofurazone is 5-nitro-2-furaldehyde semicarbazone.
Carry out the following procedure in subdued light.
Nitrofurazone contains not less than 97.0 per cent and not
A. When examined in the range 230 nm to 400 nm (2.4.7), the more than 103.0 per cent of C6H6N4O4, calculated on the dried
final solution obtained in the Assay shows absorption maxima basis.
at about 266 nm and 367 nm.
Description. A yellow to brownish-yellow, crystalline powder;
Tests odourless or almost odourless.
Related substances. Determine by thin-layer chromatography
Identification
(2.4.17), coating the plate with silica gel HF254.
Mobile phase. A mixture of 90 volumes of nitromethane and A. Determine by infrared absorption spectrophotometry (2.4.6).
10 volumes of methanol. Compare the spectrum with that obtained with nitrofurazone
RS or with the reference spectrum of nitrofurazone.
Test solution. Shake a quantity of the powdered tablets
containing 0.1 g of Nitrofurantoin with 10 ml of a mixture of B. Dissolve 1 mg in 1 ml of dimethylformamide and add 0.05 ml
9 volumes of acetone and 1 volume of dimethylformamide of 1 M ethanolic potassium hydroxide; a ruby red colour is
and filter. produced.

835
NITROUS OXIDE IP 2007

Tests less than 6 hours before carrying out the tests Keep the
cylinder in the vertical position with the outlet valve
pH (2.4.24). 5.0 to 7.0, determined in the filtrate obtained by uppermost and deliver the gas at a rate of 4 litres per hour,
shaking 1.0 g with 100 ml of carbon dioxide-free water and unless otherwise directed. The test for Carbon monoxide
filtering. should be carried out on the first portion of gas drawn from
Related substances. Carry out the following procedure in the cylinder and that for Nitric oxide and nitrogen dioxide
subdued light. immediately thereafter.
Determine by thin-layer chromatography (2.4.17), coating the Description. A colourless gas; odourless.
plate with silica gel G.
Mobile phase. A mixture of 95 volumes of toluene and Identification
5 volumes of dioxan. A. A glowing splinter of wood bursts into flame on contact
Test solution. Dissolve 0.1 g of the substance under with the gas.
examination in 10 ml of a mixture of equal volumes of acetone B. Shake with alkaline pyrogallol solution; the gas being
and dimethylformamide. examined is not absorbed and the solution does not become
Reference solution (a). A 0.002 per cent w/v solution of brown.
5-nitrofurfurylidene azine RS in a mixture of equal volumes of
acetone and dimethylformamide. Tests
Reference solution (b). A 0.01 per cent w/v solution of Acidity or alkalinity. Use hermetically-closed, flat-bottomed,
nitrofurfural diacetate RS in a mixture of equal volumes of glass cylinders with dimensions such that 50 ml of liquid
acetone and dimethylformamide. reaches a height of 12 to 14 cm, fitted with an outlet tube and
Apply to the plate 10 µl of each solution. After development, with an inlet tube with an orifice of 1 mm in internal diameter
dry the plate in air, heat it at 105° for 5 minutes and spray with reaching to within 2 mm of the bottom of the cylinder. For
phenylhydrazine hydrochloride solution. In the solution (1) pass 2.0 litres of the gas under examination through
chromatogram obtained with the test solution any spots a mixture of 0.1 ml of 0.01 M hydrochloric acid and 50 ml of
corresponding to 5-nitrofurfurylidene azine and nitrofurfural carbon dioxide-free water. For solution (2) use 50 ml of carbon
diacetate are not more intense than the spots in the dioxide-free water. For solution (3) add 0.2 ml of 0.01 M
chromatograms obtained with reference solutions (a) and (b) hydrochloric acid to 50 ml of carbon dioxide-free water. To
respectively. each solution add 0.1 ml of a 0.02 per cent w/v solution of
methyl red in ethanol (70 per cent). The intensity of the colour
Sulphated ash (2.3.18). Not more than 0.1 per cent.
of solution (1) is between those of solutions (2) and (3).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Arsine and phosphine. Through a mercuric chloride paper
on 1.0 g by drying in an oven at 105°.
attached to a glass tube as in the limit test for arsenic (2.3.10),
Assay. Carry out the following procedure in subdued light. pass 2.0 litres of the gas; no visible stain is produced.
Weigh accurately about 60 mg, add 20.0 ml of Carbon dioxide. Not more than 300 ppm v/v determined by the
dimethylformamide, swirl to dissolve and add sufficient water following method. Use the apparatus described in the test for
to produce 500.0 ml. Dilute 5.0 ml of the solution to 100.0 ml Acidity or alkalinity. Pass 1.0 litre through 50 ml of clear barium
with water and mix. Measure the absorbance of the resulting hydroxide solution. Any turbidity produced in the resulting
solution at the maximum at about 375 nm (2.4.7). Calculate the solution is not more than that obtained in a reference solution
content of C6H6N4O4 taking 822 as the specific absorbance at prepared at the same time by adding 1 ml of a 0.11 per cent
375 nm. w/v solution of sodium bicarbonate in carbon dioxide-free
Storage. Store protected from light and moisture. water to 50 ml of barium hydroxide solution.
Carbon monoxide. Not more than 10 ppm v/v, determined by
the following method. Connect in series a U-tube containing
Nitrous Oxide silica gel impregnated with chromium trioxide, a drechsel
N2O Mol. Wt. 44.0 bottle containing 100 ml of a 40 per cent w/v solution of
potassium hydroxide, a U-tube containing pellets of
Nitrous Oxide contains not less than 98.0 per cent v/v of N2O. potassium hydroxide, a U-tube containing phosphorus
NOTE — Carry out the following tests on a full cylinder from pentoxide dispersed on previously granulated, fused pumice,
which no gas has been withdrawn. The cylinder from which a tube containing iodine pentoxide in granules, previously
the gas is taken should be kept at room temperature for not dried at 200° and kept at a temperature of 120°, packed in 1-cm

836
IP 2007 NORADRENALINE BITARTRATE

columns separated by 1-cm columns of glass wool giving an solution of 0.5 g of soluble starch and 0.5 g of potassium
effective length of 5 cm, and a flask containing 2.0 ml of 1 M iodide in 100 ml of water containing 0.05 ml of glacial acetic
potassium iodide and 0.15 ml of starch solution. acid; the colour of the liquid is not changed.
Flush the apparatus with 5.0 litres of carbon dioxide-free air Water. Pass a measured quantity at a rate of 6 litres per hour
and, if necessary, discharge the blue colour in the iodide through an absorption tube containing magnesium
solution by adding a small quantity of freshly prepared perchlorate; the increase in weight of the tube does not exceed
0.002 M sodium thiosulphate. Continue flushing until not 2 mg per litre of gas, the initial and final weighings of the tube
more than 0.045 ml of 0.002 M sodium thiosulphate is required being made when the air in it has been displaced by the nitrous
after passing 5.0 litres of carbon dioxide-free air. Pass 5.0 litres oxide.
of the gas under examination through the apparatus and flush Assay. Carry out the assay of nitrous oxide (2.3.32), using
the last traces of liberated iodine into the reaction flask by 100 ml of the gas under examination. Use a cylinder of the gas
passing through the apparatus 1.0 litre of carbon monoxide- under examination from which at least 1 per cent w/w of the
free air. Titrate the liberated iodine with 0.002 M sodium contents have been removed.
thiosulphate. Carry out a blank titration under the same
Storage. Store under pressure in metal cylinders of the type
conditions, using 5.0 litres of carbon dioxide-free air. The
conforming to the appropriate safety regulations and at a
difference between the volumes of 0.002 M sodium
temperature not exceeding 37°.
thiosulphate used in the two titrations is not greater than
1.0 ml. Labelling. The cylinder is painted blue and carries a label
stating “Nitrous Oxide”. In addition, “Nitrous Oxide” or the
Halogens and hydrogen sulphide. Pass a volume containing symbol “N2O” should be stencilled in paint on the shoulder of
1.0 litre measured at 25° and at 101.3 kPa through a mixture of the cylinder.
100 ml of water and 1 ml of silver nitrate solution; neither
opalescence nor darkening is produced.
Nitric oxide and nitrogen dioxide. Not more than 2 ppm v/v in Noradrenaline Bitartrate
both the liquid and gaseous phases, determined by the
following method. Use two of the cylinders described in the Noradrenaline Acid Tartrate; Levarterenol Bitartrate;
test for Acidity or alkalinity connected in series. Examine Norepinephrine Bitartrate
separately both the liquid and gaseous phases of the gas
OH
under examination. To obtain the liquid phase invert the
cylinder. The liquid vaporises on leaving the valve. NH2 H OH
, HOOC COOH , H2O
For solution A dissolve 1 g of sulphanilic acid in a mixture of
HO
10 ml of glacial acetic acid and 180 ml of water. For solution H OH
B dissolve 0.2 g of N-(1-naphthyl)ethylenediamine OH
dihydrochloride in 10 ml of a 50 per cent v/v solution of
C8H11NO3,C4H6O6,H2O Mol. Wt. 337.3
glacial acetic acid, heating gently, and dilute to 200 ml with
water. Mix 9 volumes of solution A with 1 volume of solution Noradrenaline Bitartrate is (R)-2-amino-1-(3,4-
B (reagent A). dihydroxyphenyl)ethanol hydrogen (2R,3R)-tartrate
monohydrate.
In the first cylinder place 15 ml of a solution containing 2.5 per
cent w/v solution of potassium permanganate and 1.2 per Noradrenaline Bitartrate contains not less than 98.5 per cent
cent v/v of sulphuric acid (96 per cent). Place 20 ml of reagent and not more than 101.0 per cent of C8H11NO3,C4H6O6,
A in the second cylinder and connect the outlet tube of the calculated on the anhydrous basis.
first cylinder to the inlet tube of the second cylinder. Pass Description. A white or almost white, crystalline powder;
2.5 litres of the gas under examination through the reagents at odourless. It gradually darkens on exposure to air and light.
a rate of 15 litres per hour. Prepare a reference solution by
adding 0.25 ml of a 0.00616 per cent w/v solution of sodium Identification
nitrite to 20 ml of reagent A. Allow both the sample and Test A may be omitted if tests B, C, D, E and F are carried out.
reference solutions to stand for 10 minutes. For both liquid Tests C, D, E may be omitted if tests A, B and F are carried out.
and gaseous phases, any red colour in the sample solution is
not more intense than that in the reference solution. A. Dissolve 0.2 g in 2 ml of water containing about 10 mg of
sodium sulphite and add sufficient dilute ammonia solution
Oxidising substances. Pass a volume containing 2.0 litres to give an alkaline reaction. Keep the mixture at about 4° for
measured at 25° and at 101.3 kPa through a freshly prepared 1 hour and filter.

837
NORADRENALINE BITARTRATE INJECTION IP 2007

On the residue (residue R) determine by infrared absorption Apply to the plate 6 µl of each of test solution, reference
spectrophotometry (2,4,6). Compare the spectrum with that solutions (a) and (b) and 12 µl of reference solution (c) as
obtained with noradrenaline acid tartrate RS treated in the bands 20 mm by 2 mm. Allow the applied bands to dry in air,
same manner. spray them with a saturated solution of sodium bicarbonate,
B. When examined in the range 230 nm to 360 nm (2.4.7), a allow to dry in air and spray the bands twice with acetic
0.005 per cent w/v solution in 0.01 M hydrochloric acid shows anhydride, drying between the two sprayings. Heat the plate
an absorption maximum at about 279 nm; absorbance at about at 50° for 90 minutes and develop the chromatograms. After
279 nm, about 0.40. removal of the plate, allow it to dry in air and spray with a
freshly prepared mixture of 8 volumes of methanol, 2 volumes
C. Wash residue R obtained in test A with three quantities, of ethylenediamine and 2 volumes of a 0.5 per cent w/v
each of 2 ml, of water, followed by 5 ml of ethanol (95 per solution of potassium ferricyanide. Dry the plate at 60° for
cent) and 5 ml of ether and dry the precipitate under pressure 10 minutes and examine in ultraviolet light at 254 and 365 nm.
of 1.5 to 2.5 kPa for 3 hours. The specific optical rotation In the chromatogram obtained with the test solution any band
(2.4.22), determined in a 2.0 per cent w/v solution of the dried with a slightly higher Rf value than the principal band is not
precipitate in 0.5 M hydrochloric acid is –44° to –48°. more intense than the corresponding band in the chromatogram
D. To 1 ml of a 1 per cent w/v solution, add 0.05 ml of ferric obtained with reference solution (b). The chromatogram
chloride solution; an intense green colour is produced. Add, obtained with reference solution (c) shows a clearly separated
drop by drop, sodium bicarbonate solution; the colour band corresponding to the most intense band in the
changes to blue and then red. chromatogram obtained with reference solution (a) at a higher
Rf value than the most intense band.
E. To 1 ml of a 0.1 per cent w/v solution add 10 ml of phthalate
buffer pH 3.6, add 1 ml of 0.05 M iodine, set aside for 5 minutes Noradrenalone. Absorbance of a 0.2 per cent w/v solution in
and add 2 ml of 0.1 M sodium thiosulphate; not more than a 0.01 M hydrochloric acid at 310 nm, not more than 0.40 (2.4.7).
faint red colour is produced. Repeat the test using buffer Sulphated ash (2.3.18). Not more than 0.1 per cent.
solution pH 6.6 instead of phthalate buffer pH 3.6; a strong Water (2.3.43). 4.5 to 5.8 per cent, determined on 0.5 g.
reddish violet colour is produced (distinction from adrenaline Assay. Weigh accurately about 0.6 g, dissolve in 50 ml of
and isoprenaline). anhydrous glacial acetic acid, warming if necessary.Titrate
F. The filtrate obtained in test A gives the reactions of tartrates with 0.1 M perchloric acid, using crystal violet solution as
(2.3.1). indicator, until a bluish green colour is obtained. Carry out a
blank titration.
Tests 1 ml of 0.1 M perchloric acid is equivalent to 0.03193 g of
Appearance of solution. A freshly prepared 2.0 per cent w/v C8H11NO3,C4H6O6.
solution is clear (2.4.1), and not more intensely coloured than Storage. Store protected from moisture.
reference solution BYS5 (2.4.1).
pH (2.4.24). 3.5 to 5.0, determined in a 1.0 per cent w/v solution.
Noradrenaline Bitartrate Injection
Melting range (2.4.21). 100° to 106°, with decomposition.
Noradrenaline Acid Tartrate Injection; Noradrenaline
Adrenaline. Determine by thin-layer chromatography (2.4.17),
Injection; Levarterenol Bitartrate Injection;
coating the plate with silica gel G.
Norepinephrine Bitartrate Injection
Mobile phase. A mixture of 50 volumes of acetone, 50 volumes
Noradrenaline Bitartrate Injection is a sterile solution of
of dichloromethane and 0.5 volume of anhydrous formic acid.
Noradrenaline Bitartrate. It is prepared by diluting Sterile
Prepare the following solutions immediately before use. Noradrenaline Concentrate to 250 times its volume with Sodium
Test solution. Dissolve 0.25g of the substance under Chloride and Dextrose Injection or with Dextrose Injection
examination in 10 ml of water. (5 per cent w/v) immediately before use.
Reference solution (a). A 0.125 per cent w/v solution of Noradrenaline Bitartrate Injection contains in 1 ml 8 µg of
adrenaline tartrate RS in water. Noradrenaline Bitartrate equivalent to approximately 4 µg of
noradrenaline.
Reference solution (b). A 0.025 per cent w/v solution of
adrenaline tartrate RS in water. Tests
Reference solution (c). A mixture of equal volumes of the test Other Tests. Complies with the tests stated under Parenteral
solution and reference solution (b). Preparations (Injections).

838
IP 2007 NORETHISTERONE

Sterile Noradrenaline Concentrate Description. A white or yellowish-white, crystalline powder;


Sterile Noradrenaline Concentrate is a sterile, isotonic solution odourless.
containing 0.2 per cent w/v of Noradrenaline Bitartrate in Identification
Water for Injections.
Sterile Noradrenaline Concentrate contains not less than A. Determine by infrared absorption spectrophotometry (2.4.6).
0.18 per cent and not more than 0.23 per cent w/v of Compare the spectrum with that obtained with norethisterone
noradrenaline bitartrate, C8H11NO3,C4H6O6,H2O. RS or with the reference spectrum of norethisterone.
B. Determine by thin-layer chromatography (2.4.17), coating
Identification the plate with silica gel G.
Mix 0.5 ml with 10 ml of phthalate buffer pH 3.6, add 1 ml of Solvent mixture. A mixture of 90 volumes of acetone and
0.05 M iodine, allow to stand for 5 minutes and add 2 ml of 10 volumes of formamide.
0.1 M sodium thiosulphate; not more than a very faint red
colour is produced. Repeat the test using phosphate buffer Mobile phase. A mixture of 40 volumes of hexane and
pH 6.6 instead of phthalate buffer pH 3.6; a strong reddish 10 volumes of dioxan.
violet colour is produced. Test solution. Dissolve 10 mg of the substance under
examination in 10 ml of chloroform.
Tests
Reference solution. Dissolve 10 mg of norethisterone RS in
pH (2.4.24). 3.0 to 4.6.
10 ml of chloroform.
Other Tests. Complies with the tests stated under Parenteral
Preparations (Concentrated Solutions for Injection). Place the dry plate in a tank containing a shallow layer of the
solvent mixture, allow the solvent mixture to ascend to the
Assay. Dilute 5.0 ml to 200.0 ml with water and measure the top, remove the plate from the tank and allow the solvent to
absorbance of the resulting solution at the maximum at about evaporate. Use within 2 hours, with the flow of the mobile
279 nm (2.4.7). Calculate the content of C8H11NO3,C4H6O6,H2O phase in the direction in which the aforementioned treatment
taking 80 as the specific absorbance at 279 nm. was done.
Storage. Store protected from light, in single dose containers.
Apply to the plate 2 µl of each solution. Allow the mobile
Labelling. The label states (1) “Sterile Noradrenaline phase to rise 12 cm. Dry the plate in a current of warm air, allow
Concentrate”; (2) that 1 volume of the solution diluted to the solvent to evaporate, heat at 120° for 15 minutes and spray
250 volumes with Sodium Chloride and Dextrose Injection or the hot plate with ethanolic sulphuric acid (20 per cent v/v).
with Dextrose Injection (5 per cent w/v) produces Heat at 120° for a further 10 minutes, allow to cool and examine
Noradrenaline Bitartrate Injection, which must be used in daylight and in ultraviolet light at 365 nm. The principal
immediately after preparation; (3) that if the solution is brown spot in the chromatogram obtained with the test solution
it should not be used. corresponds to and exhibits fluorescence similar to that in the
chromatogram obtained with the reference solution.
C. Dissolve about 2 mg in 2 ml of ethanol (95 per cent) and
Norethisterone add 1 ml of a 1 per cent w/v solution of butylated
Norethindrone hydroxytoluene in ethanol (95 per cent) and 2 ml of 1 M
sodium hydroxide. Heat in a water-bath for 30 minutes and
cool; a yellowish pink colour is produced.
H3C OH
C CH
Tests
H H
Appearance of solution. Dissolve 0.2 g in sufficient dioxan to
H H produce 10 ml (solution A). The solution is clear (2.4.1), and
O not more intensely coloured than reference solution YS6 (2.4.1).
C20H26O2 Mol. Wt. 298.4 Specific optical rotation (2.4.22). –33.0° to –37.0°, determined
Norethisterone is 17β-hydroxy-19-nor-17α-pregn-4-en-20-yn- in a solution prepared by diluting 5.0 ml of solution A to
3-one. 10.0 ml with dioxan.
Norethisterone contains not less than 98.0 per cent and not Light absorption. Dissolve 10 mg in sufficient ethanol
more than 102.0 per cent of C20H26O2, calculated on the dried (95 per cent) to produce 100 ml, dilute 10 ml of the solution to
basis. 100 ml with methanol (98 per cent). Absorbance of the

839
NORETHISTERONE TABLETS IP 2007

resulting solution at the maximum at about 240 nm, 0.55 to 0.59 each of 5 ml, of light petroleum (60° to 80°) and discard the
(2.4.7). washings. Extract the residue with 15 ml of chloroform,
Related substances. Determine by thin-layer chromatography evaporate the extract to dryness and recrystallise from aqueous
(2.4.17), coating the plate with silica gel GF254. methanol. The residue complies with the following test.

Mobile phase. A mixture of 90 volumes of chloroform and Determine by thin-layer chromatography (2.4.17), coating the
10 volumes of acetone. plate with silica gel G.

Test solution. Dissolve 0.5 g of the substance under Solvent mixture. A mixture of 90 volumes of acetone and
examination in 100 ml of the mobile phase. 10 volumes of 1,2-propanediol.
Reference solution (a). A 0.0025 per cent w/v solution of the Mobile phase. A mixture of 40 volumes of cyclohexane and 10
substance under examination in the mobile phase. volumes of toluene.
Reference solution (b). A solution containing 0.025 per cent Test solution. Dissolve 25 mg of the substance under
w/v each of the substance under examination and ethisterone examination in 10 ml of the solvent mixture.
RS in the mobile phase.
Reference solution (a). Dissolve 25 mg of norethisterone RS
Apply to the plate 10 µl of each solution. After development, in 10 ml of the solvent mixture.
dry the plate in air, spray with ethanolic sulphuric acid
Reference solution (b). Mix equal volumes of the test solution
(20 per cent v/v), heat at 105° for 5 minutes and examine in
and reference solution (a).
ultraviolet light at 365 nm. Any secondary spot in the
chromatogram obtained with the test solution is not more Place the dry plate in a tank containing a shallow layer of the
intense than the spot in the chromatogram obtained with solvent mixture, allow the solvent mixture to ascend to the
reference solution (a). The chromatogram obtained with top, remove the plate from the tank and allow the solvent to
reference solution (b) shows two clearly separated spots of evaporate. Use within 2 hours, with the flow of the mobile
equal intensities. phase in the direction in which the aforementioned treatment
Sulphated ash (2.3.18). Not more than 0.1 per cent. was done.

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Apply to the plate 2 µl of each solution. Allow the mobile
on 1.0 g by drying in an oven at 105° for 3 hours. phase to rise 12 cm. Dry the plate in a current of warm air, allow
the solvent to evaporate, heat at 120° for 15 minutes and spray
Assay. Weigh accurately about 0.4 g, dissolve in 40 ml of the hot plate with ethanolic sulphuric acid (20 per cent v/v).
tetrahydrofuran, add 10 ml of a 10 per cent w/v solution of Heat at 120° for a further 10 minutes, allow to cool and examine
silver nitrate and titrate with 0.1 M sodium hydroxide using in daylight and in ultraviolet light at 365 nm. The principal
2 ml of bromocresol green solution as indicator. Repeat the spot in the chromatogram obtained with the test solution
operation without the substance under examination. The corresponds to that in the chromatogram obtained with
difference between the titrations represents the amount of reference solution (a). The principal spot in the chromatogram
sodium hydroxide required. obtained with reference solution (b) appears as a single,
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02984 g of compact spot.
C20H26O2.
Tests
Storage. Store protected from light and moisture.
Uniformity of content. Comply with the test stated under
Tablets.
Norethisterone Tablets Powder one tablet and warm with about 75 ml of ethanol
(95 per cent) with stirring. Cool, transfer to a 100-ml volumetric
Norethindrone Tablets
flask and dilute to volume with ethanol (95 per cent).
Norethisterone Tablets contain not less than 90.0 per cent Centrifuge a few ml of the mixture until a clear supernatant
and not more than 110.0 per cent of the stated amount of liquid is obtained. Dilute 10.0 ml of the supernatant liquid to
norethisterone, C20H26O2. 50.0 ml with ethanol (95 per cent) and mix. Measure the
absorbance of the resulting solution at the maximum at about
Identification 240 nm (2.4.7). Calculate the content of C20H26O2 in the tablet
taking 570 as the specific absorbance at 240 nm.
Place a quantity of the powdered tablets containing 25 mg of
Norethisterone on a small filter, wash with three quantities, Other Tests. Comply with the tests stated under Tablets.

840
IP 2007 NORFLOXACIN

Assay. Weigh and powder 20 tablets. Weigh accurately a Tests


quantity of the powder containing about 0.2 g of
Norethisterone and transfer to a glass column closed at the Related substances. Determine by thin-layer chromatography
bottom with a small piece of absorbent cotton. The glass (2.4.17), coating the plate with silica gel G, previously washed
column consists of a piece of glass tubing (150 mm x 10 mm) with methanol and dried.
tapered at the bottom and sealed at the top to another piece of Mobile phase. A mixture of 40 volumes of dichloromethane,
glass tubing (150 mm x 25 mm). Place a small piece of absorbent 40 volumes of methanol, 20 volumes of toluene, 14 volumes
cotton on top of the powder, pass 200 ml of light petroleum of diethylamine and 8 volumes of water.
(60° to 80°) through the column and discard the effluent.
Extract the residue with 200 ml of chloroform, evaporate the Test solution. Dissolve 0.8 g of the substance under
chloroform from the extract and dry the residue at 105° for examination in 100 ml of a mixture of equal volumes of methanol
2 hours. Allow to cool, dissolve in 40 ml of tetrahydrofuran, and dichloromethane.
add 10 ml of a 10 per cent w/v solution of silver nitrate. Titrate Reference solution. Dissolve 4.0 mg of norfloxacin RS in 1 ml
with 0.1 M sodium hydroxide, determining the end-point of glacial acetic acid, add 4 ml of methanol and mix; dilute
potentiometrically (2.4.25). Repeat the operation without the 1 ml of the solution with 9 ml of a mixture of equal volumes of
substance under examination. The difference between the methanol and dichloromethane (reference solution A). Dilute
titrations represents the amount of sodium hydroxide required. a portion of reference solution A with an equal volume of the
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02984 g of methanol-dichloromethane mixture (reference solution B).
C20H26O2. Apply separately to the plate spots of the three solutions in
Storage. Store protected from light and moisture. quantities indicated below. For spot 1 use 5 µl of the test
solution; for spots 2, 3 and 4 use 1 µl, 1.5 µl and 2 µl respectively
of reference solution A; for spot 5 use 5 µl of reference solution
B. Place the plate in a paper-lined chamber previously
equilibrated with the mobile phase and allow the solvent front
Norfloxacin to move about nine-tenths of the length of the plate. After
development, dry the plate in air and examine in ultraviolet
light at 254 nm and 365 nm. Compare the intensities of any
CH3 secondary spots in the chromatogram obtained with the test
HN
solution with those of the principal spots (2), (3), (4) and (5).
N N The sum of the intensities of secondary spots obtained with
the test solution is not more than 0.5 per cent of impurities.
F COOH (The spots (2) (3) (4) and (5) represent 0.2 per cent, 0.3 per
O cent, 0.4 per cent and 0.5 per cent respectively of impurities).
Heavy metals (2.3.13). 1.33 g complies with the limit test for
C16H18FN3O3 Mol. Wt. 319.3 heavy metals, Method B (15 ppm).

Norfloxacin is 1-ethyl-6-fluoro-4-oxo-7-(piperazin-1-yl)-1,4- Sulphated ash (2.3.18). Not more than 0.1 per cent, determined
dihydroquinoline-3-carboxylic acid. on 1.0 g in a platinum crucible.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Norfloxacin contains not less than 99.0 per cent and not more
on 0.5 g by drying in an oven at 100° at a pressure not exceeding
than 101.0 per cent of C16H18FN3O3, calculated on the dried
0.7 kPa.
basis.
Assay. Weigh accurately about 0.3 g and dissolve in 100 ml of
Description. A white to pale yellow, crystalline powder.
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
acid, determining the end-point potentiometrically (2.4.25) and
Identification
using a suitable anhydrous electrode system. (The electrode
A. Determine by infrared absorption spectrophotometry (2.4.6). system may be rendered anhydrous by filling the electrode
Compare the spectrum with that obtained with norfloxacin with 0.1 M lithium perchlorate in acetic anhydride after
RS or with the reference spectrum of norfloxacin. removing any aqueous solution contained in it). Carry out a
blank titration.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.0005 per cent w/v solution in 0.1 M sodium hydroxide shows 1 ml of 0.1 M perchloric acid is equivalent to 0.03193 g of
an absorption maximum at about 273 nm. C16H18FN3O3.

841
NORFLOXACIN EYE DROPS IP 2007

Storage. Store protected from light and moisture. Norfloxacin Tablets


Norfloxacin Tablets contain not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
Norfloxacin Eye Drops norfloxacin, C16H18FN3O3. The tablets may be coated.
Norfloxacin Eye Drops are a sterile solution of Norfloxacin in
Purified water.
Identification

Norfloxacin Eye Drops contain not less than 90.0 per cent and A. Determine by thin-layer chromatography (2.4.17), coating
not more than 110.0 per cent of the stated amount of the plate with silica gel GF254.
norfloxacin, C16H18FN3O3. Mobile phase. A mixture of 40 volumes of chloroform,
40 volumes of methanol, 20 volumes of toluene, 14 volumes
Identification of diethylamine and 8 volumes of water.
In the Assay, the principal peak in the chromatogram obtained Test solution. Shake a quantity of the finely powdered tablets
with the test solution corresponds to the peak in the containing 75 mg of Norfloxacin with 50 ml of a mixture of
chromatogram obtained with the reference solution. equal volumes of acidified methanol (containing 0.9 per cent
v/v of hydrochloric acid) and dichloromethane, centrifuge
Tests and use the clear supernatant solution.
pH (2.4.24). 4.6 to 5.5. Reference solution. A 0.15 per cent w/v solution of norfloxacin
Other tests. Comply with the tests stated under Eye Drops. RS in the same solvent mixture.

Assay. Determine by liquid chromatography (2.4.14). Apply to the plate 50 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Test solution. Dilute a suitable volume of the eye drops with a The principal spot in the chromatogram obtained with the test
0.1 per cent v/v solution of orthophosphoric acid to produce solution corresponds to that in the chromatogram obtained
a solution containing 0.005 per cent w/v of Norfloxacin. with the reference solution.
Reference solution. A 0.005 per cent w/v solution of B. In the Assay, the principal peak in the chromatogram
norfloxacin RS in 0.1 per cent v/v solution of orthophosphoric obtained with the test solution corresponds to the peak due
acid. to norfloxacin in the chromatogram obtained with the reference
Chromatographic system solution.
– a stainless steel column 30 cm x 3.9 mm, packed with
octadecylsilyl silica gel (10 µm) (such as Bondapack Tests
C18), Dissolution (2.5.2).
– column temerature 50°,
Apparatus. No 1
– mobile phase: a mixture of 300 volumes of methanol
Medium. 750 ml of acetate buffer pH 4.0.
and 700 volumes of 0.1 per cent v/v orthophosphoric
acid, Speed and time. 50 rpm and 30 minutes.
– flow rate. 2 ml per minute, Withdraw a suitable volume of the medium and filter. Measure
– spectrophotometer set at 280 nm, the absorbance of the filtrate, suitably diluted with acetate
– a 20 µl loop injector. buffer pH 4.0, if necessary, at the maximum at about 278 nm
Precondition the column using 0.01 M anhydrous sodium (2.4.7). Concomitantly measure the absorbance of a solution
dihydrogen orthophosphate, adjusted to pH 4.0 with of known concentration of norfloxacin RS in the same medium.
orthophosphoric acid, at a flow rate of 0.5 ml per minute for 8 Calculate the total content of C16H18FN3O3 in the medium.
hours. Equilibrate the column with the mobile phase for about D. Not less than 70 per cent of the stated amount of
30 minutes before starting the chromatography. C16H18FN3O3.
Inject the reference solution. The test is not valid unless the
Other Tests. Comply with the tests stated under Tablets.
tailing factor is not more than 2.0. The relative standard
deviation for replicate injections is not more than 2.0 per cent. Assay. Determine by liquid chromatography (2.4.14).
Inject alternately the test solution and the reference solution. Test solution. Weigh and powder 20 tablets. Add 80 ml of the
mobile phase to an accurately weighed quantity of the
Calculate the content of C16H18FN3O3 in the eye drops. powdered tablets containing about 100 mg of Norfloxacin, mix
Storage. Store protected from light. with the aid of ultrasound for 10 minutes, dilute with a 0.1 per

842
IP 2007 NORGESTREL AND ETHINYLOESTRADIOL TABLETS

cent v/v solution of phosphoric acid to 200.0 ml and mix. C. Melting range (2.4.21). 205° to 212°, but the range between
Dilute 10.0 ml of this solution to 25.0 ml with the mobile phase, beginning and end of melting does not exceed 4°.
mix and use the resulting solution after filtration through a
filter with porosity of not more than 0.1 µm. Tests
Reference solution. A 0.02 per cent w/v solution of norfloxacin Specific optical rotation (2.4.22). –0.1° to +0.1°, determined in
RS in the mobile phase. a 5.0 per cent w/v solution in chloroform.
Chromatographic system Related substances. Determine by thin-layer chromatography
– a stainless steel column 30 cm x 3.9 mm, packed with (2.4.17), coating the plate with silica gel G.
octadecylsilane bonded to porous silica (5 µm), Mobile phase. A mixture of 80 volumes of dichloromethane
– mobile phase: 85 volumes of a 0.1 per cent v/v solution and 20 volumes of ethyl acetate.
of phosphoric acid and 15 volumes of acetonitrile,
– temperature. column 40° ± 1°, after preconditioning with Test solution. Dissolve 0.2 g of the substance under
degassed 0.01 M sodium dihydrogen phosphate examination in 10 ml of chloroform.
adjusted to pH 4.0 with phosphoric acid flowing at a Reference solution (a). A 0.01 per cent w/v solution of the
rate of 0.5 ml per minute for 8 hours, substance under examination in chloroform.
– flow rate. 2 ml per minute,
Reference solution (b). A 0.004 per cent w/v solution of the
– spectrophotometer set at 275 nm,
substance under examination in chloroform.
– a 20 µl loop injector.
Apply to the plate 10 µl of each solution. After development,
Inject the test solution and the reference solution. The assay
dry the plate in air and spray with phosphomolybdic acid
is not valid unless the capacity factor is not less than 2, the
solution. Any secondary spot in the chromatogram obtained
column efficiency is not less than 1500 theoretical plates, the
with the test solution is not more intense than the spot in the
tailing factor for the norfloxacin peak is not more than 2.0 and
chromatogram obtained with reference solution (a) and not
the relative standard deviation for replicate injections is not
more than two such spots are more intense than the spot in
more than 2.0 per cent.
the chromatogram obtained with reference solution (b).
Calculate the content of C16H18FN3O3 in the tablets. Sulphated ash. (2.3.18) Not more than 0.3 per cent.
Storage. Store protected from light and moisture. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105° for 5 hours.
Assay. Weigh accurately about 0.1 g, dissolve in sufficient
Norgestrel ethanol (95 per cent) to produce 100.0 ml, dilute stepwise
with ethanol (95 per cent) to obtain a solution containing
H3 C OH 0.001 per cent w/v of Levonorgestrel and measure the
C CH absorbance of the resulting solution at the maximum at about
H H 241 nm, (2.4.7). Calculate the content of C21H28O2 from the
absorbance obtained with a 0.001 per cent w/v solution of
H H norgestrel RS in ethanol (95 per cent).
O Storage. Store protected from moisture.
C21H28O2 Mol. Wt. 312.5
Norgestrel is rac-13-ethyl-17-hydroxy-18,19-dinor-17α-
pregn-4-en-20-yn-3-one. Norgestrel and Ethinyloestradiol
Description. A white or almost white, crystalline powder; Tablets
practically odourless. Norgestrel and Ethinyloestradiol Tablets contain not less than
90.0 per cent and not more than 110.0 per cent of the stated
Identification
amounts of norgestrel, C21H28O 2 and ethinyloestradiol,
A. Determine by infrared absorption spectrophotometry (2.4.6). C20H24O2. The tablets may be film-coated.
Compare the spectrum with that obtained with norgestrel RS.
Identification
B. When examined in the range 220 nm to 360 nm (2.4.7), a
0.001 per cent w/v solution in methanol shows an absorption Determine by thin-layer chromatography (2.4.17), coating the
maximum only at about 240 nm. plate with silica gel G.

843
NORTRIPTYLINE HYDROCHLORIDE IP 2007

Mobile phase. A mixture of 96 volumes of dichloromethane – mobile phase: a mixture of 35 volumes of acetonitrile,
and 4 volumes of ethanol (95 per cent). 15 volumes of methanol and 45 volumes of water,
Test solution. Powder 20 tablets finely, triturate with 20 ml of – flow rate. 1 to1.5 ml per minute,
dichloromethane, allow the solids to sediment and use the – spectrophotometer set at 215 nm,
clear supernatant liquid. – a 20 µl loop injector.

Reference solution. A solution containing 0.06 per cent w/v Inject the reference solution. The resolution between the two
solution of norgestrel RS and 0.006 per cent w/v solution of major peaks is not less 2.5 and the relative standard deviation
ethinyloestradiol RS. for replicate injections is not more than 2.0 per cent.

Apply to the plate 40 µl of each solution. After development, Inject the test solution and the reference solution. The relative
dry the plate in air, spray with ethanolic sulphuric acid retention times are about 0.7 for ethinyloestradiol and about
(80 per cent v/v), heat at 110° for 10 minutes and examine in 1.0 for norgestrel.
ultraviolet light at 365 nm. The principal spots in the Calculate the contents of norgestrel, C 21 H 28 O 2 , and
chromatogram obtained with the test solution correspond to ethinyloestradiol, C20H24O2 in the tablets.
the spots for norgestrel (red fluorescence) and
Storage. Store protected from light.
ethinyloestradiol (orange-yellow fluorescence) in the
chromatogram obtained with the reference solution.

Tests
Nortriptyline Hydrochloride
Uniformity of content. Comply with the test stated under
Tablets.
Carry out the procedure described under Assay but using the
following test solution. , HCl
Test solution. Add 2.0 ml of methanol (70 per cent) and 2.0 ml CH3
of a 0.00002 per cent w/v solution of diphenyl in methanol N
H
(70 per cent) (internal standard solution) to one tablet, shake
for 20 minutes, centrifuge, filter the supernatant liquid through C19H21N,HCl Mol. Wt. 299.8
a membrane filter with a pore size of not more than 0.2 mm and
Nortriptyline Hydrochloride is 3-(10,11-dihydro-5H-dibenzo
use the filtrate.
[a,d]cyclohept-5-ylidene)propyl(methyl)amine hydrochloride.
Calculate the contents of norgestrel C 21 H 28 O 2 , and
Nortriptyline Hydrochloride contains not less than 98.0 per
ethinyloestradiol, C20H24O2, in the tablet.
cent and not more than 101.5 per cent of C19H21N,HCl, calculated
Other Tests. Comply with the tests stated under Tablets. on the dried basis.
Assay. Determine by liquid chromatography (2.4.14) using the Description. A white to off-white powder; odour slight and
chromatographic system described under Uniformity of characteristic.
content.
Test solution. Weigh and powder 20 tablets. To a quantity of Identification
the powder equivalent to one tablet add 2.0 ml of methanol Test A may be omitted if tests B, C and D are carried out. Tests
(70 per cent) and 2.0 ml of a 0.00002 per cent w/v solution of B and C may be omitted if tests A and, D are carried out.
diphenyl in methanol (70 per cent) (internal standard
solution), shake for 20 minutes, centrifuge, filter the A. Dissolve 0.1 g in 10 ml of water, make alkaline with 1 M
supernatant liquid through a membrane filter with a pore size sodium hydroxide, extract with 5 ml of chloroform and
of not more than 0.2 mm and use the filtrate. evaporate to dryness using a current of nitrogen.
Reference solution. A solution in methanol (70 per cent ) Determine by infrared absorption spectrophotometry (2.4.6).
containing 0.15 mg per ml of norgestrel RS and 0.015 mg per Compare the spectrum with that obtained with nortriptyline
ml of ethinyloestradiol RS. Take 2.0 ml of this solution and hyrdochloride RS treated in the same manner or with the
add 2.0 ml of a 0.00002 per cent w/v solution of diphenyl in reference spectrum of nortriptyline.
methanol (70 per cent) and use the resulting solution. B. When examined in the range 230 nm to 360 nm (2.4.7), a
Chromatographic system 0.001 per cent w/v solution in methanol shows an absorption
– a stainless steel column 15 cm x 4.6 mm, packed with maximum only at about 239 nm; absorbance at about 239 nm,
octadecylsilane bonded to porous silica (5 to 7 µm), about 0.48.

844
IP 2007 NORTRIPTYLINE TABLETS

C. To about 50 mg dissolved in 3 ml of warm water, add 1 drop Identification


of a 2.5 per cent w/v solution of quinhydrone in methanol; a
red colour is produced after a few minutes (distinction from A. Shake a quantity of the powdered tablets containing about
amitriptyline). 5 mg of nortriptyline with 20 ml of methanol and filter. To 1 ml
of the filtrate add 1 ml of a 2.5 per cent w/v solution of sodium
D. Gives the reactions of chlorides (2.3.1). bicarbonate, 1 ml of a 2 per cent w/v solution of sodium
periodate and 1 ml of a 0.3 per cent w/v solution of potassium
Tests permanganate. Allow to stand for 15 minutes, acidify with
Related substances. Determine by thin-layer chromatography 1 M sulphuric acid and extract with 10 ml of 2,2,4-
(2.4.17), coating the plate with silica gel G. trimethylpentane.

Mobile phase. A mixture of 85 volumes of cyclohexane, When examined in the range 230 nm to 360 nm (2.4.7), the
15 volumes of ethyl acetate and 3 volumes of diethylamine. resulting trimethylpentane solution shows an absorption
maximum only at about 265 nm.
Test solution. Dissolve 0.2 g of the substance under
B. Triturate a quantity of the powdered tablets containing
examination in 10 ml of ethanol (95 per cent) prepared in
0.1 g of nortriptyline with 10 ml of chloroform, filter and
subdued light.
evaporate the filtrate to a low volume. Add ether until a
Reference solution. A 0.001 per cent w/v solution of turbidity is produced and allow to stand. Dissolve 50 mg of
dibenzosuberone RS in ethanol (95 per cent) prepared in the precipitate in 3 ml of warm water, cool and add 1 drop of a
subdued light. 2.5 per cent w/v solution of quinhydrone in methanol; a red
Apply to the plate 5 µl of each solution. Allow the mobile colour is produced after a few minutes (distinction from
phase to rise 14 cm in an unsaturated tank protected from amitriptyline).
light. Dry the plate in air, spray with a freshly prepared solution
of sulphuric acid containing 4 per cent v/v of formaldehyde
Tests
solution and examine immediately in ultraviolet light at Related substances. Determine by thin-layer chromatography
365 nm. Any secondary spot in the chromatogram obtained (2.4.17), coating the plate with silica gel G.
with the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution. Mobile phase. A mixture of 85 volumes of cyclohexane,
15 volumes of ethyl acetate and 3 volumes of diethylamine.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
Test solution. Extract a quantity of the powdered tablets
heavy metals, Method B (10 ppm).
containing 20 mg of nortriptyline with 5 ml of a mixture of
Sulphated ash (2.3.18). Not more than 0.1 per cent. 9 volumes of ethanol (95 per cent) and 1 volume of 2 M
Loss on drying (2.4.19). Not more than 0.5 per cent, determined hydrochloric acid, centrifuge and use the supernatant liquid.
on 1.0 g by drying in an oven at 105° for 3 hours. Reference solution. A 0.001 per cent w/v solution of
Assay. Weigh accurately about 0.25 g and dissolve in 25 ml of dibenzosuberone RS in ethanol (95 per cent) prepared in
anhydrous glacial acetic acid, warm slightly, if necessary, to subdued light.
effect solution. Cool, add 5 ml of mercuric acetate solution. Apply to the plate 5 µl of each solution. Allow the mobile
Titrate with 0.1 M perchloric acid, determining the end-point phase to rise 14 cm in an unsaturated tank protected from
potentiometrically (2.4.25). Carry out a blank titration. light. Dry the plate in air, spray with a freshly prepared solution
of sulphuric acid containing 4 per cent v/v of formaldehyde
1 ml of 0.1 M perchloric acid is equivalent to 0.02998 g of
solution and examine immediately in ultraviolet light at
C19H21N,HCl.
365 nm. Any secondary spot in the chromatogram obtained
Storage. Store protected from light and moisture. with the test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
Uniformity of content (For tablets containing 10 mg or less).
Comply with the test stated under Tablets.
Nortriptyline Tablets
Determine by liquid chromatography (2.4.14)
Nortriptyline HydrochlorideTablets
Test solution. Powder one tablet, add 2.5 ml of water, shake
Nortriptyline Tablets contain less than 90.0 per cent and not vigorously to completely disperse the tablet, add 5 ml of
more than 110.0 per cent of the stated amount of nortriptyline, methanol and shake for 30 minutes. Add sufficient water to
C19H21N. The tablets are coated. produce 10 ml, centrifuge and use the clear supernatant liquid.

845
NOSCAPINE IP 2007

Reference solution. A 0.01 per cent w/v solution of Noscapine contains not less than 98.5 per cent and not more
nortriptyline hydrochloride RS in methanol (50 per cent). than 100.5 per cent of C22H23NO7, calculated on the dried basis.
Follow the procedure given in the Assay. Calculate the content Description. Colourless crystals or a white crystalline powder.
of C20H23N in the tablet.
Identification
Other Tests. Comply with the tests stated under Tablets.
Assay. Determine by liquid chromatography (2.4.14). Test A may be omitted if tests B, C and D are carried out. Tests
B, C and D may be omitted if test A is carried out.
Test solution. Shake vigorously 20 tablets with 50 ml of water
until the tablets disintegrate completely, add 100.0 ml of A. Determine by infrared absorption spectrophotometry (2.4.6).
methanol and shake for 30 minutes. Add sufficient water to Compare the spectrum with that obtained with noscapine RS
produce 200.0 ml, filter and dilute a volume of the filtrate or with the reference spectrum of noscapine.
containing about 25 mg of nortriptyline to 100.0 ml with B. When examined in the range 230 nm to 360 nm (2.4.7), a
methanol (50 per cent). 0.005 per cent w/v solution in methanol shows absorption
Reference solution. A 0.025 per cent w/v solution of maxima at about 291 nm and 310 nm; ratio of absorbance at the
nortriptyline hydrochloride RS in methanol (50 per cent). maximum at about 310 nm to that at the maximum at about
291 nm, 1.2 to 1.3.
Chromatographic system
– a stainless steel column 20 cm x 4.6 mm, packed with C. To 0.1 g in a porcelain dish add a few drops of sulphuric
octadecylsilane bonded to porous silica (10 µm), acid and stir; a greenish-yellow solution is formed which on
– mobile phase: a 0.56 per cent w/v solution of sodium warming becomes red and finally violet.
hexanesulphonate in a mixture of equal volumes of D. Dissolve 50 mg in 5 ml of 5 M hydrochloric acid, add 10 ml
water and acetonitrile adjusted to pH 4.5 with glacial of a mixture of equal volumes of ethanol (95 per cent) and a
acetic acid, saturated solution of sodium acetate, mix and allow to stand
– flow rate. 2 ml per minute, for about 3 minutes; shining crystals separate.
– spectrophotometer set at 239 nm,
– a 20 µl loop injector. Tests
Inject the test solution and the reference solution. Appearance of solution. A 2.0 per cent w/v solution in acetone
Calculate the content of C19H21N in the tablets. examined immediately after preparation is clear (2.4.1), and not
more intensely coloured than reference solution YS6 (2.4.1).
Storage. Store protected from light and moisture.
Specific optical rotation (2.4.22). +42.0° to +48.0°, determined
at 20° in a solution prepared by dissolving 0.5 g in sufficient
0.1 M hydrochloric acid to produce 25.0 ml.
Noscapine Related substances. Determine by thin-layer chromatography
Narcotine (2.4.17), coating the plate with silica gel G.
Mobile phase. A mixture of 60 volumes of acetone, 60 volumes
O of toluene, 9 volumes of ethanol (95 per cent) and 3 volumes
of strong ammonia solution.
O N
CH3 Test solution. Dissolve 0.25 g of the substance under
H
OCH3 examination in 10 ml of acetone.
H Reference solution. A 0.0125 per cent w/v solution of the
substance under examination in acetone.
O
Apply to the plate 10 µl of each solution. After development,
H3CO dry the plate in air, spray with dilute potassium
OCH3 O iodobismuthate solution. Any secondary spot in the
chromatogram obtained with the test solution is not more
C22H23NO7 Mol. Wt. 413.4 intense than the spot in the chromatogram obtained with the
reference solution.
Noscapine is (3S)-6,7-dimethoxy-3-[(5R)-5,6,7,8-tetrahydro-
4-methoxy-6-methyl-1,3-dioxolo[4,5-g]isoquinolin-5- Morphine. Dissolve 0.1 g in 10 ml of 0.1 M hydrochloric acid.
yl]phthalide, an alkaloid obtained from opium. To 1 ml of the resulting solution add a mixture of 1 ml of

846
IP 2007 NOVOBIOCIN SODIUM

potassium ferricyanide solution, 0.05 ml of ferric chloride Assay. Weigh accurately a quantity containing 60 mg of
test solution and 4 ml of water; no blue or dark green colour Noscapine, add 20 ml of water, 2 g of sodium chloride and
develops within 1 minute. 2 ml of 5 M sodium hydroxide and extract with successive
Sulphated ash (2.3.18). Not more than 0.1 per cent. quantities of 50, 50, 25 and 25 ml of ether. Combine the extracts,
wash with three quantities, each of 5 ml, of water and evaporate
Loss on drying (2.4.19). Not more than 1.0 per cent, determined to dryness. To the residue add 50.0 ml of 0.1 M hydrochloric
on 1.0 g by drying in an oven at 105°. acid, warm on a water-bath to dissolve and to remove any
Assay. Weigh accurately about 0.35 g, dissolve in 40 ml of traces of ether and dilute to 100.0 ml with water. Dilute 3.0 ml
anhydrous glacial acetic acid, warming gently. Titrate with to 50.0 ml with water and measure the absorbance of the
0.1 M perchloric acid, determining the end-point resulting solution at the maximum at about 310 nm (2.4.7).
potentiometrically (2.4.25). Carry out a blank titration. Calculate the content of C22H23NO7 taking 90.7 as the specific
absorbance at 310 nm.
1 ml of 0.1 M perchloric acid is equivalent to 0.04134 g of
C22H23NO7. Determine the weight per ml of the linctus (2.4.29), and calculate
the content of C22H23NO7, weight in volume.
Storage. Store protected from light and moisture.
Storage. Store protected from light and moisture.

Noscapine Linctus Novobiocin Sodium


Narcotine Linctus CH3
CH3
Noscapine Linctus is a solution of Noscapine in a suitable H3CO O O O
O O CH3
flavoured vehicle. It may contain up to 1 per cent w/v solution CH3
of Citric Acid. N CH3
H3 C O OH
Noscapine Linctus contains not less than 90.0 per cent and ONa H
OH
not more than 110.0 per cent of the stated amount of O
noscapine, C22H23NO7. C31H35N2NaO11 Mol. Wt. 634.6

Identification Novobiocin Sodium is the monosodium salt of novobiocin,


N-[7-{3-O-(aminocarbonyl)-6-deoxy-5-C-methyl-4-O-methyl-
To a quantity containing 60 mg of Noscapine add 20 ml of β-L-lyxo-hexopyranosyl}-4-hydroxy-8-methyl-2-oxo-2H-1-
water, 2 g of sodium chloride and 2 ml of 5 M sodium benzopyran-3-yl]-4-hydroxy-3-(3-methyl-2-
hydroxide. Extract with successive quantities of 50, 50, 25 and butenyl)benzamide, an antimicrobial substance produced by
25 ml of ether. Combine the extracts, wash with three quantities, the growth of certain strains of Streptomyces niveus or
each of 5 ml, of water and evaporate to dryness. Dissolve the related organisms or by other means.
residue in 20 ml of chloroform. Wash with three quantities,
Novobiocin Sodium contains the equivalent of not less than
each of 20 ml, of water, dry the chloroform layer with anhydrous
850 µg of novobiocin per mg, calculated on the dried basis.
sodium sulphate, filter and evaporate the solvent. If necessary,
induce crystallisation by scratching with a glass rod. The Description. A white or yellowish white, crystalline powder.
crystals comply with the following tests.
Identification
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with noscapine RS A. Determine by thin-layer chromatography (2.4.17), coating
or with the reference spectrum of noscapine. the plate with silica gel G.
B. When examined in the range 230 nm to 360 nm (2.4.7), a Mobile phase. A mixture of 75 volumes of chloroform,
0.005 per cent w/v solution in methanol shows absorption 25 volumes of methanol and 1 volume of strong ammonia
maxima at about 291 nm and 310 nm; ratio of absorbance at the solution.
maximum at about 310 nm to that at the maximum at about Test solution. Dissolve a quantity of the substance under
291 nm, 1.2 to 1.3. examination in methanol so as to obtain a solution containing
0.1 per cent w/v solution of novobiocin.
Tests
Reference solution. A 0.1 per cent w/v solution of novobiocin
Other tests. Complies with the tests stated under Oral Liquids. RS in methanol.

847
NYSTATIN IP 2007

Apply to the plate 1 µl of each solution. After development, Identification


dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with the test A. When examined in the range 220 nm to 360 nm (2.4.7), the
solution corresponds to that in the chromatogram obtained final solution obtained in the test for Light absorption shows
with the reference solution. absorption maxima at about 230 nm, 291 nm, 305 nm and
319 nm. The ratios of the absorbances at the maxima at about
B. When examined in the range 230 nm to 360 nm (2.4.7), a 291 nm and about 319 nm to the absorbance at the maximum at
0.001 per cent w/v solution in a 0.4 per cent w/v solution of about 305 nm are 0.61 to 0.73 and 0.83 to 0.96, respectively.
potassium hydroxide shows an absorption maximum only at Use as the blank a solution prepared in the same manner
about 307 nm. without the substance under examination.
C. The residue obtained by igniting it gives the tests for sodium
B. Shake 30 mg with 5 ml of water for 2 minutes, add 2 ml of
salts (2.3.1).
sodium molybdotungstophosphate solution, and allow to
Tests stand for 1 hour; the green colour produced is darker than
that produced by repeating the test without the substance
pH (2.4.24). 6.6 to 8.5, determined in a 2.5 per cent w/v solution. under examination.
Specific optical rotation (2.4.22). –50.0° to –58.0°, determined C. Shake 30 mg with 5 ml of water for 2 minutes, add 2 ml of a
in a 5.0 per cent w/v solution in methanol containing 1 per solution prepared by dissolving 0.1 g of pyrogallol in 100 ml
cent v/v of hydrochloric acid. of decolorised magenta solution, heat on a water-bath until a
Loss on drying (2.4.19). Not more than 6 per cent, determined dark pink colour is produced, cool and allow to stand for
on 0.2 g by drying in an oven at 60° over phosphorus pentoxide 1 hour; the pink colour is retained.
at a pressure not exceeding 0.7 kPa for 3 hours. D. To 2 mg add 0.1 ml of hydrochloric acid; a brown colour is
Assay. Determine by the microbiological assay of antibiotics, produced.
Method A (2.2.10), and express the results in µg of novobiocin E. To 2 mg add 0.1 ml of sulphuric acid; a brown colour is
per mg. produced which becomes violet on standing.
Novobiocin Sodium intended for use in the manufacture of
Parenteral Preparations complies with the following Tests
additional requirements.
pH (2.4.24). 6.5 to 8.0, determined in a 3.0 per cent w/v
Bacterial endotoxins (2.2.3). Not more than 0.7 Endotoxin units suspension in water.
per mg.
Light absorption (2.4.7). Dissolve 0.1 g in a mixture of 5.0 ml of
Sterility. Complies with the test for sterility (2.2.11). glacial acetic acid and 50 ml of methanol, add sufficient
Storage. Store protected from light and moisture at a methanol to produce 100.0 ml and dilute 1.0 ml of the resulting
temperature not exceeding 30°. If it is intended for use in the solution to 100.0 ml with methanol. Absorbance of the resulting
manufacture of parenteral preparations, the container should solution, measured within 30 minutes of preparation, at the
be sterile and sealed so as to exclude micro-organisms. maximum at about 305 nm, not less than 0.60. Use as the blank
a solution prepared in the same manner without the substance
Labelling. The label states whether or not the contents are under examination.
intended for use in the manufacture of parenteral preparations.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Sulphated ash (2.3.18). Not more than 3.5 per cent.
Nystatin
Loss on drying (2.4.19). Not more than 5.0 per cent, determined
Nystatin is an antifungal substance produced by the growth on 1.0 g by drying in an oven at 60° at a pressure not exceeding
of certain strains of Streptomyces noursei or by any other 0.1 kPa for 3 hours.
means. It consists mainly of polyenes, the principal component
being nystatin A1. Assay. Protect the solution from light throughout the assay.
Nystatin has a potency of not less than 4400 Units per mg, Weigh accurately about 75 mg and dissolve in sufficient
calculated on the dried basis. dimethylformamide to produce 50.0 ml; dilute 10.0 ml of the
resulting solution to 200.0 ml with buffer solution No 4 (2.2.10).
Description. A yellow to slightly brown powder; odour,
characteristic; hygroscopic. Determine by the microbiological assay of antibiotics (2.2.10).

848
IP 2007 NYSTATIN TABLETS

Nystatin intended for oral administration complies with the Nystatin Pessaries
following additional requirement.
Nystatin Vaginal Tablets
Abnormal toxicity (2.2.1). Complies with the test for abnormal
toxicity, using a quantity containing not less than 600 Units Nystatin Pessaries contain not less than 90.0 per cent and not
suspended in not more than 0.5 ml of a 0.5 per cent w/v solution more than 130.0 per cent of the stated number of Units of
of acacia and injecting the suspension intraperitoneally. nystatin.

Storage. Store protected from light and moisture. Identification


Labelling. The label states the strength in terms of the number Extract a quantity of the powdered pessaries containing
of Units of Nystatin per mg. 300,000 Units with a mixture of 50 ml of methanol and 5 ml of
glacial acetic acid, add sufficient methanol to produce
100 ml and filter. Dilute 1 ml of the filtrate to 100 ml with
methanol. The resulting solution complies with the follwing
Nystatin Ointment test.
Disperse a quantity containing 25,000 Units in 10 ml of
Nystatin Ointment is a dispersion of Nystatin in microfine chloroform, add 40 ml of methanol and shake. Filter and dilute
powder in a suitable ointment basis. 1 ml of the filtrate to 25 ml with methanol.
Nystatin Ointment contains not less than 90.0 per cent and When examined in the range 230 nm to 360 nm (2.4.7), the
not more than 130.0 per cent of the stated number of Units of resulting solution shows absorption maxima at about 291 nm,
nystatin. 305 nm and 319 nm. The ratios of the absorbances at the maxima
at about 291 nm and about 319 nm to the absorbance at the
Identification maximum at about 305 nm are 0.61 to 0.73 and 0.83 to 0.96,
Disperse a quantity containing 25,000 Units in 10 ml of respectively. Use as the blank a solution prepared exactly in
chloroform, add 40 ml of methanol and shake. Filter and dilute the same manner without the substance under examination.
1 ml of the filtrate to 25 ml with methanol.
Tests
When examined in the range 230 nm to 360 nm (2.4.7), the
resulting solution shows absorption maxima at about 291 nm, Other Tests. Comply with the tests stated under Pessaries.
305 nm and 319 nm. The ratios of the absorbances at the maxima Loss on drying (2.4.19). Not more than 5 per cent, determined
at about 291 nm and about 319 nm to the absorbance at the on 1.0 g of the powdered pessaries by drying in an oven at 60°
maximum at about 305 nm are 0.61 to 0.73 and 0.83 to 0.96, at a pressure not exceeding 0.7 kPa for 3 hours.
respectively. Use as the blank a solution prepared exactly in
the same manner without the substance under examination. Assay. Protect the solution from light throughout the assay.
Weigh and powder 20 pessaries. Weigh accurately a quantity
Tests of the powder containing 200,000 Units and shake with 50.0 ml
Other tests. Complies with the tests stated under Ointments. of dimethylformamide for 1 hour. Centrifuge, dilute 10.0 ml of
the clear, supernatant liquid to 200.0 ml with buffer solution
Assay. Protect the solution from light throughout the assay. No 4 (2.2.10).
Weigh accurately a quantity containing 400,000 Units, disperse Determine by the microbiological assay of antibiotics (2.2.10).
in 20 ml of ether in a stoppered flask, add 70 ml of
Storage. Store protected from light and moisture.
dimethylformamide, shake for a few minutes, add 10 ml of
water, shake vigorously for a few minutes and add sufficient Labelling. The label states the strength in terms of the number
dimethylformamide to produce 100.0 ml. Mix well, filter and of Units of Nystatin.
dilute 10.0 ml of the filtrate to 100.0 ml with buffer solution No
4 (2.2.10).
Determine by the microbiological assay of antibiotics (2.2.10).
Nystatin Tablets
Storage. Store protected from moisture.
Nystatin Tablets contain not less than 90.0 per cent and not
Labelling. The label states the strength in terms of the number more than 130.0 per cent of the stated number of Units of
of Units of Nystatin per g. nystatin. The tablets are coated.

849
NYSTATIN TABLETS IP 2007

Identification tablets fail to disintegrate, wash them rapidly by immersion in


water and continue the test using phosphate buffer pH 6.8;
Extract a quantity of the powdered tablets containing the tablets then disintegrate within a further 30 minutes.
300,000 Units with a mixture of 50 ml of methanol and 5 ml of
glacial acetic acid, add sufficient methanol to produce Loss on drying (2.4.19). Not more than 5.0 per cent, determined
100 ml and filter. Dilute 1 ml of the filtrate to 100 ml with on 1.0 g of the powdered tablets by drying in an oven at 60° at
methanol. The resulting solution complies with the follwing a pressure not exceeding 0.7 kPa for 3 hours.
test. Other Tests. Comply with the tests stated under Tablets.
When examined in the range 230 nm to 360 nm (2.4.7), the Assay. Weigh and powder 20 tablets. Weigh accurately a
solution shows absorption maxima at about 291 nm, 305 nm quantity of the powder containing 200,000 Units and shake
and 319 nm. The ratios of the absorbances at the maxima at with 50.0 ml of dimethylformamide for 1 hour. Centrifuge, dilute
about 291 nm and about 319 nm to the absorbance at the 10.0 ml of the clear, supernatant liquid to 200.0 ml with buffer
maximum at about 305 nm are 0.61 to 0.73 and 0.83 to 0.96, solution No 4 (2.2.10).
respectively. Use as the blank a solution prepared exactly in
the same manner without the substance under examination. Determine by the microbiological assay of antibiotics (2.2.10).
Storage. Store protected from moisture.
Tests
Labelling. The label states the strength in terms of the number
Disintegration (2.5.1). 30 minutes, but using a 0.6 per cent of Units of Nystatin.
v/v solution of hydrochloric acid in place of water. If the

850
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

O
Oestradiol Benzoate ....
Oestradiol Injection ....
Ofloxacin ....
Ofloxacin Infusion ....
Ofloxacin Opthalmic Solution ....
Ofloxacin Tablets ....
Olanzapine ....
Olanzapine Tablets ....
Oleic Acid ....
Omeprazole ....
Omeprazole Capsules ....
Oral Rehydration Salts ....
Ormeloxifen Hydrochloride ....
Ormeloxifen Hydrochloride Tablets ....
Orphenadrine Citrate ....
Orphenadrine Hydrochloride ....
Orphenadrine Tablets ....
Oseltamivir Phosphate ....
Oseltamivir Capsules ....
Oseltamivir Oral Suspension ....
Oxazepam ....
Oxazepam Tablets ....
Oxprenolol Hydrochloride ....
Oxprenolol Tablets ....
Oxygen ....
Oxygen 93 Per Cent ....
Oxyphenbutazone ....
Oxyphenbutazone Tablets ....
Oxytetracycline Dihydrate ....
Oxytetracycline Injection ....

851
MONOGRAPHS INDIAN PHARMACOPOEIA 2007 2007

Oxytetracycline Hydrochloride ....


Oxytetracycline Capsules ....
Oxytetracycline Eye Ointment ....
Oxytetracycline Hydrochloride Injection ....
Oxytocin ....
Oxytocin Injection ....
Oxytocin Nasal Solution ....

852
IP 2007 OESTRADIOL INJECTION

Oestradiol Benzoate Test solution (b). Dissolve 0.1 g of the substance under
examination in 100 ml of the same solvent mixture.

OH Reference solution (a). A 0.02 per cent w/v solution of the


H3C
substance under examination in the same solvent mixture.
H Reference solution (b). A 0.1 per cent w/v solution of
O oestradiol benzoate RS in the same solvent mixture.
H H
Apply to the plate 5 µl of each solution. After development,
O
dry the plate in air until the odour of the solvent is no longer
detectable, heat at 110º for 10 minutes, spray the plate while
hot with ethanolic sulphuric acid (20 per cent), heat again at
C25H28O3 Mol. Wt. 376.5 110º for 10 minutes and examine in ultraviolet light at 365 nm.
Any secondary spot in the chromatogram obtained with test
Oestradiol Benzoate is 17β-hydroxyestra-1,3,5(10)-trien-3-yl
solution (a) is not more intense than the spot in the
benzoate.
chromatogram obtained with reference solution (a).
Oestradiol Benzoate contains not less than 97.0 per cent and
Sulphated ash (2.3.18). Not more than 0.2 per cent, determined
not more than 103.0 per cent of C25H28O3, calculated on the
on 0.5 g.
dried basis.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Description. Colourless crystals or a white, crystalline powder;
on 0.5 g by drying in an oven at 105º for 3 hours.
odourless.
Assay. Weigh accurately about 25 mg, dissolve in sufficient
Identification ethanol (95 per cent) to produce 250.0 ml. Dilute 10.0 ml to
100.0 ml with ethanol (95 per cent) and measure the
Test A may be omitted if tests B and C are carried out. Test C
absorbance of the resulting solution at the maximum at about
may be omitted if tests A and B are carried out.
231 nm (2.4.7). Calculate the content of C25H28O3 taking 500 as
A. Determine by infrared absorption spectrophotometry (2.4.6). the specific absorbance at 231 nm.
Compare the spectrum with that obtained with oestradiol Storage. Store protected from light and moisture.
benzoate RS or with the reference spectrum of oestradiol
benzoate.
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Oestradiol Injection
that in the chromatogram obtained with reference solution (b)
when examined in daylight and in ultraviolet light at 365 nm. Oestradiol Benzoate Injection
C. To about 1 mg add 0.5 ml of a 5 per cent w/v solution of Oestradiol Injection is a sterile solution of Oestradiol Benzoate
ammonium molybdate in sulphuric acid; a yellowish green in Ethyl Oleate or other suitable ester, in a suitable fixed oil or
colour develops which exhibits an intense green fluorescence in any mixture of these. It may contain suitable alcohols.
when examined in ultraviolet light at 365 nm. Add 1 ml of Oestradiol Benzoate Injection contains not less than 90.0 per
sulphuric acid and 9 ml of water; the solution becomes pink cent and not more than 110.0 per cent of the stated amount of
with a yellowish fluorescence. oestradiol benzoate, C25H28O3.
Tests Identification
Specific optical rotation (2.4.22). +57.0º to +63.0º, determined Determine by thin-layer chromatography (2.4.17), coating the
in a 1.0 per cent w/v solution in dioxan. plate with silica gel G.
Related substances. Determine by thin-layer chromatography Mobile phase. A mixture of 80 volumes of toluene and
(2.4.17), coating the plate with silica gel G. 20 volumes of ethyl acetate.
Mobile phase. A mixture of 90 volumes of toluene and Test solution. Add 10 ml of 2,2,4-trimethylpentane to a volume
10 volumes of ethanol (95 per cent). of the injection containing 2 mg of Oestradiol Benzoate and
Test solution (a). Dissolve 0.2 g of the substance under extract with three quantities, each of 10 ml, of ethanol (70 per
examination in 10 ml of a mixture of 90 volumes of chloroform cent). Wash the combined extracts with 15 ml of 2,2,4-
and 10 volumes of methanol. trimethylpentane, evaporate the ethanolic extract to dryness

853
OFLOXACIN IP 2007

using a rotary evaporator and dissolve the residue in 2 ml of Ofloxacin


chloroform.
Reference solution. A 0.1 per cent w/v solution of oestradiol H 3C CH3
N O
benzoate RS in chloroform.
N N
Apply to the plate 5 µl of each solution. After development,
dry the plate in air, spray with ethanolic sulphuric acid
(20 per cent), heat at 105º for 10 minutes and examine in F COOH
ultraviolet light at 365 nm. The principal spot in the O
chromatogram obtained with the test solution corresponds to
C18H20FN3O4 Mol. Wt. 361.4
that in the chromatogram obtained with the reference solution.
Ofloxacin is (RS)-9-fluoro-3-methyl-10-(4-methylpiperazin-1-
Tests yl)-7-oxo-2,3-dihydro-7H-pyrido[1,2,3,-de]-1,4-benzoazeine-
6-carboxylic acid
Other Tests. Complies with the tests stated under Parenteral
Preparations (Injections). Ofloxacin contains not less than 98.5 per cent and not more
than of 101.5 per cent of C18H20FN3O4, calculated on the dried
Assay. Determine by liquid chromatography (2.4.14).
basis.
Test solution (a). Dilute an accurately measured quantity of
Description. A pale yellow or bright yellow, crystalline powder.
the injection containing about 1 mg of Oestradiol Benzoate to
10.0 ml with a mixture of 90 volumes of cyclohexane and Identification
10 volumes of dioxan.
Determine by infrared absorption spectrophotometry (2.4.6).
Test solution (b). Add 1 ml of a solution prepared by dissolving
Compare the spectrum with that obtained with ofloxacin RS.
15 mg of 4-hydroxybenzaldehyde (internal standard) in
10.0 ml of dioxan, adding sufficient cyclohexane to produce Tests
100.0 ml (solution A), to an accurately measured quantity of
the injection containing about 1 mg of Oestradiol Benzoate Light absorption. Absorbance at 440 nm (2.4.7), of 0.5 per cent
and dilute to 10.0 ml with sufficient of a mixture of 90 volumes w/v solution in 0.1 M hydrochloric acid is not more than 0.25.
of cyclohexane and 10 volumes of dioxan. Related substances. Determine by liquid chromatography
Reference solution. Add 10 ml of solution A to 10 mg of (2.4.14).
oestradiol benzoate RS, accurately weighed, and dilute to Test solution. Dissolve 10 mg of the substance under
100.0 ml with the same solvent mixture. examination in 10 ml of methanol.
Chromatographic system Reference solution (a). A 0.1 per cent w/v solution of
– a stainless steel column 30 cm x 4 mm, packed with porous ofloxacin RS in methanol.
silica particles (10 µm), Reference solution (b). Dilute 1 ml of reference solution (a) to
– mobile phase: 90 volumes of cyclohexane and 100 ml with methanol.
10 volumes of dioxan,
– flow rate. 2 ml per minute, Chromatographic system
– spectrophotometer set at 254 nm, – a stainless steel column 10 cm x 4.6 mm packed with
– a 20 µl loop injector. octadecylsilyl silica gel for chromatography (5 µm),
– mobile phase: a mixture of 10 volumes of acetonitrile
Inject test solutions (a), (b) and the reference solution. The and 90 volumes of phosphate buffer pH 2.4 prepared by
assay is not valid unless the resolution between the peaks dissolving 27.2 g of monobasic potassium phosphate
due to benzyl alcohol (if present) and oestradiol benzoate and in 1000 ml of water, adjust the pH to 2.4 with
between the peaks due to oestradiol benzoate and the internal orthophosphoric acid,
standard is more than 1.5. – flow rate. 2 ml per minute,
Calculate the content of C25H28O3 in the injection. – spectrophotometer set at 294 nm,
– a 10 µl loop injector.
Storage. Store protected from light.
Inject reference solution (b). The test is not valid unless the
Labelling. The label states (1) the nature and composition of
column efficiency in not less than 1400 theoretical plates.
the solvent; (2) that it is meant for intramuscular injection
only; (3) that any solid matter that may have separated on Inject the test solution and reference solution (b). Run the
standing should be redissolved by warming before use. chromatogram three times of the principal peak. In the

854
IP 2007 OFLOXACIN OPHTHALMIC SOLUTION

chromatogram obtained with the test solution, the area of any – mobile phase: a mixture of 80 volumes of buffer solution
secondary peak is not more than 0.5 times the area of the peak prepared by dissolving 6.8 g of potassium dihydrogen
in the chromatogram obtained with reference solution (b) (0.5 orthophosphate and 0.47 g sodium 1-hexane
per cent) and the sum of areas of all the secondary peaks is sulphonate in 1000 ml of water, add 1 ml of triethylamine
not more than the area of the peak in the chromatogram and adjust the pH to 3.0 with orthophosphoric acid
obtained with the reference solution (b) (1.0 per cent). and 20 volumes of acetonitrile,
Heavy Metals (2.3.13). 2.0 g complies with the limit test for – flow rate. 1 ml per minute,
heavy metals, Method C (10 ppm). – spectrophotometer set at 294 nm,
– a 20 µl loop injector.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Inject the reference solution. The test is not valid unless the
Loss on drying (2.4.19). Not more than 0.2 per cent, determined tailing factor is not more than 2.0 and the relative standard
on 1.0 g by drying in an oven at 105° for 4 hours. deviation for replicate injections is not more than 2.0 per cent.
Assay. Weigh accurately about 0.3 g, dissolve in 100 ml Inject the test solution and the reference solution.
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
Calculate the content of C18H20FN3O4 in infusion.
acid, determining the end-point potentiometrically (2.4.25).
Carry out a blank titration.
1 ml 0.1 M perchloric acid is equivalent of 0.03614 g of
C18H20FN3O4.
Ofloxacin Ophthalmic Solution
Ofloxacin Ophthalmic Solution is a sterile aqueous solution of
Storage. Store protected from light and moisture.
Ofloxacin.
Ofloxacin Ophthalmic Solution contain not less than 90.0 per
cent and more than 110.0 per cent of the stated amount of
Ofloxacin Infusion ofloxacin, C18H20FN3O4.
Ofloxacin Infusion contain Ofloxacin in water for injection. Identification
Ofloxacin Infusion contain not less than 90.0 per cent and not In the Assay, the principal peak in the chromatogram obtained
more than 120.0 per cent of the stated amount of ofloxacin, with the test solution corresponds to the peak in the
C18H20FN3O4. chromatogram obtained with the reference solution.
Identification Tests
In the Assay, the principal peak in the chromatogram obtained pH (2.4.24). 6.0 to 7.2.
with the test solution corresponds to the peak in the Sterility (2.2.11). Comply with the test for sterility.
chromatogram obtained with the reference solution.
Assay. Determine by liquid chromatography (2.4.14).
Tests Solvent mixture. Phosphate buffer pH 7.25 prepared by
dissolving 3.56 g of disodium hydrogen phosphate in 1000 ml
pH (2.4.24). 3.8 to 5.8
water, adjusted pH to 7.25 using orthophosphoric acid or
Other tests. Complies with the tests stated under Parenteral sodium hydroxide solution.
Preparation (Infusions).
Test solution. Measure accurately a volume of Opthalmic
Assay. Determine by liquid chromatography (2.4.14). Solution containing 30 mg of Ofloxacin in 100.0 ml of solvent
NOTE —Protect the solutions from the light. mixture. Dilute 1.0 ml of the solution to 10.0 with solvent
mixture.
Test solution. Measure accurately a volume containing 50 mg
Reference solution. A 0.003 per cent w/v solution of ofloxacin
of Ofloxacin in 100.0 ml with mobile phase. Dilute 5.0 ml of the
RS in solvent mixture.
solution to 50.0 ml with mobile phase.
Chromatographic system
Reference solution. A 0.005 per cent w/v solution of ofloxacin
– a stainless steel column 15 cm x 4.6 mm packed with
RS in mobile phase.
octadecylsilane bonded to porous silica (5 µm) (such as
Chromatographic system TSK GEL),
– a stainless steel column 25 cm x 4.6 mm packed with – mobile phase: a mixture of 20 volumes of acetonitrile,
octadecylsilane bonded to porous silica (5 µm), 80 volumes of phosphate buffer pH 7.25 prepared by

855
OFLOXACIN TABLETS IP 2007

dissolving 2.54 g of tetrabutyl ammonium hydrogen Reference solution (b). Dilute 1 ml of reference solution (a) to
sulphate and 3.56 g of disodium hydrogen phosphate 100 ml with methanol.
in 1000 ml water, Chromatographic system
– flow rate. 1.5 ml per minute, – a stainless steel column 10 cm x 4.6 mm packed with
– spectrophotometer set at 254 nm, octadecylsilyl silica gel for chromatography (5 µm),
– a 20 µl loop injector. – mobile phase: a mixture of 10 volumes of acetonitrile
Inject the reference solution. The test is not valid unless the and 90 volumes of phosphate buffer pH 2.4 prepared by
relative standard deviation for replicate injections is not more dissolving 27.2 g of monobasic potassium phosphate
than 2.0 per cent. in 1000 ml of water, adjust the pH to 2.4 with
orthophosphoric acid,
Inject the test solution and the reference solution.
– flow rate. 2 ml per minute,
Calculate the content of C18H20FN3O4. – spectrophotometer set at 294 nm,
Storage. Store protected from light. – a 10 µl loop injector.
Inject reference solution (a). The test is not valid unless the
column efficiency in not less than 1400 theoretical plates.
Ofloxacin Tablets Inject the test solution and reference solution (b). Run the
chromatogram three times of the principal peak. In the
Ofloxacin Tablets contain Ofloxacin. chromatogram obtained with the test solution, the area of any
Ofloxacin Tablets contain not less than 90.0 per cent and not secondary peak is not more than the area of the peak in the
more than 110.0 per cent of the stated amount of ofloxacin, chromatogram obtained with reference solution (b) (1.0 per
C18H20FN3O4. cent) and the sum of areas of all the secondary peaks is not
more than twice the area of the peak in the chromatogram
Identification obtained with the reference solution (b) (2.0 per cent).

In the Assay, the principal peak in the chromatogram obtained Other tests. Comply with the tests stated under the Tablets.
with the test solution corresponds to the peak in the Assay. Determine by liquid chromatography (2.4.14).
chromatogram obtained with the reference solution (a).
Test solution. Weigh and powder 20 tablets. Weigh accurately
Tests a quantity of powdered tablet containing 25 mg of Ofloxacin,
disperse in 60 ml of methanol and dilute to 100 ml with methanol
Dissolution (2.5.2). and filter.
Apparatus. No 1 Reference solution. A 0.025 per cent w/v solution of ofloxacin
Medium. 900 ml of 0.1 M hydrochloride acid. RS in methanol.
Speed and time. 50 rpm for 30 minutes. Chromatographic system
Withdraw a suitable volume of the medium and filter. Measure – a stainless steel column 15 cm x 4.6 mm packed with
the absorbance of the filtrate, suitably diluted with the medium octadecylsilane bonded to porous silica (5 µm),
if necessary, at the maximum at about 294 nm (2.4.7). Calculate – mobile phase: a mixture of 92 volumes of buffer solution
the content of C18H20FN3O4 in the medium from the absorbance prepared by dissolving 27.2 g of potassium dihydrogen
obtained from a solution of known concentration of phosphate in 1000 ml of water and adjust the pH to 2.4
ofloxacin RS in the same medium. with orthophosphoric acid and 8 volumes of
acetonitrile,
D. Not less than 75 per cent of the stated amount of
– flow rate. 2 ml per minute,
C18H20FN3O4.
– spectrophotometer set at 294 nm,
Related substances. Determine by liquid chromatography – a 10 µl loop injector.
(2.4.14).
Inject the reference solution. The test is not valid unless the
Test solution. Weigh and powder 20 tablets. Weigh accurately relative standard deviation for replicate injections is not more
a quantity of powdered tablet containing 100 mg of Ofloxacin, than 2.0 per cent.
disperse in 60 ml of methanol and dilute to 100 ml with methanol
Inject the test solution and the reference solution.
and filter.
Calculate the content of C18H20FN3O4.
Reference solution (a). A 0.1 per cent w/v solution of
ofloxacin RS in methanol. Storage. Store protected from light and moisture.

856
IP 2007 OLANZAPINE TABLETS

Olanzapine – spectrophotometer set at 230 nm,


– a 10 µl loop injector.
Time Mobile phase A Mobile phase B
CH3 (in min.) ( per cent v/v) ( per cent v/v)
N 0 100 0
20 63 37
N
N 30 45 55
32 100 0
N CH3 38 100 0
S
H Inject reference solution (b). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
C17H20N4S Mol. Wt. 312.4 not less than 2000 theoretical plates.

Olanzapine is 2-methyl-4-(4-methyl-1-piperazinyl)-10H- Inject the test solution and reference solution (b). Run the
thieno[2,3-b][1,5]benzodiazepine chromatogram for three times, the principal peak due to
olanzapine. In the chromatogram obtained with the test
Olanzapine contains not less than 98.0 per cent and not more solution, the area of any secondary peak is not more than 0.5
than 102.0 per cent of C17H20N4S, calculated on the anhydrous times the area of the peak in the chromatogram obtained with
basis. reference solution (b) (0.5 per cent) and the sum of areas of all
Description. A yellow crystalline powder. the secondary peaks is not more than twice the area of the
peak in the chromatogram obtained with the reference solution
Identification (b) (2.0 per cent).
Determine by infrared absorption spectrophotometry (2.4.6). Heavy metals (2.3.13). 1.0 g complies with the limit test for
Compare the spectrum with that obtained with olanzapine RS heavy metals, Method B (20 ppm).
or with the reference spectrum of olanzapine. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Tests Water (2.3.43). Not more than 1.0 per cent, determined on
1.0 g.
Related substances. Determine by liquid chromatography
(2.4.14). Assay. Weigh accurately about 0.2 g, dissolve in 40 ml of
glacial acetic acid. Titrate with 0.1 M perchloric acid,
Solvent mixture. 40 volumes of water and 60 volumes of determining the end-point potentiometrically (2.4.25). Carry
acetonitrile. out a blank titration.
Test solution. Dissolve 50 mg of the substance under
1 ml of 0.1 M perchloric acid is equivalent to 0.01562 g of
examination in 25 ml of solvent mixture.
C17H20N4S.
Reference solution (a). A 0.2 per cent w/v solution of
Storage. Store protected from light and moisture, at a
olanzapine RS in solvent mixture.
temperature not exceeding 30º.
Reference solution (b). Dilute 1 ml of reference solution (a) to
100 ml with solvent mixture.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with Olanzapine Tablets
octadecylsilane bonded to porous silica (5 µm),
Olanzapine Tablets contain not less than 90.0 per cent and not
– column temperature 40º,
more than 110.0 per cent of the stated amount of olanzapine,
– mobile phase: A. a mixture of 80 volumes of buffer
C17H20N4S.
solution pH 6.8 prepared by dissolving 4.825 g of
sodium dihydrogen orthophosphate monohydrate in
Identification
1000 ml of water, adjust pH to 6.8 with 10 per cent w/v of
sodium hydroxide and 20 volumes of acetonitrile, In the Assay, the principal peak in the chromatogram obtained
B. acetonitrile, with the test solution corresponds to the peak in the
– flow rate. 1.2 ml per minute, chromatogram obtained with the reference solution.

857
OLEIC ACID IP 2007

Tests less than 2500 theoretical plates. The relative standard


deviation for replicate injections is not more than 2 per cent.
Dissolution (2.5.2).
Inject the test solution and the reference solution.
Apparatus. No 1
Medium. 900 ml of 0.01 M hydrochloric acid. Calculate the content of C17H20N4S.
Speed and time. 50 rpm for 45 minutes. Storage. Store protected from light and moisture, at a
Withdraw a suitable volume of the medium and filter. Determine temperature not exceeding 25º.
by liquid chromatography (2.4.14).
NOTE – Protect all the solutions from light.
Test solution. Use the filtrate, if necessary dilute with Oleic Acid
dissolution medium.
Oleic Acid consists mainly of (Z)-octadec-9-enoic acid,
Reference solution. Weigh 16 mg of olanzapine RS, dissolve
C18H34O2, together with varying amounts of saturated and
in about 2.5 ml of acetonitrile and dilute to 25 ml with 0.01 M
other unsaturated fatty acids and is obtained by the hydrolysis
hydrochloroc acid.Dilute suitabely to get 0.0016 per cent w/v
of fats or fixed oils and separation of the liquid acids by
in dissolution medium.
expression or other suitable means. It may contain a suitable
Chromatographic system as described under Assay, using 50 antioxidant.
µl loop injector.
Description. A clear, yellowish to pale brown, oily liquid; odour,
Inject the reference solution . The test is not valid unless the characteristic. On exposure to air it darkens in colour and the
tailing factor is not more than 2.0. The column efficiency in odour becomes more pronounced.
not less than 2500 theoretical plates.
Inject the test solution and the reference solution. Identification
Calculate the content of C17H20N4S. A.To 1 ml add 1 ml of ethanol (95 per cent); the solution is
D. Not less than 70 per cent of the stated amount of C17H20N4S. clear. It turns orange or red on addition of 0.1 ml of methyl
orange solution.
Other tests. Comply with the tests stated under Tablets.
B.Take a mixture of 1 ml of nitric acid and 1 ml of water in a
Assay. Determine by liquid chromatography (2.4.14). test-tube with an internal diameter of about 12.5 mm and add
Test solution. Take 10 intact tablets to a suitable volumetric 1 ml of the substance under examination on the surface of the
flask, disperse in acetonitrile and dilute with 0.01 M mixture. Introduce 0.5 g of copper turnings into the lower
hydrochloroc acid to get final concentration of 0.01 per cent layer and allow to stand under a hood for 4 hours; the upper
w/v of Olanzapine. layer solidifies.
Reference solution. Weigh 10 mg of olanzapine RS, dissolve C.Complies with the test for Iodine value (2.3.28).
in about 25 ml of acetonitrile and dilute to 100 ml with 0.01 M
hydrochloroc acid. Tests
Chromatographic system Weight per ml (2.4.29). 0.889 g to 0.895 g.
– a stainless steel column 25 cm x 4.6 mm packed with
packed with octadecylsilyl silica gel for chromatography Peroxide value (2.3.35). Not more than 10.0.
(5 µm), Acid value (2.3.23). 195 to 202, determined on 0.5 g.
– mobile phase: a mixture of 70 volumes of buffer solution
Iodine value (2.3.28). 85 to 95, determined by Method A.
prepared by dissolving 3 g of ammonium dihydrogen
orthophosphate in 900 ml water, add 2 ml of Water-soluble acids. Shake 5 ml with 5 ml of water for
triethylamine and dilute to 1000 ml with water. Adjust 2 minutes, allow the liquids to separate and filter the aqueous
pH to 2.5 with orthophosphoric acid and 30 volumes layer through paper moistened with water. To the filtrate add
of methanol, 0.05 ml of methyl orange solution; the liquid does not become
– flow rate. 1 ml per minute, red.
– spectrophotometer set at 220 nm,
Neutral fats and mineral oils. Boil 1 ml with 5 ml of 0.5 M
– a 10 µl loop injector.
sodium carbonate and 25 ml of water in a large flask. The
Inject the reference solution. The test is not valid unless the solution, while still hot, is not more opalescent than
tailing factor is not more than 2.0, the column efficiency in not opalescence standard OS2 (2.4.1).

858
IP 2007 OMEPRAZOLE

Congealing point. Dry about 10 ml by heating to 110º with Tests


frequent stirring, transfer to a test-tube about 20 mm in diameter,
cool and when at 15º immerse the tube in a suitable water-bath Appearance of solution. A 2.0 per cent w/v solution in
so that the cooling takes place at the rate of 2º per minute. Stir dichloromethane is clear (2.4.1).
the sample with a thermometer; it does not become cloudy Light absorption (2.4.7). Absorbances of a freshly prepared
until the temperature has fallen to 10º. On further cooling it 2.0 per cent w/v solution in dichloromethane at 400 nm,
congeals to a white solid or semi-solid mass at about 4º. 500 nm and 600 nm are not more than 0.25, 0.10 and 0.10
Sulphated ash (2.3.18). Not more than 0.1 per cent. respectively.
Related substances. A. Determine by thin-layer chromato-
Storage. Store protected from light and moisture in a refrigerator
graphy (2.4.17), coating the plate with silica gel GF254.
(8º to 15º).
Mobile phase. A mixture of 50 volumes of benzene, 30 volumes
Labelling. The label states (1) where applicable, that it is used
of ethyl acetate and 10 volumes of methanol.
for external use only; (2) the name and concentration of any
added antioxidant. Test solution. Dissolve 0.4 g of the substance under
examination in 100 ml of ethanol.
Reference solution (a). A 0.4 per cent w/v solution of
omeprazole RS in ethanol.
Omeprazole Reference solution (b). A 0.004 per cent w/v solution of
omeprazole RS in ethanol.
H CH3 Reference solution (c). A 0.002 per cent w/v solution of
N O N
omeprazole RS in ethanol.
S
N OCH3 Apply to the plate 10 µl of each solution. After development,
H3CO
CH3 dry the plate in air and examine in ultraviolet light at 254 nm
immediately. Any secondary spot in the chromatogram
C17H19N3O3S Mol. Wt. 345.4 obtained with the test solution is not more intense than the
spot in the chromatogram obtained with reference solution (c)
Omeprazole is 5-methoxy-2-[[(4-methoxy-3,5- and the total intensity of all such spots in the chromatogram
dimethylpyridin-2-yl)methyl]sulfinyl] -1H-benzimidazole. obtained with the test solution is not more than the intensity
Omeprazole contains not less than 99.0 per cent and not more of the spot obtained with reference solution (b).
than 101.0 per cent of C17H19N3O3S, calculated on the dried B. In the Assay, the sum of the areas of all the secondary
basis. peaks is not greater than 1.5 per cent of the total area of all
NOTE — Perform the tests and assay in subdued light and peaks.
use low-actinic glassware. Heavy metals (2.3.13). The residue obtained in the test for
Description. A white or almost white powder. Sulphated ash, complies with the limit test for heavy metals,
Method B (20 ppm).
Identification Sulphated ash (2.3.18). Not more than 0.2 per cent.
Test A may be omitted if tests B and C are carried out. Tests B Loss on drying (2.4.19). Not more than 0.2 per cent, determined
and C may be omitted if test A is carried out. on 1.0 g by drying in an oven at 60º at a pressure not exceeding
0.7 kPa for 4 hours.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with omeprazole Assay. Determine by liquid chromatography (2.4.14).
RS or with the reference spectrum of omeprazole. Test solution. A 0.005 per cent w/v solution of the substance
B. When examined in the range 230 nm to 360 nm (2.4.7), a under examination in the mobile phase.
0.002 per cent w/v solution in 0.1 M sodium hydroxide shows Reference solution. A 0.005 per cent w/v solution of
absorption maxima at about 276 nm and 305 nm; the ratio of omeprazole RS in the mobile phase.
the absorbance at about 305 nm to that at about 276 nm, 1.6 to
Chromatographic system
1.8.
– a stainless steel column 30 cm x 3.9 mm, packed with
C. In the test for Related substances, the principal spot in the octadecylsilane bonded to porous silica (5 µm),
chromatogram obtained with the test solution corresponds to – mobile phase: a mixture of 65 volumes of phosphate
that in the chromatogram obtained with reference solution (a). buffer pH 7.4 and 35 volumes of acetonitrile,

859
OMEPRAZOLE CAPSULES IP 2007

– flow rate. 1 ml per minute, for 2 hours, drain the solution slowly without losing any
– spectrophotometer set at 302 nm, granules. Transfer them quantitatively to a 100-ml volumetric
– a 20 µl loop injector. flask, add 20 ml of 0.1 M sodium hydroxide and mix with the
Inject the reference solution. Repeat the procedure at least aid of ultrasound. Dilute to volume with 0.1 M sodium
five times and measure the peak responses of the peak due to hydroxide, centrifuge about 15 ml for 5 minutes and dilute
omeprazole. The relative standard deviation of the replicate 5.0 ml of the clear supernatant liquid to 50.0 ml with the mobile
injections is not more than 2.0 per cent. phase. Using the resulting solution as the test solution, carry
out the determination as described in the Assay. Calculate the
Calculate the content of C17H19N3O3S. content of C17H19N3O3S in the supernatant liquid. Calculate
Storage. Store protected from light and moisture in a refrigerator the percentage of omeprazole released in the acid medium by
(2º to 8º). subtracting the content of C17H19N3O3S in the test solution
from the total content of omeprazole determined in the Assay.
NOTE — A combination of elevated temperatures (37º-50º)
and high humidity degrades Omeprazole. It rapidly degrades Not more than 15 per cent of the stated amount of C17H19N3O3S
under acidic conditions. is dissolved in 2 hours.
B. Apparatus. No 1
Medium. 900 ml of phosphate buffer pH 6.8,
Omeprazole Capsules Speed and time. 100 rpm and 45 minutes.
Omeprazole Capsules contain enteric-coated granules of Tap the granules from a capsule slightly with a glass rod to
Omeprazole. make them settle to the bottom. Rotate the paddle at 100 rpm
for 45 minutes and filter the solution. Using the filtered medium
Omeprazole Capsules contain not less than 90.0 per cent and as the test solution, carry out the determination as described
not more than 110.0 per cent of the stated amount of in the Assay. Calculate the content of C17H19N3O3S in the
omeprazole, C17H19N3O3S. medium.
NOTE — Perform the tests and assay in subdued light and D. Not less than 70 per cent of the stated amount of
use low-actinic glassware. C17H19N3O3S.
Identification Other Tests. Comply with the tests stated under Capsules.
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
A.To a quantity of the contents of the capsules containing
on 0.5 g of the contents of the capsules by drying in an oven
50 mg of Omeprazole in a 100-ml volumetric flask add about
at 60º at a pressure not exceeding 0.7 kPa for 4 hours.
70 ml of 0.1 M sodium hydroxide. Mix in an ultrasonic bath for
about 5 minutes and heat on a water-bath for 10 minutes. Assay. Determine by liquid chromatography (2.4.14).
Cool, make up to volume with 0.1 M sodium hydroxide and Test solution. Mix the contents of 20 capsules. Weigh and
filter. Dilute 2 ml of the filtrate to 100 ml with 0.1 M sodium transfer the granules containing about 20 mg of Omeprazole
hydroxide. to a 100-ml volumetric flask, add 20 ml of 0.1 M sodium
When examined in the range 230 nm to 360 nm (2.4.7), the hydroxide, mix with the aid of ultrasound and dilute to volume
resulting solution shows absorption maxima at about 276 nm with 0.1 M sodium hydroxide. Centrifuge for 5 minutes and
and 305 nm. dilute 5.0 ml of the clear supernatant liquid to 50.0 ml with the
mobile phase.
B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak due Reference solution. Take 20 mg of omeprazole RS in a dry,
to omeprazole in the chromatogram obtained with the reference stoppered test-tube, add 20.0 ml of 0.1 M sodium hydroxide,
solution. shake vigorously for 5 minutes and dilute 1.0 ml of the solution
with the mobile phase to produce 50.0 ml.
Tests Chromatographic system
Dissolution (2.5.2). – a stainless steel column 30 cm x 3.9 mm, packed with
octadecylsilane bonded to porous silica (5 µm),
A. Apparatus. No 1
– mobile phase: a mixture of 65 volumes of phosphate
Medium. 900 ml of 0.1 M hydrochloric acid,
buffer pH 7.4 and 35 volumes of acetonitrile,
Speed and time. 100 rpm and 2 hours. – flow rate. 1 ml per minute,
Tap the granules from a capsule slightly with a glass rod to – spectrophotometer set at 302 nm,
make them settle to the bottom. Rotate the paddle at 100 rpm – a 20 µl loop injector.

860
IP 2007 ORAL REHYDRATION SALTS

Inject the reference solution. Repeat the procedure at least Oral Rehydration Salts contain not less than 90.0 per cent and
five times and measure the peak responses of the peak due to not more than 110.0 per cent of the stated amount of Dextrose
omeprazole. The relative standard deviation of the replicate (anhydrous) or Dextrose Monohydrate (as appropriate) and
injections is not more than 2.0 per cent. of the requisite amounts of sodium, Na, potassium, K, chloride,
Calculate the content of C17H19N3O3S in the capsules. Cl, and citrate, C6H5O7, calculated from the stated amounts of
the relevant constituents.
Storage. Store protected from light and moisture.
Description. A white to creamy-white, amorphous or
crystalline powder; odourless.

Oral Rehydration Salts Identification


ORS Powder A. When heated, melting and charring occurs and an odour of
burnt sugar is produced.
Oral Rehydration Salts are dry, homogeneously mixed powders
containing Dextrose, Sodium Chloride, Potassium Chloride B. Add a few drops of the solution prepared as directed in the
and either Sodium Bicarbonate or Sodium Citrate for use in label to 5 ml of potassium cupri-tartrate solution; a copious
oral rehydration therapy after being dissolved in the requisite red precipitate is produced on boiling.
amount of water. C. Gives reaction A of sodium salts, reaction A of potassium
They may contain suitable flavouring agents and, where salts and reaction A of chlorides (2.3.1).
necessary, suitable flow agents in the minimum quantity D. A quantity containing about 50 mg of citric acid gives the
required to achieve a satisfactory product but may not contain reactions of citrates (2.3.1).
artificial sweetening agents like mono- and/or poly-
saccharides. If saccharin/saccharin sodium or aspartame is Tests
used in preparations meant for paediatric use, the concentration Uniformity of weight. Comply with the test for contents of
of saccharin should be such that its daily intake is not more packaged dosage forms (2.5.6).
than 5 mg/kg of body weight and that of aspartame should be
such that its daily intake is not more than 40 mg/kg of body Seal test (only for sachets). Loosely bundle 10 sachets with a
weight. rubber band and submerge the bundle under water in a vacuum
desiccator maintained at a pressure not exceeding 18 kPa for
Strength. A formulation of reduced osmolarity (given below) one minute. Examine the bundle for any fine stream of bubbles.
recommended by the World Health Organization (WHO) for Re-establish normal pressure and open the bundle. No
the Diarrhoeal Diseases Control Programme, and of the United penetration of water is observed in any sachet.
Nations Children’s Fund (UNICEF).
Other Tests. Comply with the tests stated under Oral Powders.
Composition of the formulation in terms of the amount, in g, to
be dissolved in sufficient water to produce 1000 ml. Assay. Carry out the following assays on the well-mixed
contents of an individual sachet or on a suitable sample from
Sodium Chloride 2.6 the well-mixed contents of a bulk container. Where the amount
Dextrose (anhydrous) 13.5 in an individual sachet is insufficient to carry out all the assays,
or take a separate sachet for the Assay for citrate and for the
Dextrose Monohydrate 14.85 Assay for dextrose. For the Assays for total sodium, for
Potassium Chloride 1.5 potassium and for total chloride weigh accurately about 8.0 g
Sodium Citrate 2.9 and dissolve in sufficient water to produce 500.0 ml (solution
A).
The molar concentrations of sodium, potassium, chloride and
citrate ions in terms of millimoles per litre are given below: For total sodium — Dilute a suitable volume of solution A
mmol/l with a sufficient volume of a solution of strontium chloride
such that the final solution contains a 1500- to 2000-fold excess
Sodium 75
of strontium ions and determine by Method A for atomic
Potassium 20 absorption spectrophotometry (2.4.2), measuring at 589 nm
Chloride 65 and using sodium solution AAS, suitably diluted with the
Citrate 10 strontium chloride solution, for the standard solutions or,
Dextrose 75 alternately by Method A for flame photometry (2.4.4).
The total osmolar concentration of the solution in terms of 1 g of Sodium Chloride, and of Sodium Citrate is equivalent to
mOsmol per litre is 245. 0.3934 and 0.2345 g of Na respectively.

861
ORMELOXIFEN HYDROCHLORIDE IP 2007

For potassium — Dilute a suitable volume of solution A with Ormeloxifen Hydrochloride


a sufficient volume of solution of strontium chloride such
that the final solution contains a 1500- to 2000-fold excess of Centchroman Hydrochloride; Centchroman
strontium ions and determine by Method A for atomic
absorption spectrophotometry (2.4.2), measuring at 767 nm CH3
H3CO O
and using potassium solution AAS, suitably diluted with the CH3
strontium chloride solution, for the standard solutions or,
alternatively by Method A for flame photometry (2.4.4).
1 g of Potassium Chloride is equivalent to 0.5245 g of K. , HCl
For total chloride — Titrate 50 ml of solution A with 0.1 M
silver nitrate using potassium chromate solution as indicator.
O
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of Cl. N

1 g of Sodium Chloride, and of Potassium Chloride is


equivalent to 0.6066 and 0.4756 g of Cl respectively. C30H35NO3,HCl Mol. Wt. 493.5
For citrate — Weigh accurately about 2.0 g and dissolve in Ormeloxifen Hydrochloride is trans-7-methoxy-2,2-dimethyl-
50 ml of anhydrous glacial acetic acid by heating at about 3-phenyl-4[4-(2-pyrrolidinoethoxy)phenyl]chroman
50º. Cool, allow to stand for 10 minutes. Titrate with 0.1 M hydrochloride.
perchloric acid, using 1-naphtholbenzein solution as
indicator. Carry out a blank titration. Ormeloxifen Hydrochloride contains not less than 98.5 per
cent and not more than 101.5 per cent of C30H35NO3,HCl,
1 ml of 0.1 M perchloric acid is equivalent to 0.006303 g of calculated on the dried basis.
C6H5O7.
Description. A white to off-white powder.
1 g of Sodium Citrate is equivalent to 0.6430 g of C6H5O7.
Identification
For dextrose — Weigh accurately about 7.5 g, dissolve in
40 ml of water, add 0.5 ml of dilute ammonia solution, and A. Determine by infrared absorption spectrophotometry (2.4.6).
dilute to 50 ml with water. Mix well, allow to stand for Compare the spectrum with that obtained with ormeloxifen
30 minutes, filter the solution, if turbid, and measure the optical hydrochloride RS or with the reference spectrum of
rotation in a 2-dm tube (2.4.22). The observed rotation, in ormeloxifen.
degrees, multiplied by 0.9477 and 1.0424 represents the weight
B. When examined in the range 230 nm to 360 nm (2.4.7), a
in g of C6H12O6 and C6H12O6,H2O respectively, as appropriate,
0.01 per cent w/v solution in methanol shows absorption
in the weight taken for the Assay.
maxima at about 278 and 282 nm.
Storage. Store protected from moisture in sachets, preferably C. Determine by thin-layer chromatography (2.4.17), coating
made of aluminum foil, containing sufficient powder for a single the plate with silica gel G.
dose or for a day’s treatment or for use in hospitals, in bulk
containers containing sufficient quantity to produce a volume Mobile phase. A mixture of 90 volumes of chloroform and
of solution appropriate to the daily requirements of the 10 volumes of methanol.
hospital concerned. Test solution. Dissolve 0.25 g of the substance under
Labelling. The label states (1) for sachets, the total weights, examination in 100 ml of chloroform.
in g, of each constituent; (2) for bulk containers, the weights, Reference solution. A 0.25 per cent w/v solution of ormeloxifen
in g, of each constituent in a stated quantity, in g, of the oral hydrochloride RS in chloroform.
powder; (3) the molar concentration in millimoles per litre of
Apply to the plate 2 µl of each solution. After development,
sodium, potassium, chloride and citrate ions, and of dextrose
dry the plate in air, until the odour of the solvents is no longer
as well as the total osmolar concentration in mOsmol per litre
detectable and spray with a 0.3 per cent w/v solution of
of the solution prepared from the oral powder; (4) the total
potassium permanganate. The principal spot in the
weight of the contents of the container; (5) the directions for
chromatogram obtained with the test solution corresponds to
use; (6) that any portion of the solution prepared from the oral
that in the chromatogram obtained with the reference solution.
powder that remains unused for 24 hours after preparation
should be discarded; (7) the storage conditions. g, of the oral D. To 1 ml of a 2 per cent w/v solution in ethanol (95 per cent)
powder. add 1 ml of a saturated solution of picric acid in water, stir

862
IP 2007 ORMELOXIFEN HYDROCHLORIDE TABLETS

well and set aside for 5 minutes. The yellow precipitate obtained six replicate analyses calculate the content of trans-ormeloxifen
after washing with water and drying at 60º for 4 hours melts at hydrochloride and cis-ormeloxifen hydrochloride in the
212º to 218º (2.4.21). substance under examination by comparing with the peak
responses for trans-ormeloxifen hydrochloride and cis-
Tests ormeloxifen hydrochloride respectively obtained with
Total basic substances. Weigh accurately 0.5 g, dissolve in reference solution.
25 ml of anhydrous glacial acetic acid, add 10 ml of a 5 per Storage. Store protected from moisture.
cent w/v solution of mercuric acetate in anhydrous glacial
acetic acid. Titrate with 0.1 M perchloric acid, using crystal
violet solution as indicator to a bluish green end-point. Carry
out a blank titration. Ormeloxifen Hydrochloride Tablets
1 ml of 0.1 M perchloric acid is equivalent to 0.04935 g of Centchroman Hydrochloride Tablets; Centchroman
C30H35NO3,HCl. Tablets
cis-Isomer. Not more than 1.5 per cent of the total content of Ormeloxifen Hydrochloride Tablets contain not less than
hydrochlorides of trans-and cis-isomers determined in the 90.0 per cent and not more than 110.0 per cent of the stated
Assay. amount of Ormeloxifen hydrochloride, C30H35NO3,HCl.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Identification
Loss on drying (2.4.19). Not more than 3.5 per cent, determined Shake a quantity of the powdered tablets containing 0.1 g of
on 1.0 g by drying in an oven at 105º for 4 hours. Ormeloxifen Hydrochloride with 10 ml of chloroform, filter
Assay. Determine by liquid chromatography (2.4.14). and evaporate the filtrate to dryness at a pressure not
exceeding 0.7 kPa. The residue complies with the following
Test solution. A 0.004 per cent w/v solution of the substance
tests.
under examination in the mobile phase.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution. A solution containing 0.004 per cent w/v
Compare the spectrum with that obtained with ormeloxifen
of trans-ormeloxifen hydrochloride RS and 0.0006 per cent
hydrochloride RS or with the reference spectrum of
w/v of cis-ormeloxifen hydrochloride RS in the mobile phase.
ormeloxifen.
Chromatographic system B. When examined in the range 230 nm to 360 nm (2.4.7), a
– a stainless steel column 25 cm x 4.6 mm, packed with 0.01 per cent w/v solution in methanol shows absorption
octadecylsilane bonded to porous silica (5 µm), maxima at about 278 and 282 nm.
– mobile phase: 80 volumes of acetonitrile and 20 volumes
of water containing 0.04 per cent w/v solution of C. Determine by thin-layer chromatography (2.4.17), coating
tetramethylammonium hydroxide adjusted to pH 7.6 the plate with silica gel G.
with phosphoric acid, Mobile phase. A mixture of 90 volumes of chloroform and
– flow rate. 1.5 ml per minute, 10 volumes of methanol.
– spectrophotometer set at 280 nm, Test solution. Dissolve 0.25 g of the substance under
– a 20 µl loop injector. examination in 100 ml of chloroform.
Inject the reference solution and use an attenuation such that Reference solution. A 0.25 per cent w/v solution of ormeloxifen
the response for the principal peak due to trans-ormeloxifen hydrochloride RS in chloroform.
hydrochloride is more than 50 per cent of the full-scale
deflection of the recorder. Make a total of six injections. When Apply to the plate 2 µl of each solution. After development,
the chromatograms are recorded under the conditions dry the plate in air, until the odour of the solvents is no longer
described above, trans-ormeloxifen hydrochloride is eluted detectable and spray with a 0.3 per cent w/v solution of
before cis-ormeloxifen hydrochloride.The test is not valid potassium permanganate. The principal spot in the
unless the relative standard deviation of six replicate injections chromatogram obtained with the test solution corresponds to
is not greater than 6 per cent and the resolution between the that in the chromatogram obtained with the reference solution.
peaks due to cis-ormeloxifen hydrochloride and trans- Tests
ormeloxifen hydrochloride is greater than 1. If necessary, adjust
the proportions and the flow rate of the mobile phase to obtain Dissolution (2.5.2).
proper resolution. Inject a suitable volume of test solution Apparatus. No 2
and record the response. From the average peak areas of the Medium. 900 ml of water.

863
ORPHENADRINE CITRATE IP 2007

Speed and time. 100 rpm and 45 minutes. hydrochloride and cis-ormeloxifen hydrochloride in the
Withdraw a suitable volume of the medium and filter. Measure substance under examination.
the absorbance of the filtrate, suitably diluted if necessary, at Storage. Store protected from moisture.
the maximum at about 278 nm (2.4.7). Calculate the content of
C30H35NO3,HCl in the medium from the absorbance obtained
from a solution of known concentration of Ormeloxifen
hydrochloride RS in water. Orphenadrine Citrate
D. Not less than 70 per cent of the stated amount of
CH3
C30H35NO3,HCl.
N
cis-Isomer. Not more than 1.5 per cent of the total content of CH3 O CH3
HO COOH
hydrochlorides of trans-and cis-isomers determined in the ,
Assay. HOOC COOH

Other Tests. Comply with the tests stated under Tablets.


Assay. Determine by liquid chromatography (2.4.17).
C18H23NO,C6H807 Mol.461.5
Test solution. Weigh and powder 20 tablets. Shake a quantity
Orphenadrine Citrate is (RS)-dimethyl[2-(2-
of the powder containing 0.1 g of Ormeloxifen Hydrochloride
methylbenzhydryloxy)ethyl]amine dihydrogen citrate.
with three quantities, each of 5 ml, of methanol, centrifuge
each extract, dilute the combined extracts to 25 ml with Orphenadrine Citrate contains not less than 98.5 per cent and
methanol and then dilute 250 µl of the resulting solution to 25 not more than 101.0 per cent of C18H23NO, C6H807, calculated
ml with the mobile phase. on the dried basis.
Reference solution. A solution containing 0.004 per cent w/v Description. A white or almost white, crystalline powder;
of trans-ormeloxifen hydrochloride RS and 0.0006 per cent odourless or almost odourless.
w/v of cis-ormeloxifen hydrochloride RS in the mobile phase.
Identification
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Test A may be omitted if tests B, C, D and E are carried out.
octadecylsilane bonded to porous silica (5 µm), Tests B, C and D may be omitted if tests A and E are carried
– mobile phase: 80 volumes of acetonitrile and 20 volumes out.
of water containing 0.04 per cent w/v solution of A. Determine by infrared absorption spectrophotometry,(2.4.6).
tetramethylammonium hydroxide adjusted to pH 7.6 Compare the spectrum with that obtained with orphenadrine
with phosphoric acid, citrate RS.
– flow rate. 1.5 ml per minute,
B. When examined in the range 230 nm to 360 nm (2.4.7), a
– spectrophotometer set at 280 nm,
0.06 per cent w/v solution in ethanol (95 per cent) shows
– a 20 µl loop injector. absorption maxima at 258 nm, 264 nm and 271 nm; absorbances
Inject the reference solution and use an attenuation such that at the maxima are about 0.68, 0.72 and 0.47 respectively.
the response for the principal peak due to trans-ormeloxifen C. Dissolve 50 mg in 10 ml of ethanol (50 per cent), add 10 ml
hydrochloride is more than 50 per cent of full-scale deflection of picric acid solution and allow to stand. The precipitate.
of the recorder. Make a total of six injections. When the after recrystallisation from ethanol (95 per cent), melts at
chromatograms are recorded under the conditions described about 89º or at about 107º (2.4.21).
above, trans-ormeloxifen hydrochloride is eluted before cis-
ormeloxifen hydrochloride.The test is not valid unless the D. Dissolve 5 mg in 2 ml of sulphuric acid; an orange-red
relative standard deviation of six replicate injections is not colour is produced.
greater than 6 per cent and the resolution between the peaks E. To 1 g add 10 ml water and 2 ml of 5 M sodium hydroxide,
due to cis-ormeloxifen hydrochloride and trans-ormeloxifen shake with two quantities, each of 10 mI, of chloroform and
hydrochloride is greater than 1.If necessary, adjust the discard the chloroform. Heat the aqueous solution to boiling
proportions and the flow rate of the mobile phase to obtain with an excess of mercuric sulphate solution, filter if necessary
proper resolution. Inject a suitable volume of test solution and boil the resulting solution with 0.2 ml of dilute potassium
and record the response. From the average peak areas of the permanganate solution; the solution is decolorised and a
six replicate analyses calculate the content of trans-ormeloxifen white precipitate is produced.

864
IP 2007 ORPHENADRINE HYDROCHLORIDE

Tests Orphenadrine Hydrochloride


Quaternary ammonium salt. Determine by thin-layer C18H23NO,HCl Mol. Wt.305.8
chromatography (2.4.17), coating the plate with silica gel G.
Orphenadrine Hydrochloride is (RS)-dimethyl [2-
Mobile phase. A mixture of 50 volumes of 2-propanol, (2methylbenzhydryloxy) ethyl]amine hydrochloride.
30 volumes of butyl acetate. 15 volumes of water and
5 volumes of strong ammonia solution. Orphenadrine Hydrochloride contains not less than 98.5 per
cent and not more than 101.0 per cent of C18H23NO,HCl.
Test solution. Dissolve 0.1 g of the substance under calculated on the dried basis.
examination in 10 ml methanol.
Description. A white or almost white, crystalline powder;
Reference solution. A 0.005 per cent w/v solution of odourless or almost odourless.
ethyldimethyl [2-(2-methylbenzhydryloxy)ethyl ]ammonium
chloride RS. Identification
Apply to the plate 5 µl of each solution. After development, Tests A and F may be omitted if tests B, C, D and E are carried
dry the plate in air and spray with dilute potossium out. Tests B, C and D may be omitted if tests A, E and F are
iodobismuthate solution. Any spot corresponding to carried out.
ethyldimethyl [2-(2-methylbenzhydryloxy) ethyl] ammonium
chloride in the chromatogram obtained with the test solution A. Determine by infrared absorption spectrophotometry,(2.4.6).
is not more intense than the spot in the chromatogram obtained Compare the spectrum with that obtained with orphenadrine
with the reference solution. hydrochloride RS.
Secondary amine. Determine by thin-layer chromatography B. When examined in the range 230 nm to 360 nm (2.4.7), a
(2.4.17) coating the plate with silica gel GF254. 0.06 per cent w/v solution in ethanol (95 per cent) shows
three absorption maxima. at about 258 nm,264 nm and 271 nm;
Mobile phase. A mixture of 96 volumes of 1-butanol and
absorbances at the maxima, about 1.07, 1.13 and 0.73
4 volumes of strong ammonia solution.
respectively.
Test solution. Dissolve 0.4 g of the substance under
examination in 10 ml methanol. C. Dissolve about 5 mg in 2 ml of sulphuric acid; an orange-
red colour is produced.
Reference solution. A 0.02 per cent w/v solution of methyl [2-
(2-methylbenzhydryloxy)ethyl]amine hydrochloride RS. D. Dissolve about 50 mg in 10 ml of ethanol (95per cent), add
10 ml of picric acid solution and allow to stand. The
Apply to the plate 10 µl of each solution. After development, precipitate, after recrystallisation from ethanol (95 per cent)
dry in air and examine in ultra-violet light at 254 nm. Spray the melts at about 89º or at about 107º (2.4.21).
plate with dilute potassium iodobismuthate solution and
examine again. By each method of visualisation any spot Tests
corresponding to methyl [2-(2-methylbenzhydryloxy)
ethyl]amine hydrochloride in the chromatogram obtained with Quaternary ammonium salt. Determine by thin-layer
the test solution is not more intense than the spot in the chromatography (2.4.17), coating the plate with silica gel G.
chromatogram obtained with the reference solution. Mobile phase. A mixture of 50 volumes of 2-propanol,
Heavy Metals (2.3.13). 2.0 g complies with the limit test for 30 volumes of butyl acetate. 15 volumes of water and
heavy metals, Method B (10 ppm). Use 2 ml of lead standard 5 volumes of strong ammonia solution.
solution (10 ppm Pb) to prepare the standard. Test solution. Dissolve 0.1 g of the substance under
Sulphated ash (2.3.18). Not more than 0.1 per cent. examination in 10 of methanol.
Loss on drying (2.3.19). Not more than 0.5 per cent, determined Reference solution. A 0.005 per cent w/v solution of
on 1.0 g by drying in an oven at 105° for 3 hours. ethyldimethyl [2-(2-methylbenzhydryloxy) ethyl] ammonium
chloride RS.
Assay. Weigh accurately about I.0 g dissolve in 30 ml of
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric Apply to the plate 5 µl of each solution. After development,
acid, determining the end-point potentiometrically (2.4.25). dry the plate in air and spray with dilute potossium
Carry out a blank titration. iodobismuthate solution. Any spot corresponding to
ethyldimethyl [2-(2-methylbenzhydryloxy) ethyl] ammonium
1 ml of 0.1 M perchloric acid is equivalent to 0.04615 g of
chloride in the chromatogram obtained with the test solution
C18H23NO, C6H807.
is not more intense than the spot in the chromatogram obtained
Storage. Store protected from light. with the reference solution.

865
ORPHENADRINE TABLETS IP 2007

Secondary amine. Determine by thin-layer chromatography B. Dissolve 50 mg in 10 ml of ethanol (50 per cent), add 10 ml
(2.4.17) coating the plate with silica gel GF254. of picric acid solution and allow to stand. The precipitate.
after recrystallisation from ethanol (95 per cent), melts at
Mobile phase. A mixture of 96 volumes of l-butanol and
about 89º or at about 107º (2.4.21).
4 volumes of strong ammonia solution.
C. A 5 per cent w/v solution gives reaction A of chlorides
Test solution. Dissolve 0.4 g of the substance under
(2.3.1).
examination in 10 methanol.
Reference solution. A 0.02 per cent w/v solution of methyl [2- Tests
(2-methylbenzhydryloxy)ethyl]amine hydrochloride RS.
Other tests. Comply with the tests stated under Tablets.
Apply to the plate 10 µl of each solution. After development,
Assay. Weigh and powder 20 tablets. Weigh accurately a
dry in air and examine in ultra-violet light at 254 nm. Spray the
quantity of the powder containing about 70 mg Orphenadrine
plate with dilute potassium iodobismuthate solution and
Hydrochloride and dissolve as completely as possible in a
examine again. By each method of visualisation any spot
mixture of 5 ml of water and 5 ml of 2 M hydrochloric acid.
corresponding to methyl [2-(2-methylbenzhydryloxy)
Without delay extract with four quantities, each of 15 ml, of
ethyl]amine hydrochloride in the chromatogram obtained with
chloroform, filter the combined extracts and evaporate to about
the test solution is not more intense than the spot in the
20 ml. Add 30 ml of anhydrous glacial acetic and 2 ml of
chromatogram obtained with the reference solution.
mercuric acetate solution and titrate with 0.02 M perchloric
Heavy Metals (2.3.13). 2.0 g complies with the limit test for acid determining the end point potentiometrically (2.4.25).
heavy metals, Method B (10 ppm). Use 2 ml of lead standard Carry out a blank titration.
solution (10 ppm Pb) to prepare the standard. 1 ml of O.02 M perchloric acid is equivalent to 0.006117 g of
Sulphated ash (2.3.18). Not more than 0.1 per cent. C18H23NO,HCl.
Loss on drying (2.3.19). Not more than 0.5 per cent, determined Storage. Store protected from light and moisture.
on 1.0 g by drying in an oven at 105° for 3 hours.
Assay. Weigh accurately about 1.0 g, dissolve in 20 ml of
anhydrous glacial acetic acid, add 20 ml of mercuric acetate
solution and titrate with 0.1 M perchloric acid using Oseltamivir Phosphate
1-naphtholbenzein solution as indicator. Carry out a blank
titration. H3C
1 ml of 0.1 M perchloric acid is equivalent to 0.03058 g of H3C
C18H23NO, HCl.
O
H O CH3 , H3PO4
Storage. Store protected from light.
N
H3C H2N O
O

Orphenadrine Tablets C16H28N2O4, H3PO4 Mol. Wt. 410.4


Orphenadrine Hydrochloride Tablets Oseltamivir Phosphate is phosphoric acid salt of ethyl
(3R,4R,5S)-4-(acetylamino)-5-amino-3-(1-ethylpropoxy)-1-
Orphenadrine Tablets contain not less than 92.5 per cent and
cyclohexene-1-carboxylate.
not more than 107.5 per cent of the stated amount of
orphenadrine. hydrochloride, C18H23NO,HCl. Oseltamivir Phosphate contain not less than 98.0 per cent and
not more than 102.0 per cent of C16H28N2O4 .H3PO4, calculated
Identification on the dried basis.
Extract a quantity of the powdered tablets containing about Description. A white to off-white powder.
0.15 g of Orphenadrine Hydrochloride with chloroform, filter
and evaporate the filtrate to dryness. The residue complies Identification
with the following tests.
A. Determine by infrared absorption spectrophotometry (2.4.6).
A. Dissolve 5 mg in 2 ml of sulphuric acid; an orange-red Compare the spectrum with that obtained with oseltamivir
colour is produced. phosphate RS.

866
IP 2007 OSELTAMIVIR CAPSULES

B. In the Assay, the principal peak in the chromatogram Loss on drying (2.4.19). Not more than 0.5 per cent, determined
obtained with the test solution corresponds to the peak in the on 1 g by drying in an oven at 105º.
chromatogram obtained with the reference solution. Assay. Determine by liquid chromatography (2.4.14).
Tests Test solution. Dissolve 50 mg of the substance under
examination in 50.0 ml of the mobile phase. Dilute 10.0 ml of
Specific optical rotation (2.4.22). - 42.0º to - 48.0º, determined
the solution to 50.0 ml with the mobile phase.
in a 1.0 per cent w/v solution in methanol.
Related substances. Determine by liquid chromatography Reference solution. A 0.02 per cent w/v solution of oseltamivir
(2.4.14). phosphate RS in the mobile phase.

NOTE — Prepare the solutions immediately before use. Chromatographic system


– a stainless steel column 15 cm x 4.6 mm, packed with
Test solution. Dissolve 25 mg of the substance under octylsilane bonded to porous silica (5 µm) (such as YMC
examination in 25 ml of the mobile phase. pack PRO C8),
Reference solution (a). A 0.1 per cent w/v solution of – column temperature 50º,
oseltamivir phosphate RS in the mobile phase. – mobile phase: a mixture of 66 volumes of buffer solution
prepared by dissolving 6.8 g of anhydrous monobasic
Reference solution (b). Dilute 1 ml of reference solution (a) to
potassium phosphate in 1000 ml of water, adjusting the
100 ml with the mobile phase.
pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
Chromatographic system of methanol and 23.5 volumes of acetonitrile,
– a stainless steel column 15 cm x 4.6 mm, packed with – flow rate. 1 ml per minute,
octylsilane bonded to porous silica (5 µm) (such as YMC – spectrophotometer set at 207 nm,
pack PRO C8), – a 20 µl loop injector.
– column temperature 50º,
Inject the reference solution. The test is not valid unless the
– mobile phase: a mixture of 66 volumes of a buffer solution
tailing factor is not more than 2.0, the column efficiency in not
prepared by dissolving 6.8 g of anhydrous monobasic
less than 2000 theoretical plates and the relative standard
potassium phosphate in 1000 ml of water and adjusting
deviation for replicate injections is not more than 2.0 per cent.
the pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
of methanol and 23.5 volumes of acetonitrile, Inject the test solution and the reference solution.
– flow rate. 1 ml per minute, Calculate the content of C16H28N2O4,H3PO4.
– spectrophotometer set at 207 nm,
– a 20 µl loop injector. Storage. Store protected from light and moisture.

Inject reference solution (a). The test is not valid unless the
column efficiency is not less than 2000 theoretical plates and
the tailing factor is not more than 2.0.
Oseltamivir Capsules
Inject the test solution and reference solution (b). In the Oseltamivir Phosphate Capsules
chromatogram obtained with the test solution, the area of any Oseltamivir Capsules contain Oseltamivir Phosphate.
secondary peak is not more than 0.5 time the area of the peak
Oseltamivir Capsules contain not less than 90.0 per cent and
in the chromatogram obtained with the reference solution (b)
not more than 110.0 per cent of the stated amount of
(0.5 per cent) and the sum of all the secondary peaks is not
oseltamivir, C16H28N2O4.
more than the area of the peak in the chromatogram obtained
with the reference solution (b) (1.0 per cent). Identification
Phosphoric acid. 23.4 to 24.4 per cent, calculated on a dried
In the Assay, the principal peak in the chromatogram obtained
basis.
with the test solution corresponds to the peak in the
Weigh accurately about 0.2 g and dissolve in 40 ml of distilled chromatogram obtained with the reference solution.
water. Titrate with 0.1 M sodium hydroxide, determining the
end-point potentiometrically (2.4.25). Carry out a blank titration. Tests
1 ml of 0.1 M sodium hydroxide is equivalent to 0.0049 g of Dissolution (2.5.2).
phosphoric acid. Apparatus. No. 1
Heavy metals (2.3.13). 1 g complies with the limit test for heavy Medium. 900 ml of 0.1 M hydrochloric acid.
metals, Method A (20 ppm). Speed and time. 50 rpm and 45 minutes

867
OSELTAMIVIR ORAL SUSPENSION IP 2007

Withdraw a suitable volume of the medium and filter. Determine Test solution. Weigh accurately a quantity of the mixed
by liquid chromatography (2.4.14). contents of 20 capsules containing about 200 mg of oseltamivir,
Test solution. Use the filtrate. disperse in 100.0 ml of the mobile phase and filter. Dilute 5.0 ml
of the solution to 50.0 ml with the mobile phase.
Reference solution. Dissolve 50 mg of oseltamivir phosphate
Reference solution. A 0.02 per cent w/v solution of oseltamivir
RS in 20 ml of the mobile phase and dilute to 100 ml with the
phosphate RS in the mobile phase.
dissolution medium. Dilute 5 ml of the solution to 25 ml with
the dissolution medium. Chromatographic system
– a stainless steel column 15 cm x 4.6 mm, packed with
Use the chromatographic system described under Assay.
octylsilane bonded to porous silica (5 µm),
D. Not less than 80 per cent of the stated amount of – mobile phase: a mixture of 66 volumes of a buffer solution
C16H28N2O4. prepared by dissolving 6.8 g of anhydrous monobasic
Related substances. Determine by liquid chromatography potassium phosphate in 1000 ml of water and adjusting
(2.4.14). the pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
of methanol and 23.5 volumes of acetonitrile,
NOTE — Prepare the solutions immediately before use. – flow rate. 1.2 ml per minute,
Test solution. Weigh accurately a quantity of the mixed – spectrophotometer set at 207 nm,
contents of 20 capsules containing about 50 mg of oseltamivir, – a 20 µl loop injector.
disperse in 50 ml of mobile phase and filter. Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0, the column efficiency in not
Reference solution (a). A 0.1 per cent w/v solution of less than 2000 theoretical plates and the relative standard
oseltamivir phosphate RS in the mobile phase. deviation for replicate injections is not more than 2.0 per cent.
Reference solution (b). Dilute 1 ml of reference solution to Inject the test solution and the reference solution.
100 ml with the mobile phase.
Calculate the content of C16H28N2O4.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Storage. Store protected from moisture, at a temperature not
octylsilane bonded to porous silica (5 µm) (such as YMC exceeding 30º.
pack PRO C8), Labelling. The label states the strength in terms of the
– mobile phase: a mixture of 66 volumes of a buffer solution equivalent amount of oseltamivir.
prepared by dissolving 6.8 g of anhydrous monobasic
potassium phosphate in 1000 ml of water, adjusting the
pH to 6.0 with dilute sodium hydroxide, 24.5 volumes
of methanol and 23.5 volumes of acetonitrile,
Oseltamivir Oral Suspension
– flow rate. 1.5 ml per minute, Oseltamivir Phosphate Oral Suspension
– spectrophotometer set at 207 nm,
Oseltamivir Oral Suspension is a mixture consisting of
– a 20 µl loop injector.
Oseltamivir Phosphate with buffering agents and other
Inject reference solution (a). The test is not valid unless the excipients. It contains a suitable flavouring agent. It is filled in
column efficiency is not less than 2000 theoretical plates and a sealed container.
the tailing factor is not more than 2.0.
The suspension is constituted by dispersing the contents of
Inject the test solution and reference solution (b). In the the sealed container in the specified volume of water just
chromatogram obtained with the test solution, the area of any before issue.
secondary peak is not more than the area of the peak in the
Oseltamivir Oral Suspension contains not less than 90.0 per
chromatogram obtained with the reference solution (b) (1.0
cent and not more than 110.0 per cent of the stated amount of
per cent) and the sum of the areas of all the secondary peaks
oseltamivir C16H28N2O4.
is not more than twice the area of the peak in the chromatogram
obtained with the reference solution (b) (2.0 per cent). The contents of the sealed container comply with the
following requirement.
Other tests. Comply with the tests stated under Capsules.
Water (2.3.43). Not more than 2.5 per cent, determined on
Water (2.3.43). Not more than 5.0 per cent, determined on 0.5 g.
1.0 g.
Assay. Determine by liquid chromatography (2.4.14).
The constituted suspension complies with the requirements
NOTE — Prepare the solutions immediately before use. stated under Oral liquids and with the following requirements.

868
IP 2007 OXAZEPAM

Identification Test solution. Weigh accurately a quantity of the suspension


containing 60 mg of oseltamivir, dissolve in 250.0 ml of the
A. In the Assay, the principal peak in the chromatogram mobile phase and filter. Dilute 10.0 ml of the solution to 25.0 ml
obtained with the test solution corresponds to the peak in the with the mobile phase and filter.
chromatogram obtained with reference solution (b).
Reference solution. A 0.0125 per cent w/v solution of
B. To 2 ml of the reconstituted suspension, add 2 ml of dilute oseltamivir phosphate RS in the mobile phase.
nitric acid and 4 ml of a 10 per cent w/v solution of ammonium
molybdate and warm the solution. A bright yellow precipitate Chromatographic system
is formed. – a stainless steel column 15 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 µm),
Tests – mobile phase: a mixture of 60 volumes of a buffer solution
prepared by dissolving 6.8 g of potassium dihydrogen
pH (2.4.24). 3.0 to 5.0.
phosphate in 1000 ml of water and adjusting the pH to
Related substances. Determine by liquid chromatography 6.0 with dilute sodium hydroxide, 24.5 volumes of
(2.4.14). methanol and 23.5 volumes of acetonitrile,
NOTE — Prepare the solutions immediately before use. – flow rate. 1.2 ml per minute,
– spectrophotometer set at 207 nm,
Test solution. Weigh accurately a quantity of the suspension – a 20 µl loop injector.
containing 25 mg of oseltamivir, dissolve in 25 ml of the mobile
phase and filter. Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0, the column efficiency in not
Reference solution (a). A 0.1 per cent w/v solution of less than 2000 theoretical plates and the relative standard
oseltamivir phosphate RS in the mobile phase. deviation for replicate injections is not more than 2.0 per cent.
Reference solution (b). Dilute 1 ml of reference solution (a) to Inject the test solution and the reference solution.
100 ml with the mobile phase.
Calculate the content of C16H28N2O4.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Storage. Store protected from moisture, at a temperature not
octylsilane bonded to porous silica (5 µm), exceeding 30°.
– column temperature 35º, Labelling. The label states (1) the quantity of active ingredient
– mobile phase: 70 volumes of a buffer solution prepared in terms of the equivalent amount of oseltamivir; (b) the
by dissolving 6.8 g of potassium dihydrogen phosphate temperature of storage and the period during which the
in 1000 ml of water and adjusting the pH to 6.0 with constituted suspension may be expected to be satisfactory
dilute sodium hydroxide, 15 volumes of methanol and for use.
15 volumes of acetonitrile,
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 207 nm,
– a 20 µl loop injector. Oxazepam
Inject reference solution (a). The test is not valid unless the
H O
column efficiency is not less than 2000 theoretical plates and N
the tailing factor is not more than 2.0.
OH
Inject the test solution and reference solution (b). In the N
Cl
chromatogram obtained with the test solution, the area of any
secondary peak is not more than the area of the peak in the
chromatogram obtained with the reference solution (b)
(1.0 per cent) and the sum of areas of all the secondary peaks
is not more than 3 times the area of the peak in the
C15HIICIN2O2 Mol. Wt. 286.7
chromatogram obtained with the reference solution (b)
(3.0 per cent). Oxazepam is 7-chloro-3-hydroxy-5-phenyl-1,3-dihydro-2H-
1,4-benzodiazepin-2-one.
Other tests. Complies with the tests stated under Oral liquids.
Oxazepam contains not less than 98.5 per cent and not more
Assay. Determine by liquid chromatography (2.4.14).
than 101.0 per cent of C15HIICIN2O2, calculated on the dried
NOTE — Prepare the solutions immediately before use. basis.

869
OXAZEPAM TABLETS IP 2007

Description. A white or almost white, crystalline powder. Reference solution (c). Dilute 1 ml of reference solution (a) to
100 ml with acetone.
Identification Reference solution (d). Dilute 5 ml of solution (d) to 10 ml with
Test A may be omitted if tests B, C and D are carried out. Tests acetone.
B and D may be omitted if tests A and C are carried out. Apply to the plate 20 µl of each solution. Allow the mobile
A. Determine by infrared absorption spectrophotometry (2.4.6). phase to rise 17 cm in the same direction as the washing with
Compare the spectrum with that obtained with oxazepam RS. methanol. Dry in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with test
B. Prepare the solutions immediately before use, protected solution (a) is not more intense than the spot in the-
from light. chromatogram obtained with reference solution (c) and at most
Dissolve about 20 mg in sufficient ethanol (95 per cent) to one such spot is more intense than the spot in the
produce 100 ml. Dilute 10 ml of the solution to 50 ml with chromatogram obtained with reference solution (d). The test
ethanol (95 per cent) (solution A). Dilute 10 ml of solution A is not valid unless the chromatogram obtained with reference
to 100 ml with ethanol (95 per cent) (solution B). solution (b) shows two clearly separated spots.

When examined in the range 230 nm to 360 nm (2.4.7), solution Sulphated ash (2.3.18). Not more than 0.1 per cent.
A shows an absorption maximum at about 316 nm. When Loss on drying (2.3.19). Not more than 0.5 per cent, determined
examined in the range 220 nm to 250 nm, solution B shows an on 1.0 g by drying in an oven at 105º at a pressure not exceeding
absorption maximum at about 229 nm; absorbance at about 0.7 kPa.
229 nm, 1.220 to 1.300.
Assay. Weigh accurately about 0.25 g and dissolve in a mixture
C. In the test for Related substances, the principal spot in the of 10 ml of anhydrous glacial acetic acid and 90 ml of acetic
chromatogram obtained with test solution (b) corresponds to anhydride and titrate with 0.1 M perchloric acid determining
that in the chromatogram obtained with reference solution the end point potentiometrically (2.4.25). Carry out a blank
(b). titration.
D. Dissolve about 20 mg in a mixture of 5 ml of hydrochloric 1 ml of 0.1 M perchloric acid is equivalent to 0.02867 g of
acid and 10 ml of water. Heat to boiling for 5 minutes and cool. Cl5HIICIN2O2.
Add 2 ml of a 0.1 per cent w/v solution of sodium nitrite and Storage. Store protected from light and moisture.
allow to stand for 1 minute. Add 1 ml of a 0.5 per cent w/v
solution of sulphamic acid, mix and allow to stand for 1 minute.
Add 1 ml of 0.1 per cent w/v solution of a N- (1-naphthyl)
ethylenediamine dihydrochloride; a red colour is produced. Oxazepam Tablets
Oxazepam Tablets contain not less than 90.0 per cent and not
Tests more than 110.0 per cent of the stated amount of oxazepam,
CI5H11CIN2O2.
Related substances: Carry out the test protected from light.
Determine by thin-layer chromatography (2.4.17), coating the Identification
plate with silica gel GF254.
A. Extract a quantity of the powdered tablets containing
Wash the plate with methanol before use. 20 mg of Oxazepam with 25 ml of chloroform, filter, evaporate
Mobile phase. A mixture of 100 volumes of dichloromethane to dryness and dry the residue at 60° at a pressure not exceeding
and 10 volumes of methanol. 0.7 kPa.

Test solution (a). Dissolve 50 mg of the substance under On the residue, determine by infrared absorption
examination in sufficient acetone to produce 10 ml. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with oxazepam RS or with the reference spectrum of
Test solution (b). Dilute 2 ml of test solution (a) to 10 ml with oxazepam.
acetone.
B. When examined in the range 210 nm to 360 nm (2.4.7), the
Reference solution (a). Dissolve 10 mg of oxazepam RS in solution obtained in the Assay shows absorption maxima at
sufficient acetone to produce 10 ml. about 230 nm and 316 nm.
Reference solution (b). Dissolve 10 mg each of oxazepam RS
Tests
and bromazepam RS in sufficient acetone to produce
10 ml. Related substances. Carry out the test protected from light.

870
IP 2007 OXPRENOLOL HYDROCHLORIDE

Determine by thin-layer chromatography (2.4.17) coating the Oxprenolol Hydrochloride contains not less than 98.5 per cent
plate with silica gel GF 254. and not more than 101.5 per cent of C15H23NO3,HCl, calculated
Wash the plate with methanol before use. on the dried basis.

Mobile phase. A mixture of 100 volumes of dichloromethane Description. A white or almost white, crystalline powder.
and 10 volumes of methanol. Identification
Test solution. Shake a quantity of powdered tablets containing
30 mg of Oxazepam with 6 ml of acetone and centrifuge. Test A may be omitted if tests B and C are carried out. Test B
may be omitted if tests A and C are carried out.
Reference solution (a). Dilute 1 volume of the test solution to
100 volumes with acetone. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with oxprenolol RS
Reference solution (b). Dilute 1 volume of the test solution to or with the reference spectrum of oxprenolol.
500 volumes with acetone.
B. In the test for Related substances, the principal spot in the
Reference solution (c). A solution containing 0.1 per cent chromatogram obtained with test solution (b) corresponds to
w/v each of oxazepam RS and bromazepam RS. that in the chromatogram obtained with reference solution (c).
Apply to the plate 20 µl of each of the solutions. Allow the C. Gives reaction A of chlorides (2.3.1).
mobile phase to rise 17 cm in the same direction as in the
washing with methanol. Dry in air and examine in ultraviolet Tests
light at 254 nm. Any secondary spot in the chromatogram Appearance of solution. A 10.0 per cent w/v solution in carbon
obtained with the test solution is not more intense than the dioxide-free water is clear (2.4.1), and not more intensely
spot in the chromatogram obtained with reference solution (a) coloured than reference solution GYS6 (2.4.1).
(1 per cent) and not more than one such spot is more intense
than the spot in the chromatogram obtained with reference pH (2.4.24). 4.5 to 6.0, determined in a freshly prepared
solution (b) (0.2 per cent). The test is not valid unless the 10.0 per cent w/v solution.
chromatogram obtained with reference solution (c) shows two Related substances. Determine by thin-layer chromatography
clearly separated spots. (2.4.17), coating the plate with silica gel G.
Other tests. Comply with the tests stated under Tablets. Mobile phase. A mixture of 88 volumes of chloroform,
Assay. Weigh and powder 20 tablets. Weigh accurately a 12 volumes of methanol and 2 volumes of strong ammonia
quantity of the powder containing about 25 mg of Oxazepam solution.
and shake with 150 mI of ethanol (95 per cent) for 30 minutes. Test solution (a). Dissolve 0.5 g of the substance under
Add sufficient ethanol (95 per cent) to produce 250.0 ml and examination in 10 ml of a mixture of 90 volumes of chloroform
centrifuge. Dilute 5.0 ml of the supernatant liquid to 100.0 ml and 10 volumes of methanol.
with the same solvent and measure the absorbance of the Test solution (b). Dissolve 0.5 g of the substance under
resulting solution at the maximum at about 230 nm (2.4.7). examination in 100 ml of a mixture of 90 volumes of chloroform
Calculate the content of CI5HI1CIN2O2 taking 1250 as the and 10 volumes of methanol.
specific absorbance at 230 nm.
Reference solution (a). A 0.02 per cent w/v solution of the
Storage. Store protected from light at a temperature not substance under examination in a mixture of 90 volumes of
exceeding 30º. chloroform and 10 volumes of methanol.
Reference solution (b). A 0.01 per cent w/v solution of the
substance under examination in a mixture of 90 volumes of
Oxprenolol Hydrochloride chloroform and 10 volumes of methanol.
Reference solution (c). A 0.5 per cent w/v solution of
OH H oxprenolol hydrochloride RS in a mixture of 90 volumes of
O N CH3 chloroform and 10 volumes of methanol.
, HCl Apply to the plate 2 µl of each solution. Allow the mobile
CH2 CH3
O phase to rise 13 cm. Dry the plate in warm air for 10 minutes,
allow to cool, spray with anisaldehyde solution, heat at 105º
C15H23NO3,HCl Mol. Wt. 301.8
for 10 minutes and examine in daylight. Any secondary spot
Oxprenolol Hydrochloride is (RS)-1-(2-allyloxyphenoxy)-3- in the chromatogram obtained with test solution (a) is not
isopropylaminopropan-2-ol hydrochloride. more intense than the spot in the chromatogram obtained

871
OXPRENOLOL TABLETS IP 2007

with reference solution (a) and not more than one such spot is Mobile phase. A mixture of 88 volumes of chloroform,
more intense than the spot in the chromatogram obtained 12 volumes of methanol and 2 volumes of strong ammonia
with reference solution (b). solution.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Test solution. Extract a quantity of the powdered tablets
heavy metals, Method B (20 ppm). containing 0.25 g of Oxprenolol Hydrochloride with 5 ml of
Sulphated ash (2.3.18). Not more than 0.1 per cent. water, centrifuge and use the supernatant liquid.

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Reference solution. Dilute 1 volume of the test solution to
on 1.0 g by drying in an oven at 60º over phosphorus pentoxide 200 volumes with water.
at a pressure of 1.5 to 2.5 kPa for 6 hours. Apply to the plate 2 µl of each solution. Allow the mobile
Assay. Weigh accurately about 0.25 g, dissolve in 60 ml of phase to rise 13 cm. Dry the plate in warm air for 10 minutes,
anhydrous glacial acetic acid, add 5 ml of mercuric acetate allow to cool, spray with anisaldehyde solution, heat at 105º
solution. Titrate with 0.1 M perchloric acid, determining the for 10 minutes and examine in daylight. Any secondary spot
end-point potentiometrically (2.4.25).Carry out a blank in the chromatogram obtained with the test solution is not
titration. more intense than the spot in the chromatogram obtained
with the reference solution.
1 ml of 0.1 M perchloric acid is equivalent to 0.03018 g of
C15H23NO3,HCl. Other Tests. Comply with the tests stated under Tablets.
Storage. Store protected from light and moisture. Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 20 mg of Oxprenolol
Hydrochloride, add 25 ml of 0.1 M hydrochloric acid and
sufficient water to produce 250.0 ml. Mix with the aid of
Oxprenolol Tablets ultrasound for 5 minutes, shake for 15 minutes and filter.
Oxprenolol Hydrochloride Tablets Measure the absorbance of the resulting solution at the
maximum at about 273 nm (2.4.7). Calculate the content of
Oxprenolol Hydrochloride Tablets contain not less than C15H23NO3,HCl taking 74.5 as the specific absorbance at
95.0 per cent and not more than 105.0 per cent of the stated 273 nm.
amount of oxprenolol hydrochloride, C15H23NO3,HCl. The
tablets are coated. Storage. Store protected from moisture.

Identification
A.To a quantity of the powdered tablets containing 50 mg of
Oxprenolol Hydrochloride add 10 ml of water and 2 ml of
Oxygen
dilute sodium hydroxide solution, extract with 10 ml of O2 Mol. Wt. 32.0
chloroform and reserve the aqueous layer for test C. Dry the Oxygen contains not less than 99.0 per cent v/v of O2.
chloroform extract over anhydrous sodium sulphate, filter and
evaporate the filtrate to dryness. This monograph applies to oxygen for medicinal use only.
On the residue, determine by infrared absorption Description. A colourless gas; odourless.
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with oxprenolol hydrochloride RS or with the Identification
reference spectrum of oxprenolol hydrochloride. A. A glowing splinter of wood bursts into flame on contact
B. When examined in the range 230 nm to 360 nm (2.4.7), the with the gas.
solution obtained in the Assay shows an absorption maximum B. Shake with alkaline pyrogallol solution; the gas under
only at about 273 nm. examination is absorbed and the solution becoming dark
C. The aqueous layer obtained in test A gives reaction A of brown.
chlorides (2.3.1). C. When tested as described under Assay, not more than
D. The residue obtained in test A melts at about 76º (2.4.21). 1.0 ml of gas remains.

Tests Tests
Related substances. Determine by thin-layer chromatography Carbon dioxide. Not more than 300 ppm v/v, determined by
(2.4.17), coating the plate with silica gel G. using a carbon dioxide detector tube (2.1.1).

872
IP 2007 OXYPHENBUTAZONE

Carbon monoxide. Not more than 5 ppm v/v, determined by compounds and must not be treated with any compound that
using a carbon monoxide detector tube (2.1.1). will be irritating to the respiratory tract when the Oxygen
93 Per Cent is used.
Water vapour. Not more than 67 ppm v/v, determined by using
a water vapour detector tube (2.1.1). Labelling. Label each outlet “Oxygen 93 Per Cent”, when it is
piped directly from the collecting tank to the point of use. If it
Assay (2.3.33). Use 100 ml of the gas under examination and
is stored in cylinders, reduce the pressure by means of a
place spirals of freshly cleaned copper wire and 125 ml of
regulator. Measure the gases with a gas volume meter
ammonia buffer pH 10.9 in the pipette. The volume of the
downstream from the detector tube in order to minimise
residual gas in the burette is not more than 1.0 ml.
contamination or change of the specimens. The shoulder of
Storage. Store under pressure in metal cylinders of the type the cylinder should be painted white and the remainder should
conforming to the appropriate safety regulations. Valves and be painted black. The cylinder should carry a label stating
taps should not be lubricated with oil or grease. “Oxygen 93 Per Cent” and “For medicinal use”.
Labelling. The shoulder of the metal cylinder should be
painted white and the remainder should be painted black. The
cylinder should carry a label stating “Oxygen”. In addition,
“Oxygen” or the symbol “O2” should be stencilled in paint on
Oxyphenbutazone
the shoulder of the cylinder.
OH

Oxygen 93 Per Cent


Oxygen 93 Per Cent contains not less than 90.0 per cent and N , H2O
not more than 96.0 per cent, v/v of O2, the remainder consisting O N
mostly of argon and nitrogen. It is produced from air by the
H3 C
molecular sieve process. O
Description. A colourless gas; odourless.
C19H20N2O3,H2O Mol. Wt. 342.4
Identification
Oxyphenbutazone is (RS)-4-butyl-1-(4-hydroxyphenyl)-2-
A. A glowing splinter of wood bursts into flame on contact phenylpyrazolidine-3,5-dione monohydrate.
with the gas. Oxyphenbutazone contains not less than 98.0 per cent and
B. Shake with alkaline pyrogallol solution; the gas under not more than 101.0 per cent of C19H20N2O3, calculated on the
examination is absorbed and the solution becomes dark brown. anhydrous basis.
C. When tested as described under Assay, not more than Description. A white to yellowish white, crystalline powder;
10.0 ml and not less than 4.0 ml of gas remain. almost odourless.

Tests Identification
Carbon dioxide. Not more than 300 ppm v/v, determined by Test A may be omitted if tests B, C and D are carried out. Tests
using a carbon dioxide detector tube (2.1.1). B and D may be omitted if tests A and C are carried out.
Carbon monoxide. Not more than 5 ppm v/v, determined by A. Determine by infrared absorption spectrophotometry (2.4.6).
using a carbon monoxide detector tube (2.1.1). Compare the spectrum with that obtained with
oxyphenbutazone RS or with the reference spectrum of
Assay (2.3.33). Use 100 ml of the gas under examination and
oxyphenbutazone.
place spirals of freshly cleaned copper wire and 125 ml of
ammonia buffer pH 10.9 in the pipette. The volume of the B. When examined in the range 230 nm to 360 nm (2.4.7), a
residual gas in the burette is not more than 10.0 ml and not 0.001 per cent w/v solution in 0.01 M sodium hydroxide shows
less than 4.0 ml. an absorption maximum at about 254 nm; absorbance at about
254 nm, 0.71 to 0.77.
Storage. Store in cylinders or in a low pressure collecting
tank. Containers used for Oxygen 93 Per Cent must not be C. To 2 ml of a 1 per cent w/v solution in ethanol (95 per cent)
treated with any toxic, sleep-inducing or narcosis-producing add 2 ml of a 0.1 per cent w/v solution of 2,6-dichloroquinone-

873
OXYPHENBUTAZONE TABLETS IP 2007

4-chlorimide in ethanol (95 per cent) and 1 ml of sodium Oxyphenbutazone Tablets


carbonate solution; an intense green colour is produced.
Oxyphenbutazone Tablets contain not less than 92.5 per cent
D. To 0.1 g add 1 ml of glacial acetic acid and 2 ml of and not more than 107.5 per cent of the stated amount of
hydrochloric acid and boil under a reflux condenser for oxyphenbutazone, C19H20N2O3,H2O. The tablets are coated.
30 minutes. Cool, add 10 ml of water and filter. To the filtrate
add 3 ml of a 0.7 per cent w/v solution of sodium nitrite; a Identification
yellow colour develops. To 1 ml of the resulting solution add
a solution of 10 mg of 2-naphthol in 5 ml of sodium carbonate Extract a quantity of the powdered tablets containing 0.3 g of
solution; an orange-red precipitate is produced. Oxyphenbutazone with 60 ml of acetone, filter and evaporate
the filtrate to dryness. The residue complies with the following
Tests tests.

Appearance of solution. To 0.5 g add a mixture of 12 ml of 1 M A. When examined in the range 230 nm to 360 nm (2.4.7), a
sodium hydroxide and 8 ml of a 7.5 per cent w/v solution of 0.001 per cent w/v solution in 0.01 M sodium hydroxide shows
glycine, shake for 1 minute and maintain at 25º for exactly an absorption maximum at about 254 nm; absorbance at about
60 minutes. The solution is clear (2.4.1). 254 nm, 0.71 to 0.77.
B. To 2 ml of a 1 per cent w/v solution in ethanol (95 per cent)
Related substances. Determine by thin-layer chromatography
add 2 ml of a 0.1 per cent w/v solution of 2,6-dichloroquinone-
(2.4.17), coating the plate with silica gel HF254.
4-chlorimide in ethanol (95 per cent) and 1 ml of sodium
Mobile phase. A 0.02 per cent w/v solution of butylated carbonate solution; an intense green colour is produced.
hydroxytoluene in a mixture of 80 volumes of chloroform and
C. To 0.1 g add 1 ml of glacial acetic acid and 2 ml of
20 volumes of glacial acetic acid.
hydrochloric acid and boil under a reflux condenser for
Test solution. Dissolve 0.1 g of the substance under 30 minutes. Cool, add 10 ml of water and filter. To the filtrate
examination in 5 ml of a solution containing 0.02 per cent w/v add 3 ml of a 0.7 per cent w/v solution of sodium nitrite; a
of butylated hydroxytoluene in ethanol. yellow colour develops. To 1 ml of the resulting solution add
a solution of 10 mg of 2-naphthol in 5 ml of sodium carbonate
Reference solution. Dilute 1 ml of the test solution to 200 ml
solution; an orange-red precipitate is produced.
with the ethanolic butylated hydroxytoluene solution.
To prepare the plate, allow the mobile phase to rise 4 cm. Dry Tests
the plate in a current of cold air for 1 minute. Without delay Related substances. Determine by thin-layer chromatography
and under a current of nitrogen, apply separately to the plate (2.4.17), coating the plate with silica gel HF254.
5 µl of each solution prepared immediately before use. Develop
immediately, allowing the mobile phase to rise 10 cm. Dry the Mobile phase. A 0.02 per cent w/v solution of butylated
plate in a current of cold air for 15 minutes and examine in hydroxytoluene in a mixture of 80 volumes of chloroform and
ultraviolet light at 254 nm. Any secondary spot in the 20 volumes of glacial acetic acid.
chromatogram obtained with the test solution is not more Test solution. Shake a quantity of the powdered tablets
intense than the spot in the chromatogram obtained with the containing 0.2 g of Oxyphenbutazone with 10 ml of a solution
reference solution. containing 0.02 per cent w/v of butylated hydroxytoluene in
ethanol for 15 minutes and centrifuge. Use the supernatant
Heavy metals (2.3.13). 1.0 g complies with the limit test for
liquid.
heavy metals, Method B (20 ppm).
Reference solution. Dilute 1 ml of the test solution to 200 ml
Sulphated ash (2.3.18). Not more than 0.1 per cent.
with the ethanolic butylated hydroxytoluene solution.
Water (2.3.43). 5.0 to 6.0 per cent, determined on 0.5 g. To prepare the plate, allow the mobile phase to rise 4 cm. Dry
Assay. Weigh accurately about 0.5 g, dissolve in 50 ml of the plate in a current of cold air for 1 minute. Without delay
acetone and titrate with 0.1 M sodium hydroxide, using and under a current of nitrogen, apply separately to the plate
bromothymol blue solution as indicator and continuing the 5 µl of each solution prepared immediately before use. Develop
titration until the blue colour persists for at least 15 seconds. immediately, allowing the mobile phase to rise 10 cm. Dry the
Carry out a blank titration. plate in a current of cold air for 15 minutes and examine in
ultraviolet light at 254 nm. Any secondary spot in the
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03244 g of
chromatogram obtained with the test solution is not more
C19H20N2O3.
intense than the spot in the chromatogram obtained with the
Storage. Store protected from light. reference solution.

874
IP 2007 OXYTETRAXYCLINE DIHYDRATE

Dissolution (2.5.2). Oxytetracycline has a potency not less than 900 µg of


Apparatus. No 1 C22H24N2O9, per mg, calculated on the anhydrous basis.
Medium. 900 ml of phosphate buffer pH 7.6. Description. A tan yellow or light yellow (with or without a
Speed and time. 100 rpm and 30 minutes. greenish tinge), crystalline powder; odourless.
Withdraw a suitable volume of the medium and filter promptly Identification
through a membrane filter disc with an average pore diameter
not greater than 1.0 µm. Reject the first few ml of the filtrate A. Determine by thin-layer chromatography (2.4.17), coating
and dilute a suitable volume of the filtrate with the same the plate with the substance prepared by mixing 25 g of silica
solvent. Measure the absorbance of the resulting solution at gel G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of
the maximum at about 262 nm (2.4.7). Calculate the content of 0.1 M disodium edetate previously adjusted to pH 7.0 with
C19H20N2O3,H2O in the medium form the absorbance obtained dilute ammonia solution. After spreading the plate, allow it to
from a solution of known concentration of oxyphenbutazone stand at room temperature till it is dry (70 to 90 minutes).
RS in the same medium.
Mobile phase. The lower layer formed after shaking 200 ml of
D. Not less than 60 per cent of the stated amount of a mixture of 2 volumes of ethyl acetate, 2 volumes of
C19H20N2O3,H2O. chloroform and 1 volume of acetone with 25 ml of 0.1 M
Other Tests. Comply with the tests stated under Tablets. disodium edetate previously adjusted to pH 7.0 with dilute
ammonia solution.
Assay. Weigh 20 tablets and reduce to a fine powder. Weigh
accurately a quantity of the powder containing about 0.5 g of Test solution. Dissolve 0.05 g of the substance under
Oxyphenbutazone and extract successively with 30, 10 and examination in 100 ml of methanol.
10 ml of warm acetone. Filter the combined extracts, cool. Reference solution (a). A 0.05 per cent w/v solution of
Titrate with 0.1 M sodium hydroxide, using bromothymol oxytetracycline RS in methanol.
blue solution as indicator and continuing the titration until
Reference solution (b). A solution containing 0.05 per cent
the blue colour persists for at least 15 seconds. Carry out a
w/v each of demethylchlortetracycline hydrochloride RS,
blank titration.
oxytetracycline hydrochloride RS and tetracycline
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03424 g of hydrochloride RS in methanol.
C19H20N2O3,H2O.
Apply to the plate 1 µl of each solution, freshly prepared.
Storage. Store protected from moisture. After development, dry the plate in air, expose to the vapours
of ammonia and examine in ultraviolet light at 365 nm. The
principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Oxytetracycline Dihydrate with reference solution (a). The test is not valid unless the
chromatogram obtained with reference solution (b) shows
Oxytetracycline three clearly separated spots.
B. To about 0.5 mg add 2 ml of sulphuric acid; a red colour is
OH O OH O O produced. Add the solution to 1 ml of water; the colour
OH changes to yellow.
NH2
, 2H2O C. Dissolve about 2 mg in 5 ml of a 1 per cent w/v solution of
H OH sodium carbonate and add 2 ml of diazotised sulphanilic
H
H3C OH OH N acid solution; an orange-red to brownish-red colour is
H3C CH3 produced.

Tests
C22H24N2O9,2H2O Mol. Wt. 496.5
Oxytetracycline is (4S,4aR,5S,5aR,6S,12aS)-4-dimethyl pH (2.4.24). 4.5 to 7.5, determined in a 1.0 per cent w/v
amino-1,4,4a,5,5a,6,11,12a-octahydro-3,5,6,10,12,12a- suspension in freshly boiled and cooled water.
hexahydroxy-6-methyl-1,11-dioxonaphthacene-2- Specific optical rotation (2.4.22). –203º to –216º, determined
carboxamide, a substance produced by the growth of at 20º in a 1.0 per cent w/v solution in 0.1 M hydrochloric
certain strains of Streptomyces rimosus or obtained by any acid, after allowing the solution to stand protected from light
other means. It contains a variable quantity of water. for 30 minutes before measurement.

875
OXYTETRAXYCLINE DIHYDRATE IP 2007

Light absorption. Absorbance of a 0.002 per cent w/v solution Adjust the sensitivity so that the heights of the peaks in the
in buffer solution pH 2.0 at the maximum at about 353 nm, 0.58 chromatogram obtained with reference solution (d) are at least
to 0.62 (2.4.7). 50 per cent of the full-scale deflection of the recorder.
Light-absorbing impurities. A. Dissolve 20 mg in sufficient The test is not valid unless (a) the resolution between the first
of a mixture of 1 volume of 1 M hydrochloric acid and peak (4-epioxytetracycline) and the second peak
99 volumes of methanol to produce 10 ml. Absorbance of the (oxytetracycline) is at least 4.0 (b) the resolution between the
resulting solution at about 430 nm, when measured within second peak and the third peak (tetracycline) is at least 5.0
1 hour of preparing the solution, not more than 0.25, calculated (the content of 2-methyl-2-propanol in the mobile phase may
on the anhydrous basis (2.4.7). be adjusted if necessary) and (c) the symmetry factor for the
second peak is at most 1.25.
B. Dissolve 0.1 g in sufficient of a mixture of 1 volume of 1 M
hydrochloric acid and 99 volumes of methanol to produce Inject reference solution (a) six times. The test is not valid
10 ml. Absorbance of the resulting solution at about 490 nm, unless the relative standard deviation of the area of the peak
when measured within 1 hour of preparing the solution, not due to oxytetracycline is not greater than 1.0 per cent.
more than 0.20, calculated on the anhydrous basis (2.4.7).
Inject the test solution and reference solution (e). In the
Related substances. Determine by liquid chromatography chromatogram obtained with the test solution the area of any
(2.4.14). peak corresponding to 4-epioxytetracycline or tetracycline is
not greater than the area of the corresponding peak in the
Test solution. Dissolve 20 mg of the substance under
chromatogram obtained with reference solution (e). In the
examination in 25 ml of 0.01 M hydrochloric acid.
chromatogram obtained with the test solution the area of any
Reference solution (a). Dissolve 20 mg of oxytetracycline RS peak appearing on the tail of the principal peak is not greater
in 25 ml of 0.01 M hydrochloric acid. than 4.0 times that of the peak due to 4-epioxytetracycline in
the chromatogram obtained with reference solution (e).
Reference solution (b). Dissolve 20 mg of 4-epioxytetra-
cycline RS in 25 ml of 0.01 M hydrochloric acid. Heavy metals (2.3.13). 0.4 g complies with limit the test for
heavy metals, Method B (50 ppm).
Reference solution (c). Dissolve 20 mg of tetracycline
hydrochloride RS in 25 ml of 0.01 M hydrochloric acid. Sulphated ash (2.3.18). Not more than 0.5 per cent.
Reference solution (d). Dilute a mixture of 1.5 ml of reference Water (2.3.43). 4.0 to 9.0 per cent, determined on 0.5 g.
solution (a), 1.0 ml of reference solution (b) and 3.0 ml of
Assay. Determine by the microbiological assay of antibiotics,
reference solution (c) to 25 ml with 0.01 M hydrochloric
Method A or B (2.2.10), and express the results in µg of
acid.
oxytetracycline, C22H24N2O9, per mg.
Reference solution (e). Dilute a mixture of 1.0 ml of reference
Oxytetracycline intended for use in the manufacture of
solution (b) and 4.0 ml of reference solution (c) to 200 ml with
parenteral preparations without a further procedure for the
0.01 M hydrochloric acid.
removal of bacterial endotoxins complies with the following
Chromatographic system additional requirement.
– a stainless steel column 25 cm x 4.6 mm, packed with
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit
styrene- divinylbenzene co-polymer (8 to 10 µm),
per mg of Oxytetracycline.
– column. temperature 60º,
– mobile phase: add 60 g of 2-methyl-2-propanol to a Oxytetracycline intended for use in the manufacture of
volumetric flask with the aid of 200 ml of water, add parenteral preparations without a further sterilisation
60 ml of 0.33 M phosphate buffer pH 7.5, 50 ml of a procedure complies with the following additional
1.0 per cent w/v solution of tetrabutylammonium requirement.
hydrogen sulphate previously adjusted to pH 7.5 with Sterility. Complies with the test for sterility (2.2.11).
2 M sodium hydroxide and 10 ml of a 0.04 per cent w/v
solution of disodium edetate previously adjusted to Storage. Store protected from light and moisture. If it is
pH 7.5 with 2 M sodium hydroxide and dilute to intended for use in the manufacture of parenteral preparations,
1000.0 ml with water, the container should be sterile and sealed so as to exclude
– flow rate. 1 ml per minute, micro-organisms.
– spectrophotometer set at 254 nm, Labelling. The label states whether or not the contents are
– a 20 µl loop injector. intended for use in the manufacture of parenteral preparations.

876
IP 2007 OXYTETRAXYCLINE HYDROCHLORIDE

Oxytetracycline Injection Other Tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Oxytetracycline Injection is a sterile solution of oxytetracycline
Assay. Determine by the microbiological assay of antibiotics,
with or without one or more suitable buffering agents,
Method A or B (2.2.10), and express the result in mg of
anaesthetics, preservatives, antioxidants, complexing agents
oxytetracycline, C22H24N2O9, per ml.
and solvents.
Storage. Store protected from light.
Oxytetracycline Injection contains not less than 90.0 per cent
and not more than 120.0 per cent of the stated amount of Labelling. The label states (1) the strength in mg of anhydrous
anhydrous oxytetracycline, C22H24N2O9. oxytetracycline per ml; (2) that the contents are to be used for
intramuscular use only; (3) the names of any preservatives
Description. A clear, yellow to tan yellow solution. It may used.
have a greenish tinge.

Identification
Oxytetracycline Hydrochloride
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with the substance prepared by mixing 25 g of silica C22H24N2O9,HCl Mol. Wt. 496.9
gel G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of Oxytetracycline Hydrochloride is (4S,4aR,5S,5aR,6S, 12aS)-
0.1 M disodium edetate previously adjusted to pH 7.0 with 4-dimethylamino-1,4,4a,5,5a,6,11,12a-octahydro-3,5,
dilute ammonia solution. After spreading the plate, allow it to 6,10,12,12a-hexahydroxy-6-methyl-1,11-dioxonaphthacene-2-
stand at room temperature till it is dry (70 to 90 minutes). carboxamide hydrochloride, a substance produced by
Mobile phase. The lower layer formed after shaking 200 ml of the growth of certain strains of Streptomyces rimosus or
a mixture of 2 volumes of ethyl acetate, 2 volumes of obtained by any other means.
chloroform and 1 volume of acetone with 25 ml of 0.1 M Description. A pale yellow, crystalline powder; odourless;
disodium edetate previously adjusted to pH 7.0 with dilute hygroscopic.
ammonia solution.
Oxytetracycline Hydrochloride has a potency not less than
Test solution. Shake a quantity containing 10 mg of anhydrous 835 µg of C22H24N2O9, per mg, calculated on the anhydrous
oxytetracycline with 20 ml of methanol, centrifuging if basis.
necessary and use the clear supernatant liquid.
Reference solution (a). A 0.05 per cent w/v solution of Identification
oxytetracycline hydrochloride RS in methanol. A. Determine by thin-layer chromatography (2.4.17), coating
Reference solution (b). A solution containing 0.05 per cent the plate with the substance prepared by mixing 25 g of silica
w/v each of demethylchlortetracycline hydrochloride RS, gel G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of
oxytetracycline hydrochloride RS and tetracycline 0.1 M disodium edetate previously adjusted to pH 7.0 with
hydrochloride RS in methanol. dilute ammonia solution. After spreading the plate, allow it to
stand at room temperature till it is dry (70 to 90 minutes).
Apply to the plate 1 µl of each solution, freshly prepared.
After development, dry the plate in air, expose to the vapours Mobile phase. The lower layer formed after shaking 200 ml of
of ammonia and examine in ultraviolet light at 365 nm. The a mixture of 2 volumes of ethyl acetate, 2 volumes of
principal spot in the chromatogram obtained with the test chloroform and 1 volume of acetone with 25 ml of 0.1 M
solution corresponds to that in the chromatogram obtained disodium edetate previously adjusted to pH 7.0 with dilute
with reference solution (a). The test is not valid unless the ammonia solution.
chromatogram obtained with reference solution (b) shows Test solution. Dissolve 0.05 g of the substance under
three clearly separated spots. examination in 100 ml of methanol.
B. Add 0.1 ml to 2 ml of sulphuric acid; a red colour is produced. Reference solution (a). A 0.05 per cent w/v solution of
Add the solution to 1 ml of water; the colour changes to oxytetracycline hydrochloride RS in methanol.
yellow.
Reference solution (b). A solution containing 0.05 per cent
Tests w/v each of demethylchlortetracycline hydrochloride RS,
oxytetracycline hydrochloride RS and tetracycline
pH (2.4.24). 8.0 to 9.0. hydrochloride RS in methanol.
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit Apply to the plate 1 µl of each solution, freshly prepared.
per mg of Oxytetracycline. After development, dry the plate in air, expose to the vapours

877
OXYTETRAXYCLINE HYDROCHLORIDE IP 2007

of ammonia and examine in ultraviolet light at 365 nm. The Reference solution (e). Dissolve 20 mg of b -apo-
principal spot in the chromatogram obtained with the test oxytetracycline RS in 20 ml of 0.01 M sodium hydroxide and
solution corresponds to that in the chromatogram obtained dilute to 250 ml with 0.01 M hydrochloric acid.
with reference solution (a). The test is not valid unless the
Reference solution (f). Dilute a mixture of 1.5 ml of reference
chromatogram obtained with reference solution (b) shows
solution (a), 1.0 ml of reference solution (b), 3.0 ml each of
three clearly separated spots.
reference solutions (c) (d) and (e) to 25 ml with 0.01 M
B. To about 0.5 mg add 2 ml of sulphuric acid; a red colour is hydrochloric acid.
produced. Add the solution to 1 ml of water; the colour
Reference solution (g). Dilute a mixture of 1.0 ml of reference
changes to yellow.
solution (b), 4.0 ml of reference solution (c) and 40.0 ml of
C. Dissolve about 2 mg in 5 ml of a 1 per cent w/v solution of reference solution (e) to 200 ml with 0.01 M hydrochloric
sodium carbonate and add 2 ml of diazotised sulphanilic acid.
acid solution; an orange-red to brownish-red colour is
Chromatographic system
produced.
– a stainless steel column 25 cm x 4.6 mm, packed with
D. A 5 per cent w/v solution gives the reactions of chlorides styrene-divinylbenzene co-polymer (8 to 10 µm),
(2.3.1). – column. temperature 60º,
– mobile phase: transfer separately 30 g (for mobile phase
Tests A) or 100 g (for mobile phase B) of 2-methyl-2-propanol
pH (2.4.24). 2.0 to 3.0, determined in a 1.0 per cent w/v solution. to volumetric flasks with the aid of 200 ml of water; to
each flask add 60 ml of 0.33 M phosphate buffer pH 7.5,
Specific optical rotation (2.4.22). –188º to –200º, determined 50 ml of a 1.0 per cent w/v solution of tetrabutyl-
at 20º in a 1 per cent w/v solution in 0.1 M hydrochloric acid. ammonium hydrogen sulphate previously adjusted to
Light absorption (2.4.7). Absorbance of a 0.002 per cent w/v pH 7.5 with 2 M sodium hydroxide and 10 ml of a 0.04
solution in chloride buffer solution pH 2.0 at the maximum at per cent w/v solution of disodium edetate previously
about 353 nm, 0.54 to 0.58. adjusted to pH 7.5 with 2 M sodium hydroxide and
dilute each solution to 1000.0 ml with water.,
Light-absorbing impurities. A. Dissolve 20 mg in sufficient
– flow rate. 1 ml per minute,
of a mixture of 1 volume of 1 M hydrochloric acid and
– spectrophotometer set at 254 nm,
99 volumes of methanol to produce 10 ml. Absorbance of the
resulting solution at about 430 nm, when measured within – a 20 µl loop injector.
1 hour of preparing the solution, not more than 0.50, calculated Carry out a one-step gradient elution in the following manner.
on the anhydrous basis (2.4.7). Pump a mixture containing 30 volumes of mobile phase B and
B. Dissolve 0.1 g in sufficient of a mixture of 1 volume of 1 M 70 volumes of mobile phase A for 15 minutes, then pump a
hydrochloric acid and 99 volumes of methanol to produce mixture containing 30 volumes of mobile phase A and
10 ml. Absorbance of the resulting solution at about 490 nm, 70 volumes of mobile phase B for 15 minutes and finally
when measured within 1 hour of preparing the solution, not equilibrate with the first mixture. Adjust the sensitivity so that
more than 0.20, calculated on the anhydrous basis (2.4.7). the heights of the peaks in the chromatogram obtained with
reference solution (f) are at least 50 per cent of full-scale
Related substances. Determine by liquid chromatography deflection of the recorder.
(2.4.14).
The test is not valid unless, in the chromatogram obtained
Test solution. Dissolve 20 mg of the substance under with reference solution (f), (a) the resolution between the first
examination in 25 ml of 0.01 M hydrochloric acid. peak (4-epioxytetracycline) and the second peak
Reference solution (a). Dissolve 20 mg of oxytetracycline RS (oxytetracycline) is at least 4.0, (b) the resolution between the
in 25 ml of 0.01 M hydrochloric acid. second peak and the third peak (tetracycline) is at least 5.0, (c)
the resolution between the fourth peak (a -apo-oxytetracycline)
Reference solution (b). Dissolve 20 mg of 4-epioxytetra- and the fifth peak (b-apo-oxytetracycline) is at least 3.5, and
cycline RS in 25 ml of 0.01 M hydrochloric acid. (d) the symmetry factor of the second peak is at most 1.25. If
Reference solution (c). Dissolve 20 mg of tetracycline necessary adjust the proportions of the mobile phases used
hydrochloride RS in 25 ml of 0.01 M hydrochloric acid. to produce the one-step gradient elution. Adjust the time-
programme for the one-step gradient elution if necessary.
Reference solution (d). Dissolve 20 mg of a-apo-
oxytetracycline RS in 20 ml of 0.01 M sodium hydroxide and Inject reference solution (a) six times. The test is not valid if
dilute to 250 ml with 0.01 M hydrochloric acid. the relative standard deviation of the area of the peak due to

878
IP 2007 OXYTETRAXYCLINE CAPSULES

oxytetracycline is greater than 1.0 per cent. If necessary, adjust Identification


the integrator parameters.
A. Determine by thin-layer chromatography (2.4.17), coating
Inject the test solution and reference solution (g). In the the plate with the substance prepared by mixing 25 g of silica
chromatogram obtained with the test solution the area of any gel G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of
peak corresponding to 4-epioxytetracycline or tetracycline 0.1 M disodium edetate previously adjusted to pH 7.0 with
is not greater than the area of the corresponding peak in the dilute ammonia solution. After spreading the plate, allow it to
chromatogram obtained with reference solution (g) and the stand at room temperature till it is dry (70 to 90 minutes).
total area of the peaks corresponding to a -apo-oxytetracycline
and to b -apo-oxytetracycline and any peak between the latter Mobile phase. The lower layer formed after shaking 200 ml of
two is not greater than the area of the peak due to b -apo- a mixture of 2 volumes of ethyl acetate, 2 volumes of
oxytetracycline in the chromatogram obtained with reference chloroform and 1 volume of acetone with 25 ml of 0.1 M
solution (g). In the chromatogram obtained with the test disodium edetate previously adjusted to pH 7.0 with dilute
solution the area of any peak appearing on the tail of the ammonia solution.
principal peak is not greater than 4.0 times that of the peak due Test solution. Extract a quantity of the contents of the capsules
to 4-epioxytetracycline in the chromatogram obtained with containing 10 mg of Oxytetracycline Hydrochloride with 20 ml
reference solution (g). methanol, centrifuge and use the supernatant liquid.
Heavy metals (2.3.13). 0.4 g complies with the limit test for Reference solution (a). A 0.05 per cent w/v solution of
heavy metals, Method B (50 ppm). oxytetracycline hydrochloride RS in methanol.
Sulphated ash (2.3.18). Not more than 0.5 per cent.
Reference solution (b). A solution containing 0.05 per cent
Water (2.3.43). Not more than 2.0 per cent w/w, determined on w/v each of demethylchlortetracycline hydrochloride RS,
0.5 g. oxytetracycline hydrochloride RS and tetracycline
Assay. Determine by the microbiological assay of antibiotics, hydrochloride RS in methanol.
Method A or B (2.2.10), and express the result in µg of Apply to the plate 1 µl of each solution, freshly prepared.
oxytetracycline, C22H24N2O9, per mg. After development, dry the plate in air, expose to the vapours
Oxytetracycline Hydrochloride intended for use in the of ammonia and examine in ultraviolet light at 365 nm. The
manufacture of parenteral preparations without a further principal spot in the chromatogram obtained with the test
procedure for the removal of bacterial endotoxins complies solution corresponds to that in the chromatogram obtained
with the following additional requirement. with reference solution (a). The test is not valid unless the
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit chromatogram obtained with reference solution (b) shows
per mg. three clearly separated spots.

Oxytetracycline Hydrochloride intended for use in the B. To 0.5 mg of the contents of the capsules add 2 ml of
manufacture of parenteral preparations without a further sulphuric acid; a red colour is produced. Add the solution to
sterilisation procedure complies with the following 1 ml of water; the colour changes to yellow.
additional requirement. C. Dissolve about 2 mg of the contents of the capsules in 5 ml
Sterility. Complies with test for sterility (2.2.11). of a 1 per cent w/v solution of sodium carbonate and add 2 ml
of diazotised sulphanilic acid solution; a light brown colour
Storage. Store protected from light and moisture. If it is
is produced.
intended for use in the manufacture of parenteral preparations,
the container should be sterile and sealed so as to exclude D. The contents of the capsules give the reactions of chlorides
micro-organisms. (2.3.1).
Labelling. The label states whether or not the contents are
intended for use in the manufacture of parenteral preparations. Tests
Light-absorbing impurities. Dissolve a portion of the mixed
contents of five capsules as completely as possible in
Oxytetracycline Capsules sufficient of a mixture of 1 volume of 1 M hydrochloric acid
and 99 volumes of methanol to produce two solutions of
Oxytetracycline Hydrochloride Capsules Oxytetracycline Hydrochloride containing (1) 0.2 per cent w/v
Oxytetracycline Capsules contain not less than 95.0 per cent and (2) 1.0 per cent w/v and filter each solution. Absorbance
and not more than 110.0 per cent of the stated amount of of the filtrate obtained from solution (1) at about 430 nm, when
oxytetracycline hydrochloride, C22H24N2O9,HCl. measured within 1 hour of preparing the solution, not greater

879
OXYTETRAXYCLINE EYE OINTMENT IP 2007

than 0.75 and of the filtrate obtained from solution (2) at about Test solution. A solution prepared by heating a quantity
490 nm, not more than 0.40 (2.4.7). containing 20 mg of Oxytetracycline Hydrochloride with 20 ml
Dissolution (2.5.2). of methanol for 20 minutes, cooling in ice, filtering, carefully
evaporating the filtrate to dryness and dissolving the residue
Apparatus. No 1 in 20 ml of methanol.
Medium. 900 ml of 0.1 M hydrochloric acid.
Reference solution (a). A 0.05 per cent w/v solution of
Speed and time. 100 rpm and 45 minutes. oxytetracycline hydrochloride RS in methanol.
Withdraw a suitable volume of the medium and filter through
Reference solution (b). A solution containing 0.05 per cent
a membrane filter disc with an average pore diameter not greater
w/v each of demethylchlortetracycline hydrochloride RS,
than 1.0 µm. Measure the absorbance of the filtrate, suitably
oxytetracycline hydrochloride RS and tetracycline
diluted if necessary, at the maximum at about 353 nm (2.4.7).
hydrochloride RS in methanol.
Calculate the content of C22H24N2O9,HCl in the medium taking
282 as the specific absorbance at 353 nm. Apply to the plate 1 µl of each solution, freshly prepared.
After development, dry the plate in air, expose to the vapours
D. Not less than 75 per cent of the stated amount of
of ammonia and examine in ultraviolet light at 365 nm. The
C22H24N2O9,HCl.
principal spot in the chromatogram obtained with the test
Loss on drying (2.4.19). Not more than 5.0 per cent, determined solution corresponds to that in the chromatogram obtained
on 1.0 g of the mixed contents of the capsules by drying in an with reference solution (a). The test is not valid unless the
oven at 60º at a pressure not exceeding 0.7 kPa for 3 hours. chromatogram obtained with reference solution (b) shows
Other Tests. Comply with the tests stated under Capsules. three clearly separated spots.

Assay. Weigh accurately a quantity of the mixed contents of Tests


20 capsules containing about 0.25 g of Oxytetracycline
Hydrochloride, add 250.0 ml of water, shake, filter. Water (2.3.43). Not more than 1.0 per cent, determined on 0.5 g.

Determine by the microbiological assay of antibiotics, Method Other Tests. Complies with the tests stated under Eye
A or B (2.2.10), and express the results in mg of oxytetracycline Ointments.
hydrochloride per capsule taking each mg of oxytetracycline Assay. Weigh accurately about 1.0 g and transfer to a
to be equivalent to 1.079 mg of oxytetracycline hydrochloride, separating funnel. Add 25 ml of peroxide-free ether, shake
C22H24N2O9,HCl. well and extract with five quantities, each of 20 ml, of 0.1 M
Storage. Store protected from light and moisture. hydrochloric acid. Combine the extracts and dilute to
200.0 ml with 0.1 M hydrochloric acid. Dilute a suitable volume
of the resulting solution with buffer solution No 3 (2.2.10), to
produce a solution containing 1 µg of oxytetracycline per ml.
Oxytetracycline Eye Ointment
Determine by the microbiological assay of antibiotics, Method
Oxytetracycline Hydrochloride Eye Ointment B (2.2.10), and express the results as a percentage of
Oxytetracycline Eye Ointment contains not less than 90.0 per oxytetracycline hydrochloride taking each mg of
cent and not more than 115.0 per cent of the stated amount of oxytetracycline to be equivalent to 1.079 mg of oxytetracycline
oxytetracycline hydrochloride, C22H24N2O9,HCl. hydrochloride, C22H24N2O9,HCl.
Storage. Store protected from light and moisture.
Identification
Determine by thin-layer chromatography (2.4.17), coating the
plate with the substance prepared by mixing 25 g of silica gel Oxytetracycline Hydrochloride
G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of
0.1 M disodium edetate previously adjusted to pH 7.0 with
Injection
dilute ammonia solution. After spreading the plate, allow it to Oxytetracycline Hydrochloride Injection is a sterile material
stand at room temperature till it is dry (70 to 90 minutes). consisting of Oxytetracycline Hydrochloride with or without
Mobile phase. The lower layer formed after shaking 200 ml of buffering agents and other excipients. It is filled in a sealed
a mixture of 2 volumes of ethyl acetate, 2 volumes of container.
chloroform and 1 volume of acetone with 25 ml of 0.1 M The injection is constituted by dissolving the contents of the
disodium edetate previously adjusted to pH 7.0 with dilute sealed container in the requisite amount of sterile Water for
ammonia solution. Injections, immediately before use.

880
IP 2007 OXYTOCIN

The constituted solution complies with the requirements for Tests


Clarity of solution and Particulate matter stated under
Parenteral Preparations (Injections). Appearance of solution. A 10.0 per cent w/v solution is clear
(2.4.1) and yellow.
Storage. The constituted solution may be used within three
days of preparation when stored in a refrigerator (2º to 8º). pH (2.4.24). 2.0 to 3.0, determined in a 1.0 per cent w/v solution.

Oxytetracycline Hydrochloride Injection contains not less than Light-absorbing impurities. A. Dissolve 20 mg in sufficient
90.0 per cent and not more than 115.0 per cent of the stated of a mixture of 1 volume of 1 M hydrochloric acid and
amount of oxytetracycline, C22H24N2O9. 99 volumes of methanol to produce 10 ml. Absorbance of the
resulting solution at about 430 nm, when measured within
The contents of the sealed container comply with the 1 hour of preparing the solution, not more than 0.50, calculated
requirements stated under Parenteral Preparations on the anhydrous basis (2.4.7).
(Powders for Injection) and with the following requirements.
B. Dissolve 0.1 g in sufficient of a mixture of 1 volume of 1 M
Description. A pale yellow, crystalline powder. hydrochloric acid and 99 volumes of methanol to produce
10 ml. Absorbance of the resulting solution at about 490 nm,
Identification when measured within 1 hour of preparing the solution, not
A. Determine by thin-layer chromatography (2.4.17), coating more than 0.20, calculated on the anhydrous basis (2.4.7).
the plate with the substance prepared by mixing 25 g of silica Assay. Determine by the microbiological assay of antibiotics,
gel G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of method A or B (2.2.10), and express the result in µg of
0.1 M disodium edetate previously adjusted to pH 7.0 with oxytetracycline, C22H24N2O9, per mg.
dilute ammonia solution. After spreading the plate, allow it to
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit
stand at room temperature till it is dry (70 to 90 minutes).
per mg.
Mobile phase. The lower layer formed after shaking 200 ml of
Storage. Store protected from light and moisture.
a mixture of 2 volumes of ethyl acetate, 2 volumes of
chloroform and 1 volume of acetone with 25 ml of 0.1 M Labelling. The label states (1) the quantity of Oxytetracycline
disodium edetate previously adjusted to pH 7.0 with dilute Hydrochloride contained in it in terms of the equivalent
ammonia solution. amount of oxytetracycline; (2) that the contents are to be
used for intravenous injection only; (3) the names of the
Test solution. Dissolve 0.05 g of the substance under buffering agents used.
examination in 100 ml of methanol.
Reference solution (a). A 0.05 per cent w/v solution of
oxytetracycline hydrochloride RS in methanol. Oxytocin
Reference solution (b). A solution containing 0.05 per cent
Cys-Tyr-Ile-Gln-Asn-Cys-Pro-Leu-GlyNH2
w/v each of demethylchlortetracycline hydrochloride RS,
oxytetracycline hydrochloride RS and tetracycline
hydrochloride RS in methanol.
C43H66N12O12S2 Mol. Wt.1007.2
Apply to the plate 1 µl of each solution, freshly prepared.
Oxytocin is a cyclic nonapeptide hormone obtained by a
After development, dry the plate in air, expose to the vapours
process of fractionation from the posterior lobe of the pituitary
of ammonia and examine in ultraviolet light at 365 nm. The
gland of healthy oxen or other mammals or by synthesis that
principal spot in the chromatogram obtained with the test
has the property of stimulating contraction of the uterus and
solution corresponds to that in the chromatogram obtained
milk ejection in receptive animals. It may be presented as a
with reference solution (a). The test is not valid unless the
solid or as a solution in a solvent containing an appropriate
chromatogram obtained with reference solution (b) shows
antimicrobial preservative such as 0.2 per cent w/v solution of
three clearly separated spots.
chlorbutol.
B. To about 0.5 mg add 2 ml of sulphuric acid; a red colour is
If it is derived from animal species, Oxytocin contains not less
produced. Add the solution to 1 ml of water; the colour
than 90.0 per cent and not more than 111.0 per cent of the
changes to yellow.
stated number of Units of oxytocic activity. If it is a synthetic
C. Dissolve about 2 mg in 5 ml of a 1 per cent w/v solution of product presented as a solid, it contains not less than
sodium carbonate and add 2 ml of diazotised sulphanilic 560 Units per mg, calculated with reference to the peptide
acid solution; an orange-red to brownish-red colour is content and when presented as a liquid, it contains not less
produced. than 150 Units per ml.

881
OXYTOCIN IP 2007

Description. When presented as a solid, a white or almost Method A. By depression of the blood pressure in chicken —
white powder. When presented as a liquid, a clear colourless Anaesthetise a young healthy adult cockerel weighing 1.2 to
liquid. 2.3 kg with an anaesthetic that will maintain a prolonged and
constant high blood pressure. Expose the gluteus primus
Identification muscle in one thigh and cut and retract it to reveal the popliteal
Gives rise to an appropriate response when administered as artery and crural vein. Cannulate the popliteal artery and record
directed under one of the methods described for Assay. the blood pressure on a suitable recorder calibrated for use
over a linear range. Cannulate the crural or brachial vein.
Tests
Immediately before use prepare a solution of the Standard
Peptide. 90.0 to 110.0 per cent of the stated amount of oxytocin, Preparation in saline solution so that the volume to be injected
C43H66N12O12S2 expressed per mg for the solid, and in mg per is between 0.1 ml and 0.5 ml. Record the blood pressure
ml for the liquid, responses to the injection into the cannulated vein of two
Determine by liquid chromatography (2.4.14). doses of this solution; the doses should be such as to produce
clearly discriminated, precipitous, submaximal decreases in
Test solution. Dissolve 3.5 mg of the substance under blood pressure; the required doses normally lie between
examination in sufficient of a 1.56 per cent w/v solution of 20 and 100 milliUnits. The interval between injections should
sodium dihydrogen phosphate to produce 10.0 ml or use the be constant and lie between 3 and 10 minutes depending on
liquid preparation as appropriate. the rate at which the blood pressure returns to normal.
Reference solution. Dissolve 3.5 mg of oxytocin RS in sufficient Immediately before use dilute the preparation being examined
of a 1.56 per cent w/v solution of sodium dihydrogen with saline solution so as to obtain responses similar to those
phosphate to produce 10.0 ml. obtained with the Standard Preparation. The ratio between
Chromatographic system the two doses of the preparation under examination should be
– a stainless steel column 12 cm x 4.6 mm, packed with the same as that between the two doses of the Standard
octadecylsilane bonded to porous silica (5 µm), Preparation and this ratio should be kept constant throughout
– mobile phase: appropriate proportions of a 1.56 per cent the assay.
w/v solution of sodium dihydrogen phosphate (mobile The two doses of the Standard Preparation and the two doses
phase A) and a mixture of equal volumes of acetonitrile of the preparation under examination should be given
and water (mobile phase B), according to a randomised block or a Latin square design and
– flow rate. 1 ml per minute, at least six responses to each should be recorded.
– spectrophotometer set at 220 nm,
If the animal rapidly becomes insensitive to the repeated
– a 20 µl loop injector.
injections of the solutions another animal must be used.
Equilibrate the column with a mixture of 70 volumes of mobile Measure all the responses and calculate the result of the assay
phase A and 30 volumes of mobile phase B and record the by standard statistical methods.
chromatograms as follows. Operate by gradient elution
increasing continuously and linearly the proportion of mobile Method B. By contraction of the rat uterus — Inject 100 µg of
phase B by 1.0 per cent v/v per minute for 30 minutes. Finally oestradiol benzoate intramuscularly into a female rat weighing
elute using the same mixture for 15 minutes to re-equilibrate 120 to 200 g 18 to 24 hours before the assay. Immediately
the column. before the assay confirm by vaginal smear that the rat is in
oestrus or preoestrus. Kill the rat and suspend one horn of
Calculate the content of the peptide, C43H66N12O12S2. the uterus in a bath containing a solution of the following
Assay.The potency of oxytocin is determined by comparing composition.
its activity with that of the Standard Preparation of oxytocin Composition (per cent w/v)
under the conditions of a suitable method of assay.
Sodium chloride 0.662
Standard Preparation Potassium chloride 0.045
The Standard Preparation is the 4th International Standard for Calcium chloride 0.007
Oxytocin, established in 1978, consisting of freeze-dried Sodium bicarbonate 0.256
synthetic oxytocin peptide with human albumin and citric acid
Disodium hydrogen phosphate 0.029
(supplied in ampoules containing 12.5 Units), or any other
suitable preparation the potency of which has been determined Sodium dihydrogen phosphate 0.003
in relation to the International Standard. Magnesium chloride 0.010
NOTE — Any of the following methods may be followed. Dextrose 0.050

882
IP 2007 OXYTOCIN

Maintain the bath at a temperature of 32º or at some other the whole system with a 3.8 per cent w/v solution of sodium
suitable temperature at which spontaneous contractions of citrate or saline solution containing 50 Units of heparin
the uterus are abolished and the preparation maintains its sodium per ml to prevent clotting of milk. After cannulation,
sensitivity. Oxygenate the solution with a mixture of 95 per inject a small volume (0.05 to 0.2 ml) of this solution into the
cent of oxygen and 5 per cent of carbon dioxide and record teat duct through the transducer to clear the milk from the tip
the contractions of the muscle using a suitable instrument of the cannula. (This procedure may be repeated during the
giving a linear response (for example an isotonic lever with a assay should obstruction arise from milk ejected into the
load not exceeding 2 g). Record the contractions produced by cannula). Clamp the strain gauge so that a slight tension is
the addition to the bath of two doses of the Standard applied to the teat and its natural alignment is preserved and
Preparation suitably diluted with the above solution. The doses connect the gauge to a potentiometric recorder adjusted to
should be such as to produce clearly discriminated, give full-scale deflection for an increase in milk-ejection
submaximal contractions; the required doses normally lie pressure of about 5.3 kPa. Inject all solutions through the
between 10 and 50 micro Units per ml of bath liquid. When venous cannula using a 1-ml syringe graduated in 0.01 ml and
maximal contraction has been reached, replace the bath liquid wash them in with 0.2 ml of saline solution.
by a fresh solution. The doses should be added at regular Prepare a solution of the Standard Preparation and a solution
intervals of 3 to 5 minutes depending upon the rate of recovery of the preparation under examination in saline solution so
of the muscle. Dilute the preparation under examination so as that the volume to be injected is between 0.1 ml and 0.4 ml.
to obtain responses on the addition of two doses similar to Choose two doses of the Standard Preparation such that the
those obtained with the Standard Preparation. The ratio increase in milk-ejection pressure is about 1.35 kPa for the
between the two doses of the preparation under examination lower dose and about 2.7 kPa for the higher dose. As an initial
should be the same as that between the two doses of the approximation, a lower dose of between 0.1 and 0.4 milliUnit
Standard Preparation and this ratio should be kept constant and an upper dose of 1.5 to 2 times this amount may be tried.
throughout the assay. Choose two doses of the preparation under examination with
The two doses of Standard Preparation and the two doses of the same inter-dose ratio, matching the effects of the doses of
the preparation under examination should be given according the Standard Preparation as closely as possible. Inject the
to a randomised block or a Latin square design and at least six four doses (two doses of the Standard Preparation and two
responses to each should be recorded. doses of the preparation being examined) at intervals of 3 to
5 minutes. The two doses of Standard Preparation and the
Measure all the responses and calculate the result of the assay two doses of the preparation under examination should be
by standard statistical methods. given according to a randomised block or a Latin square design
Method C. By measurement of milk-ejection pressure in a and at least four responses to each should be recorded.
lactating rat— Select a lactating rat, in the third to twenty- Measure all the responses and calculate the result of the assay
first day after parturition and weighing about 300 g, separate by standard statistical methods.
it from the litter and 30 to 60 minutes later anaesthetise (for
The estimated potency is not less than 90 per cent and not
example, by the intraperitoneal injection of a solution of
more than 111 per cent of the stated potency. The fiducial
Pentobarbitone Sodium). Tie the rat to an operating table,
limits of error are not less than 80 per cent and not more than
maintained at 37º, by its hind legs leaving the front legs free.
125 per cent of the stated potency.
Cannulate the trachea with a short polyethylene tube of
internal diameter about 2.5 mm in such a manner so as to Oxytocin of natural origin obtained by extraction and
ensure a free airway; apply artificial respiration only if purification complies with the following additional
necessary. Cannulate an external jugular or femoral vein with requirement.
a polyethylene tube of internal diameter about 0.4 mm which Vasopressor activity. Not more than 0.5 Unit per 20 Units of
is filled with saline solution and closed with a pin. oxytocic activity, when assayed by the biological assay for
Shave the skin surrounding the inguinal and abdominal teats vasopressor activity described below.
and excise the tip of one teat, preferably the lower inguinal The vasopressor activity is estimated by comparing the activity
teat. Insert a polyethylene tube of internal diameter about 0.3 of the preparation under examination with that of the Standard
mm and external diameter about 0.6 mm, to a depth sufficient Preparation of arginine vasopressin under the conditions of a
to obtain appropriate measurement of pressure (3 to 10 mm suitable method of assay.
depth), into the primary teat duct which opens onto the cut
surface and tie firmly in place with a ligature. Connect this Standard Preparation
cannula with a suitable strain gauge transducer (such as that The Standard Preparation is the Ist International Standard for
used for recording arterial blood pressure in the rat) and fill Arginine vasopressin, established in 1978, consisting of

883
OXYTOCIN INJECTION IP 2007

freeze-dried synthetic arginine vasopressin peptide acetate ratio, matching the effects of the dose of the Standard
with human albumin and citric acid (supplied in ampoules Preparation as closely as possible. Inject doses at intervals of
containing 8.20 Units), or another suitable preparation the 10 to 15 minutes.
potency of which has been determined in relation to that of The two doses of the Standard Preparation and the two doses
the International Standard. of the preparation under examination should be given in a
Inject slowly into the tail vein of a male albino rat weighing randomised block or a Latin square design and four to five
about 300 g a solution of a suitable a-adrenoceptor blocking responses to each should be recorded.
agent, for example 10 ml per kg of body weight of a solution Measure all the responses and calculate the result of the assay
prepared by dissolving 5 mg of phenoxybenzamine by standard statistical methods.
hydrochloride in 0.1 ml of ethanol (95 per cent), adding
0.05 ml of 1 M hydrochloric acid and diluting to 5 ml with Oxytocin intended for use in the manufacture of parenteral
saline solution. After 18 hours, anaesthetise the rat with an preparations without a further procedure for the removal of
anaesthetic that will maintain a prolonged and uniform blood bacterial endotoxins.complies with the following additional
pressure. After 45 to 60 minutes, tie the rat on its back to the requirement.
operating table by its hind legs. Cannulate the trachea with a Bacterial endotoxins (2.2.3). Not more than 100 Endotoxin
short polyethylene tube of external diameter about 2.5 mm Units per 200 Units of oxytocin.
and dissect a carotid artery ready for cannulation. Then
Oxytocin intended for use in the manufacture of parenteral
cannulate the femoral vein close to the inguinal ligament.
preparations without a further sterilisation procedure
Retract the abdominal muscles to expose the inguinal ligament.
complies with the following additional requirement.
Retract the superficial pudendal vein to one side and dissect
the femoral vein towards the inguinal ligament from the Sterility (2.2.11). Complies with the test for sterility.
corresponding artery. When dissecting, a deep branch Storage. Store protected from moisture. If the substance is
reaching the femoral vein must be found and tied off to prevent intended for use in the manufacture of parenteral preparations,
bleeding during cannulation. Tie a short polyethylene cannula the container should be sterile, tamper-evident and sealed so
of external diameter about 1 mm into the femoral vein by two as to exclude micro-organisms.
ligatures and join by a short piece of flexible tubing to a 1-ml
burette with an attached thistle funnel containing saline Labelling. The label states (1) the number of Units of oxytocic
solution at about 37º. Firmly fix a wet absorbent cotton swab activity per mg (for solid) or per ml (for liquid); (2) either the
to the thigh so as to cover the incision and cannula. At this animal species from which it is obtained or whether it is
stage inject through the venous cannula 200 Units of heparin, synthetic, as appropriate; (3) whether or not the contents are
dissolved in saline solution, per 100 g of body weight. Then intended for use in the manufacture of parenteral preparations.
tie in a carotid cannula of external diameter about 1 mm and
connect by a column of saline solution containing heparin
with a suitable pressure measuring device such as a mercury
manometer of internal diameter about 2 to 3 mm. Oxytocin Injection
The central and peripheral nervous system including both Oxytocin Injection is a sterile solution of Oxytocin in Water
vagus and associated sympathetic nerves is left intact. No for Injections.
artificial respiration is necessary. Taking care that no air is Oxytocin Injection contains not less than 90.0 per cent and
injected, inject all solutions through the venous cannula by not more than 110.0 per cent of the stated number of Units of
means of a 1-ml syringe graduated in 0.01 ml and wash in with oxytocin activity.
0.2 ml of saline solution from the burette.
Description. A clear, colourless liquid.
Dilute the extract of the Standard Preparation and the
preparation under examination with saline solution so that Identification
the volume to be injected is between 0.1 ml and 0.5 ml.
Gives rise to an appropriate response when administered as
Choose two doses of the Standard Preparation such that the directed under one of the methods described for Assay.
elevation of the blood pressure is about 4 kPa for the lower
dose and about 7 kPa but always submaximal for the higher Tests
dose, the ratio of low to high dose being determined by the
pH (2.4.24). 3.5 to 4.5.
response and usually being 3 to 5. As an initial approximation
doses of 3 and 5 milliUnits may be tried. Choose two doses of Other Tests. Complies with the tests stated under Parenteral
the preparation under examination with the same inter-dose Preparations (Injections).

884
IP 2007 OXYTOCIN INJECTION

Bacterial endotoxins (2.2.3). Not more than 100 Endotoxin oestrus or preoestrus. Kill the rat and suspend one horn of
Units per 200 Units of oxytocin. the uterus in a bath containing a solution of the following
Assay.The potency of oxytocin is determined by comparing composition.
its activity with that of the Standard Preparation of oxytocin Composition (per cent w/v)
under the conditions of a suitable method of assay. Sodium chloride 0.662
Standard Preparation Potassium chloride 0.045
Calcium chloride 0.007
The Standard Preparation is the 4th International Standard for
Oxytocin, established in 1978, consisting of freeze-dried Sodium bicarbonate 0.256
synthetic oxytocin peptide with human albumin and citric acid Disodium hydrogen phosphate 0.029
(supplied in ampoules containing 12.5 Units), or any other Sodium dihydrogen phosphate 0.003
suitable preparation the potency of which has been determined Magnesium chloride 0.010
in relation to the International Standard.
Dextrose 0.050
NOTE — Any of the following methods may be followed.
Maintain the bath at a temperature of 32º or at some other
Method A. By depression of the blood pressure in chicken — suitable temperature at which spontaneous contractions of
Anaesthetise a young healthy adult cockerel weighing 1.2 to the uterus are abolished and the preparation maintains its
2.3 kg with an anaesthetic that will maintain a prolonged and sensitivity. Oxygenate the solution with a mixture of 95 per
constant high blood pressure. Expose the gluteus primus cent of oxygen and 5 per cent of carbon dioxide and record
muscle in one thigh and cut and retract it to reveal the popliteal the contractions of the muscle using a suitable instrument
artery and crural vein. Cannulate the popliteal artery and record giving a linear response (for example an isotonic lever with a
the blood pressure on a suitable recorder calibrated for use load not exceeding 2 g). Record the contractions produced by
over a linear range. Cannulate the crural or brachial vein. the addition to the bath of two doses of the Standard
Immediately before use prepare a solution of the Standard Preparation suitably diluted with the above solution. The doses
Preparation in saline solution so that the volume to be injected should be such as to produce clearly discriminated,
is between 0.1 ml and 0.5 ml. Record the blood pressure submaximal contractions; the required doses normally lie
responses to the injection into the cannulated vein of two between 10 and 50 micro Units per ml of bath liquid. When
doses of this solution; the doses should be such as to produce maximal contraction has been reached, replace the bath liquid
clearly discriminated, precipitous, submaximal decreases in by a fresh solution. The doses should be added at regular
blood pressure; the required doses normally lie between 20 intervals of 3 to 5 minutes depending upon the rate of recovery
and 100 milliUnits. The interval between injections should be of the muscle. Dilute the preparation being examined so as to
constant and lie between 3 and 10 minutes depending on the obtain responses on the addition of two doses similar to those
rate at which the blood pressure returns to normal. Immediately obtained with the Standard Preparation. The ratio between
before use dilute the preparation being examined with saline the two doses of the preparation under examination should be
solution so as to obtain responses similar to those obtained the same as that between the two doses of the Standard
with the Standard Preparation. The ratio between the two doses Preparation and this ratio should be kept constant throughout
of the preparation under examination should be the same as the assay.
that between the two doses of the Standard Preparation and The two doses of Standard Preparation and the two doses of
this ratio should be kept constant throughout the assay. the preparation under examination should be given according
The two doses of the Standard Preparation and the two doses to a randomised block or a Latin square design and at least six
of the preparation under examination should be given responses to each should be recorded.
according to a randomised block or a Latin square design and Measure all the responses and calculate the result of the assay
at least six responses to each should be recorded. by standard statistical methods.
If the animal rapidly becomes insensitive to the repeated Method C. By measurement of milk-ejection pressure in a
injections of the solutions another animal must be used. lactating rat — Select a lactating rat, in the third to twenty-
Measure all the responses and calculate the result of the assay first day after parturition and weighing about 300 g, separate
by standard statistical methods. it from the litter and 30 to 60 minutes later anaesthetise (for
Method B. By contraction of the rat uterus — Inject 100 µg of example, by the intraperitoneal injection of a solution of
oestradiol benzoate intramuscularly into a female rat weighing Pentobarbitone Sodium). Tie the rat to an operating table,
120 to 200 g 18 to 24 hours before the assay. Immediately maintained at 37º, by its hind legs leaving the front legs free.
before the assay confirm by vaginal smear that the rat is in Cannulate the trachea with a short polyethylene tube of

885
OXYTOCIN INJACTION IP 2007

internal diameter about 2.5 mm in such a manner so as to Oxytocin Injection containing Oxytocin of natural origin
ensure a free airway; apply artificial respiration only if obtained by extraction and purification complies with the
necessary. Cannulate an external jugular or femoral vein with following additional requirement.
a polyethylene tube of internal diameter about 0.4 mm which Vasopressor activity. Not more than 0.5 Unit per 20 Units of
is filled with saline solution and closed with a pin. oxytocic activity, when assayed by the biological assay for
Shave the skin surrounding the inguinal and abdominal teats vasopressor activity described below.
and excise the tip of one teat, preferably the lower inguinal The vasopressor activity is estimated by comparing the activity
teat. Insert a polyethylene tube of internal diameter about of the preparation under examination with that of the Standard
0.3 mm and external diameter about 0.6 mm, to a depth sufficient Preparation of arginine vasopressin under the conditions of a
to obtain appropriate measurement of pressure (3 to 10 mm suitable method of assay.
depth), into the primary teat duct which opens onto the cut
surface and tie firmly in place with a ligature. Connect this Standard Preparation
cannula with a suitable strain gauge transducer (such as that
used for recording arterial blood pressure in the rat) and fill The Standard Preparation is the Ist International Standard for
the whole system with a 3.8 per cent w/v solution of sodium Arginine vasopressin, established in 1978, consisting of
citrate or saline solution containing 50 Units of heparin freeze-dried synthetic arginine vasopressin peptide acetate
sodium per ml to prevent clotting of milk. After cannulation, with human albumin and citric acid (supplied in ampoules
inject a small volume (0.05 to 0.2 ml) of this solution into the containing 8.20 Units), or any other suitable preparation the
teat duct through the transducer to clear the milk from the tip potency of which has been determined in relation to that of
of the cannula. (This procedure may be repeated during the the International Standard.
assay should obstruction arise from milk ejected into the Inject slowly into the tail vein of a male albino rat weighing
cannula). Clamp the strain gauge so that a slight tension is about 300 g a solution of a suitable a-adrenoceptor blocking
applied to the teat and its natural alignment is preserved and agent, for example 10 ml per kg of body weight of a solution
connect the gauge to a potentiometric recorder adjusted to prepared by dissolving 5 mg of phenoxybenzamine
give full-scale deflection for an increase in milk-ejection hydrochloride in 0.1 ml of ethanol (95 per cent), adding
pressure of about 5.3 kPa. Inject all solutions through the 0.05 ml of 1 M hydrochloric acid and diluting to 5 ml with
venous cannula using a 1-ml syringe graduated in 0.01 ml and saline solution. After 18 hours, anaesthetise the rat with an
wash them in with 0.2 ml of saline solution. anaesthetic that will maintain a prolonged and uniform blood
Prepare a solution of the Standard Preparation and a solution pressure. After 45 to 60 minutes, tie the rat on its back to the
of the preparation under examination in saline solution so operating table by its hind legs. Cannulate the trachea with a
that the volume to be injected is between 0.1 ml and 0.4 ml. short polyethylene tube of external diameter about 2.5 mm
Choose two doses of the Standard Preparation such that the and dissect a carotid artery ready for cannulation. Then
increase in milk-ejection pressure is about 1.35 kPa for the cannulate the femoral vein close to the inguinal ligament.
lower dose and about 2.7 kPa for the higher dose. As an initial Retract the abdominal muscles to expose the inguinal ligament.
approximation, a lower dose of between 0.1 and 0.4 milliUnit Retract the superficial pudendal vein to one side and dissect
and an upper dose of 1.5 to 2 times this amount may be tried. the femoral vein towards the inguinal ligament from the
Choose two doses of the preparation under examination with corresponding artery. When dissecting, a deep branch
the same inter-dose ratio, matching the effects of the doses of reaching the femoral vein must be found and tied off to prevent
the Standard Preparation as closely as possible. Inject the bleeding during cannulation. Tie a short polyethylene cannula
four doses (two doses of the Standard Preparation and two of external diameter about 1 mm into the femoral vein by two
doses of the preparation under examination at intervals of 3 to ligatures and join by a short piece of flexible tubing to a 1-ml
5 minutes. The two doses of Standard Preparation and the burette with an attached thistle funnel containing saline
two doses of the preparation under examination should be solution at about 37º. Firmly fix a wet absorbent cotton swab
given according to a randomised block or a Latin square design to the thigh so as to cover the incision and cannula. At this
and at least four responses to each should be recorded. stage inject through the venous cannula 200 Units of heparin,
dissolved in saline solution, per 100 g of body weight. Then
Measure all the responses and calculate the result of the assay tie in a carotid cannula of external diameter about 1 mm and
by standard statistical methods. connect by a column of saline solution containing heparin
The estimated potency is not less than 90 per cent and not with a suitable pressure measuring device such as a mercury
more than 111 per cent of the stated potency. The fiducial manometer of internal diameter about 2 to 3 mm.
limits of error are not less than 80 per cent and not more than The central and peripheral nervous system including both
125 per cent of the stated potency. vagus and associated sympathetic nerves is left intact. No

886
IP 2007 OXYTOCIN NASAL SOLUTION

artificial respiration is necessary. Taking care that no air is synthetic oxytocin peptide with human albumin and citric acid
injected, inject all solutions through the venous cannula by (supplied in ampoules containing 12.5 Units), or any other
means of a 1-ml syringe graduated in 0.01 ml and wash in with suitable preparation the potency of which has been determined
0.2 ml of saline solution from the burette. in relation to the International Standard.
Dilute the extract of the Standard Preparation and the NOTE — Any of the following methods may be followed.
preparation under examination with saline solution so that
Method A. By depression of the blood pressure in chicken —
the volume to be injected is between 0.1 ml and 0.5 ml.
Anaesthetise a young healthy adult cockerel weighing 1.2 to
Choose two doses of the Standard Preparation such that the 2.3 kg with an anaesthetic that will maintain a prolonged and
elevation of the blood pressure is about 4 kPa for the lower constant high blood pressure. Expose the gluteus primus
dose and about 7 kPa but always submaximal for the higher muscle in one thigh and cut and retract it to reveal the popliteal
dose, the ratio of low to high dose being determined by the artery and crural vein. Cannulate the popliteal artery and record
response and usually being 3 to 5. As an initial approximation the blood pressure on a suitable recorder calibrated for use
doses of 3 and 5 milliUnits may be tried. Choose two doses of over a linear range. Cannulate the crural or brachial vein.
the preparation under examination with the same inter-dose
Immediately before use prepare a solution of the Standard
ratio, matching the effects of the dose of the Standard
Preparation in saline solution so that the volume to be injected
Preparation as closely as possible. Inject doses at intervals of
is between 0.1 ml and 0.5 ml. Record the blood pressure
10 to 15 minutes.
responses to the injection into the cannulated vein of two
The two doses of the Standard Preparation and the two doses doses of this solution; the doses should be such as to produce
of the preparation under examination should be given in a clearly discriminated, precipitous, submaximal decreases in
randomised block or a Latin square design and four to five blood pressure; the required doses normally lie between 20
responses to each should be recorded. and 100 milliUnits. The interval between injections should be
Measure all the responses and calculate the result of the assay constant and lie between 3 and 10 minutes depending on the
by standard statistical methods. rate at which the blood pressure returns to normal. Immediately
before use dilute the preparation being examined with saline
Storage. Store at temperature not exceeding 30º. Do not freeze. solution so as to obtain responses similar to those obtained
Labelling. The label states (1) the number of Units of oxytocin with the Standard Preparation. The ratio between the two doses
activity per ml; (2) either the animal spieces from which it is of the preparation under examination should be the same as
obtained or whether it is synthetic, as appropriate. that between the two doses of the Standard Preparation and
this ratio should be kept constant throughout the assay.
The two doses of the Standard Preparation and the two doses
Oxytocin Nasal Solution of the preparation under examination should be given
according to a randomised block or a Latin square design and
Oxytocin Nasal Solution is a solution of Oxytocin in a suitable
at least six responses to each should be recorded.
solvent containing an appropriate antimicrobial preservative.
If the animal rapidly becomes insensitive to the repeated
Oxytocin Nasal Solution contains not less than 85.0 per cent
injections of the solutions another animal must be used.
and not more than 120.0 per cent of the stated number of Units
Measure all the responses and calculate the result of the assay
of oxytocic activity.
by standard statistical methods.
Description. A clear, colourless solution.
Method B. By contraction of the rat uterus — Inject 100 µg of
Tests oestradiol benzoate intramuscularly into a female rat weighing
120 to 200 g 18 to 24 hours before the assay. Immediately
pH (2.4.24). 3.5 to 4.5. before the assay confirm by vaginal smear that the rat is in
Other Tests. Complies with the tests stated under Nasal oestrus or preoestrus. Kill the rat and suspend one horn of
Preparations. the uterus in a bath containing a solution of the following
Assay.The potency of oxytocin is determined by comparing composition.
its activity with that of the Standard Preparation of oxytocin Composition (per cent w/v)
under the conditions of a suitable method of assay. Sodium chloride 0.662
Standard Preparation Potassium chloride 0.045
The Standard Preparation is the 4th International Standard for Calcium chloride 0.007
Oxytocin, established in 1978, consisting of freeze-dried Sodium bicarbonate 0.256

887
OXYTOCIN NASAL SOLUTION IP 2007

Disodium hydrogen phosphate 0.029 to obtain appropriate measurement of pressure (3 to 10 mm


Sodium dihydrogen phosphate 0.003 depth), into the primary teat duct which opens onto the cut
surface and tie firmly in place with a ligature. Connect this
Magnesium chloride 0.010
cannula with a suitable strain gauge transducer (such as that
Dextrose 0.050 used for recording arterial blood pressure in the rat) and fill
Maintain the bath at a temperature of 32º or at some other the whole system with a 3.8 per cent w/v solution of sodium
suitable temperature at which spontaneous contractions of citrate or saline solution containing 50 Units of heparin
the uterus are abolished and the preparation maintains its sodium per ml to prevent clotting of milk. After cannulation,
sensitivity. Oxygenate the solution with a mixture of 95 per inject a small volume (0.05 to 0.2 ml) of this solution into the
cent of oxygen and 5 per cent of carbon dioxide and record teat duct through the transducer to clear the milk from the tip
the contractions of the muscle using a suitable instrument of the cannula. (This procedure may be repeated during the
giving a linear response (for example an isotonic lever with a assay should obstruction arise from milk ejected into the
load not exceeding 2 g). Record the contractions produced by cannula). Clamp the strain gauge so that a slight tension is
the addition to the bath of two doses of the Standard applied to the teat and its natural alignment is preserved and
Preparation suitably diluted with the above solution. The doses connect the gauge to a potentiometric recorder adjusted to
should be such as to produce clearly discriminated, give full-scale deflection for an increase in milk-ejection
submaximal contractions; the required doses normally lie pressure of about 5.3 kPa. Inject all solutions through the
between 10 and 50 micro Units per ml of bath liquid. When venous cannula using a 1-ml syringe graduated in 0.01 ml and
maximal contraction has been reached, replace the bath liquid wash them in with 0.2 ml of saline solution.
by a fresh solution. The doses should be added at regular Prepare a solution of the Standard Preparation and a solution
intervals of 3 to 5 minutes depending upon the rate of recovery of the preparation under examination in saline solution so
of the muscle. Dilute the preparation being examined so as to that the volume to be injected is between 0.1 ml and 0.4 ml.
obtain responses on the addition of two doses similar to those Choose two doses of the Standard Preparation such that the
obtained with the Standard Preparation. The ratio between increase in milk-ejection pressure is about 1.35 kPa for the
the two doses of the preparation under examination should be lower dose and about 2.7 kPa for the higher dose. As an initial
the same as that between the two doses of the Standard approximation, a lower dose of between 0.1 and 0.4 milliUnit
Preparation and this ratio should be kept constant throughout and an upper dose of 1.5 to 2 times this amount may be tried.
the assay. Choose two doses of the preparation under examination with
The two doses of Standard Preparation and the two doses of the same inter-dose ratio, matching the effects of the doses of
the preparation under examination should be given according the Standard Preparation as closely as possible. Inject the
to a randomised block or a Latin square design and at least six four doses (two doses of the Standard Preparation and two
responses to each should be recorded. doses of the preparation under examination at intervals of 3 to
5 minutes. The two doses of Standard Preparation and the
Measure all the responses and calculate the result of the assay
two doses of the preparation under examination should be
by standard statistical methods.
given according to a randomised block or a Latin square design
Method C. By measurement of milk-ejection pressure in a and at least four responses to each should be recorded.
lactating rat — Select a lactating rat, in the third to twenty- Measure all the responses and calculate the result of the assay
first day after parturition and weighing about 300 g, separate by standard statistical methods.
it from the litter and 30 to 60 minutes later anaesthetise (for
example, by the intraperitoneal injection of a solution of The estimated potency is not less than 90 per cent and not
Pentobarbitone Sodium). Tie the rat to an operating table, more than 111 per cent of the stated potency. The fiducial
maintained at 37º, by its hind legs leaving the front legs free. limits of error are not less than 80 per cent and not more than
Cannulate the trachea with a short polyethylene tube of 125 per cent of the stated potency.
internal diameter about 2.5 mm in such a manner so as to Oxytocin Nasal Solution containing Oxytocin of natural
ensure a free airway; apply artificial respiration only if origin obtained by extraction and purification complies with
necessary. Cannulate an external jugular or femoral vein with the following additional requirement.
a polyethylene tube of internal diameter about 0.4 mm which
is filled with saline solution and closed with a pin. Vasopressor activity. Not more than 0.5 Unit per 20 Units of
oxytocic activity, when assayed by the biological assay for
Shave the skin surrounding the inguinal and abdominal teats
vasopressor activity described below.
and excise the tip of one teat, preferably the lower inguinal
teat. Insert a polyethylene tube of internal diameter about The vasopressor activity is estimated by comparing the activity
0.3 mm and external diameter about 0.6 mm, to a depth sufficient of the preparation under examination with that of the Standard

888
IP 2007 OXYTOCIN NASAL SOLUTION

Preparation of arginine vasopressin under the conditions of a tie in a carotid cannula of external diameter about 1 mm and
suitable method of assay. connect by a column of saline solution containing heparin
with a suitable pressure measuring device such as a mercury
Standard Preparation manometer of internal diameter about 2 to 3 mm.
The Standard Preparation is the Ist International Standard for The central and peripheral nervous system including both
Arginine vasopressin, established in 1978, consisting of vagus and associated sympathetic nerves is left intact. No
freeze-dried synthetic arginine vasopressin peptide acetate artificial respiration is necessary. Taking care that no air is
with human albumin and citric acid (supplied in ampoules injected, inject all solutions through the venous cannula by
containing 8.20 Units), or any other suitable preparation the means of a 1-ml syringe graduated in 0.01 ml and wash in with
potency of which has been determined in relation to that of 0.2 ml of saline solution from the burette.
the International Standard.
Dilute the extract of the Standard Preparation and the
Inject slowly into the tail vein of a male albino rat weighing preparation under examination with saline solution so that
about 300 g a solution of a suitable a-adrenoceptor blocking the volume to be injected is between 0.1 ml and 0.5 ml.
agent, for example 10 ml per kg of body weight of a solution
Choose two doses of the Standard Preparation such that the
prepared by dissolving 5 mg of phenoxybenzamine
elevation of the blood pressure is about 4 kPa for the lower
hydrochloride in 0.1 ml of ethanol (95 per cent), adding
dose and about 7 kPa but always submaximal for the higher
0.05 ml of 1 M hydrochloric acid and diluting to 5 ml with
dose, the ratio of low to high dose being determined by the
saline solution. After 18 hours, anaesthetise the rat with an
response and usually being 3 to 5. As an initial approximation
anaesthetic that will maintain a prolonged and uniform blood
doses of 3 and 5 milliUnits may be tried. Choose two doses of
pressure. After 45 to 60 minutes, tie the rat on its back to the
the preparation under examination with the same inter-dose
operating table by its hind legs. Cannulate the trachea with a
ratio, matching the effects of the dose of the Standard
short polyethylene tube of external diameter about 2.5 mm
Preparation as closely as possible. Inject doses at intervals of
and dissect a carotid artery ready for cannulation. Then
10 to 15 minutes.
cannulate the femoral vein close to the inguinal ligament.
Retract the abdominal muscles to expose the inguinal ligament. The two doses of the Standard Preparation and the two doses
Retract the superficial pudendal vein to one side and dissect of the preparation under examination should be given in a
the femoral vein towards the inguinal ligament from the randomised block or a Latin square design and four to five
corresponding artery. When dissecting, a deep branch responses to each should be recorded.
reaching the femoral vein must be found and tied off to prevent Measure all the responses and calculate the result of the assay
bleeding during cannulation. Tie a short polyethylene cannula by standard statistical methods.
of external diameter about 1 mm into the femoral vein by two
ligatures and join by a short piece of flexible tubing to a 1-ml Storage. Store at a temperature not exceeding 30º.
burette with an attached thistle funnel containing saline Labelling. The label states (1) the number of Units of oxytocic
solution at about 37º. Firmly fix a wet absorbent cotton swab activity per ml; (2) either the animal species from which it is
to the thigh so as to cover the incision and cannula. At this obtained or whether it is synthetic, as appropriate; (3) that the
stage inject through the venous cannula 200 Units of heparin, preparation is intended for intranasal administration only.
dissolved in saline solution, per 100 g of body weight. Then

889
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

P
Paclitaxel ....
Paclitaxel Injection ....
Pancreatin ....
D-Panthenol ....
Paracetamol ....
Paracetamol Syrup ....
Paracetamol Tablets ....
Hard Paraffin ....
Liquid Paraffin ....
Light Liquid Paraffin ....
Liquid Paraffin Emulsion ....
White Soft Paraffin ....
Yellow Soft Paraffin ....
Paraffin Ointment ....
Paraldehyde ....
Penicillamine ....
Penicillamine Tablets ....
Diluted Pentaerythritol Tetranitrate ....
Pentaerythritol Tetranitrate Tablets ....
Pentamidine Isethionate ....
Pentamidine Injection ....
Pentazocine ....
Pentazocine Hydrochloride ....
Pentazocine Tablets ....
Pentazocine Lactate ....
Pentazocine Injection ....
Pentobarbitone Sodium ....
Pentobarbitone Tablets ....
Pepsin ....
Peritoneal Dialysis Solutions ....

891
MONOGRAPHS INDIAN PHARMACOPOEIA 2007 2007

Pethidine Hydrochloride
Pethidine Injection
Pethidine Tablets
Phenindamine Tartrate
Phenindamine Tablets
Phenindione
Phenindione Tablets
Pheniramine Maleate
Pheniramine Injection
Pheniramine Tablets
Phenobarbitone
Phenobarbitone Tablets
Phenobarbitone Sodium
Phenobarbitone Injection
Phenobarbitone Sodium Tablets
Phenol
Phenolphthalein
Phenoxymethylpenicillin Potassium
Phenoxymethylpenicillin Potassium Tablets
Phentolamine Mesylate
Phentolamine Injection
Phenylbutazone
Phenylbutazone Tablets
Phenylephrine Hydrochloride
Phenylephrine Injection
Phenylmercuric Acetate
Phenylmercuric Nitrate
Phenytoin Sodium
Phenytoin Injection
Phenytoin Tablets
Pholcodine
Pholcodine Linctus
Phosphoric Acid

892
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

Physostigmine Salicylate ....


Physostigmine Injection ....
Pilocarpine Nitrate ....
Pindolol ....
Pindolol Tablets ....
Piperazine Adipate ....
Piperazine Adipate Tablets ....
Piperazine Citrate ....
Piperazine Citrate Syrup ....
Piperazine Hydrate ....
Piperazine Phosphate ....
Piperazine Phosphate Tablets ....
Piroxicam ....
Piroxicam Capsules ....
Plaster Of Paris ....
Polyethylene Glycol 1500 ....
Polyethylene Glycol 4000 ....
Polyethylene Glycol 6000 ....
Polysorbate 20 ....
Polysorbate 80 ....
Potassium Chloride ....
Potassium Chloride And Dextrose Injection ....
Potassium Chloride, Sodium Chloride And Dextrose Injection ....
Potassium Citrate ....
Potassium Clavulanate ....
Potassium Clavulanate Diluted ....
Potassium Iodide ....
Potassium Permanganate ....
Povidone ....
Povidone-Iodine ....
Povidone-Iodine Solution ....
Pralidoxime Chloride ....

893
MONOGRAPHS INDIAN PHARMACOPOEIA 2007 2007

Pralidoxime Chloride Injection ....


Prazosin Hydrochloride ....
Prazosin Tablets ....
Prednisolone ....
Prednisolone Tablets ....
Prednisone ....
Prednisone Tablets ....
Prednisolone Sodium Phosphate ....
Prednisolone Sodium Phosphate Injection ....
Primaquine Phosphate ....
Primaquine Tablets ....
Probenecid ....
Probenecid Tablets ....
Procainamide Hydrochloride ....
Procainamide Injection ....
Procainamide Tablets ....
Procaine Hydrochloride ....
Procaine And Adrenaline Injection ....
Procaine Penicillin ....
Fortified Procaine Penicillin Injection ....
Prochlorperazine Maleate ....
Prochlorperazine Tablets ....
Prochlorperazine Mesylate ....
Prochlorperazine Injection ....
Procyclidine Hydrochloride ....
Procyclidine Tablets ....
Proguanil Hydrochloride ....
Proguanil Tablets ....
Promethazine Hydrochlorid ....
Promethazine Injection ....
Promethazine Syrup ....
Promethazine Tablets ....
Promethazine Theoclate ....

894
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

Promethazine Theoclate Tablets ....


Propantheline Bromide ....
Propantheline Tablets ....
Propranolol Hydrochloride ....
Propranolol Injection ....
Propranolol Tablets ....
Propyl Gallate ....
Propylene Glycol ....
Propylparaben ....
Propylthiouracil ....
Propylthiouracil Tablets ....
Propyphenazone ....
Protamine Sulphate ....
Protamine Sulphate Injection ....
Prothionamide ....
Prothionamide Tablets ....
Pseudoephedrine Hydrochloride ....
Pseudoephedrine Syrup ....
Pseudoephedrine Tablets ....
Psoralen ....
Pyrazinamide ....
Pyrazinamide Tablets ....
Pyridoxine Hydrochloride ....
Pyridoxine Tablets ....
Pyrimethamine ....
Pyrimethamine And Sulphadoxine Tablets ....

895
IP 2007 PACLITAXEL

Paclitaxel – mobile phase: A. a mixture of 60 volumes of water and


40 volumes of acetonitrile,
Taxol B. acetonitrile
– flow rate. 1.2 ml per minute,
O – a linear gradient programme using the conditions given
O below,
O
H3C – spectrophotometer set at 227nm,
CH3 – a 10 µl loop injector.
H3C CH3 OH
O NH O CH3 Time Mobile phase A Mobile phase B
(in min) (per cent v/v) (per cent v/v)
O O
OH H 0 100 0
OH O O
CH3 20 100 0
O O 60 10 90
62 100 0
C47H51NO14 Mol. Wt. 853.9 70 100 0
A taxane derivative first isolated from the bark of the Pacific Inject reference solution (b). The test is not valid unless the
yew tree, Taxus brevifolia. resolution between the peak due to paclitaxel and 10-dactyl -
7-epipactitaxyl is not less than 1.2 relative retention time for
Paclitaxel contains not less than 97.0 per cent and not more
particles and 10-dactyl -7-epipactitaxyl is about 0.94.
than 102.0 per cent of C47H51NO14, calculated on the anhydrous
basis. Inject the test solution and reference solution (a). In the
chromatogram obtained with the test solution, the area of any
Description. A white or almost white powder. secondary peak is not more than 0.5 times the area of the peak
CAUTION — Paclitaxel is potentially cytotoxic. Great care in the chromatogram obtained with the reference solution (a)
should be taken in handling the powder and preparing (0.5 per cent) and the sum of areas of all the secondary peaks
solutions. is not more than twice the area of the peak in the chromatogram
obtained with the reference solution (a) (2.0 per cent).
Identification
Heavy metals. (2.3.13). 1 g complies with the limit test for heavy
A. Determine by infrared absorption spectrophotometry (2.4.6). metals, Method A ( 20ppm ).
Compare the spectrum with that obtained with paciltaxel RS.
Loss on ignition (2.4.20). Not more than 0.2 per cent.
B. In the Assay, the principal peak in the chromatogram Water (2.3.43). Not more than 4.0 per cent, determined on 0.1
obtained with the test solution corresponds to the peak in the g by coloumetry method.
chromatogram obtained with the reference solution.
Bacterial endotoxins (2.2.3). Not more than 0.4 Endotoxin Unit
Tests per mg of paclitaxel.
Specific optical rotation (2.4.22). – 49.0º to – 55.0º, determined Microbial contamination (2.2.9). The total viable aerobic count
in 1.0 per cent w/v solution in methanol. does not exceed 100 cfu per g. It meets the requirements of the
tests for the absence of Staphylococcus aureus, Pseudomonas
Related substances. Determine by liquid chromatography
aeruginosa, Salmonella species, and Escherichia coli.
(2.4.14).
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Dissolve 10 mg of substance under examination
in 10 ml of acetonitrile. Solvent mixture. Dissolve 200 µl of glacial acetic acid in
1000 ml of methanol.
Reference solution (a). A 0.001 per cent w/v solution of
paclitaxel RS in acetonitrile. Test solution. Dissolve 0.1 g of the substance under
examination in 100.0 ml in solvent mixture.
Reference solution (b). A sloution containing 0.008 per cent
w/v of 10 deacetyl -7-epipoelitoral and 0.1 per cent w/v of Reference solution. A 0.1 per cent w/v solution of paclitaxil
paclitaxel RS in acetonitrile. RS in solvent mixture.
Chromatographic system Chromatographic system
– a stainless steel column 15 cm x 4.6 mm, packed with – a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (3µm). pentafluoro phenyl groups chemically bonded to
– column temperature 35°, porous silica (5 µm),

897
PACLITAXEL INJECTION IP 2007

– column temperature 35°, System suitability solution- A sloution containing 0.006 per
– mobile phase: a mixture of 11 volumes of water and 9 cent w/v of 10 deacetyl -7-epipoelitoral and 0.12 per cent w/v
volumes of acetonitrile, filter. of paclitaxel in acetonitrile.
– flow rate. 1.5 ml per minute, Chromatographic system
– spectrophotometer set at 227nm, – a stainless steel column 15 cm x 4.6 mm, packed with
– a 10 µl loop injector. octadecylsilane bonded to porous silica (3 µm).
Inject the reference solution. The test is not valid unless the – column temperature 35°,
tailing factor is not more than 1.5. The relative standard – mobile phase: A. a mixture of 60 volumes of water and
deviation for replicate injections is not more than 2.0 per cent. 40 volumes of acetonitrile,
B. acetonitrile
Inject the test solution and reference solution.
– flow rate. 1.2 ml per minute,
Calculate the content of C47H51NO14. – a linear gradient programme using the conditions given
Storage. Store protected from light, at a temperature not below,
exceeding 25º. – spectrophotometer set at 227nm,
– a 10 µl loop injector.
Time Mobile phase A Mobile phase B
(in min) (per cent v/v) (per cent v/v)
Paclitaxel Injection 0 100 0
26 100 0
Paclitaxel Injection is a sterile solution of Paclitaxel suitable
for dilution,for intravenous use. 66 17 83
67 100 0
Paclitaxel Injection contains not less than 90.0 per cent and
not more than 105.0 per cent of of the related amount of Inject reference solution (b). The test is not valid unless the
paclitaxel, C47H51NO14. column efficiency is not less than 2000 theoretical plates and
the tailing factor is not more than 2.0.
Description. A clear colourless to slight yellow viscous
solution. Inject the test solution and reference solution (a). In the
chromatogram obtained with the test solution, the area of any
Identification secondary peak is not more than 0.8 times the area of the peak
in the chromatogram obtained with the reference solution (a)
A. In the Related substances, the major peak in the (0.8 per cent) and the sum of areas of all the secondary peaks
chromatogram obtained with test solution corresponds to the is not more than twice the area of the peak in the chromatogram
peak in the chromatogram obtained with reference solution. obtained with the reference solution (a) (2.0 per cent).
B. In the Assay, the principal peak in the chromatogram Other tests. Complies with the tests stated under Parenteral
obtained with test solution corresponds to the peak in the Preparations (Injections).
chromatogram obtained with reference solution.
Bacterial endotoxins (2.2.3). Not more than 0.67 Endotoxin
Tests Unit per mg of paclitaxel.
Sterility (2.2.11). Complies with the test for sterility.
pH (2.4.24). 3.0 to 7.0, determined in a solution 10 per cent v/v
solution in water. Assay. Determine by liquid chromatography (2.4.14).

Light absorption. Absorbance of the injection at about 425 Solvent mixture. Dissolve 200 µl of glacial acetic acid in
nm, not more than 0.01. 1000 ml of methanol.
Test solution. Accurately measure the volume of injection
Related substances. Determine by liquid chromatography
containing 6 mg of Paclitaxel and dissolve in 10 ml of solvent
(2.4.14).
mixture.
Test solution. Accurately measure the volume of injection
Reference solution. A 0.06 per cent w/v solution of paclitaxel
containing 12 mg of Paclitaxel, dilute to 10 ml with acetonitrile.
RS in solvent mixture.
Reference solution (a). A 0.12 per cent w/v solution of
Chromatographic system
paclitaxel RS in aceotnitrile.
– a stainless steel column 30 cm x 3.9 mm, packed with
Reference solution (b). Dilute 1 ml of reference solution (a) to penta fluro phenyl group chemically bonded to porous
100 ml with acetonitrile. silica (5 µm),

898
IP 2007 PANCREATIN

– column temperature 35°, Tests


– mobile phase: a mixture of 11 volumes of water and 9
volumes of acetonitrile, Fat. Not more than 5.0 per cent, determined by the following
– flow rate. 1.5 ml per minute, method. Extract 1 g with light petroleum (40º to 60º) for
– spectrophotometer set at 227nm, 3 hours in an apparatus for the continuous extraction of drugs
– a 10 µl loop injector. (2.1.8), evaporate the extract and dry the residue at 105º for
2 hours.
Inject the reference solution. The test is not valid unless the
tailing factor is not more than 1.5. The relative standard Microbial contamination (2.2.9). 1 g is free from Escherichia
deviation for replicate injections is not more than 2.0 per cent. coli; 10 g is free from salmonellae.

Inject the test solution and reference solution. Loss on drying (2.4.19). Not more than 5.0 per cent, determined
on 0.5 g by dying in an oven at 60º at a pressure not exceeding
Calculate the content of C47H51NO14. 0.7 kPa for 4 hours.
Storage. Store protected from light, at a temperature not Assay. For protease activity — Weigh accurately 4.0 g of
exceeding 25º. purified casein and dissolve in about 90 ml of water containing
3 ml of 1 M sodium hydroxide, adjust the pH of the solution to
8.7 and add sufficient water to produce 100.0 ml. Weigh
accurately about 0.5 g of the substance under examination,
Pancreatin triturate with water and add sufficient water to produce
300.0 ml to give the test solution. Dilute 15.0 ml of the casein
Pancreatin is a preparation of mammalian pancreas containing solution with 30 ml of water, warm to 55º and add 10.0 ml of the
protease, lipase and amylase activity. It may contain Sodium unfiltered test solution. Heat rapidly to 55º and keep at this
Chloride. temperature for 20 minutes. Cool rapidly to room temperature.
Dilute a further portion of 15.0 ml of the casein solution with
Pancreatin contains not less than the minimum protease
30 ml of water, add 10.0 ml of the unfiltered test solution,
activity, amylase activity and lipase activity determined under
previously boiled and cooled, heat rapidly to 55º and keep at
the conditions of the Assay.
this temperature for 20 minutes. Cool to room temperature. To
Description. A white or buff-coloured, amorphous powder; each solution add 0.75 ml of phenolphthalein solution and 10
odour, meaty and not unpleasant. ml of formaldehyde solution. Titrate each solution with 0.1 M
sodium hydroxide until the colour of the solution matches
Identification that produced by mixing 10 ml of buffer solution pH 8.7 and
0.15 ml of phenolphthalein solution. The difference between
A. Triturate 0.5 g with 10 ml of water and adjust to pH 8.0 by
the two titrations is not less than 4.5 ml.
the addition of 1 M sodium hydroxide using cresol red
solution as indicator. Divide the resulting solution into two For lipase activity — To 95 ml of water add 6.5 ml of triacetin
equal portions. Boil one portion [solution (1)] and leave the and 0.2 ml of a 0.1 per cent w/v solution of bromocresol purple,
other untreated [solution (2)]. To each add a few shreds of neutralise with 0.5 M sodium hydroxide and add sufficient
congo red fibrin, warm to 39º ± 1º and maintain at this water to produce 110 ml. Place 50 ml of this solution in each of
temperature for 1 hour. Solution (2) is stained red and solution two large tubes 3 cm ´ 20 cm A and B contained in a thermostat
(1) is colourless or not more than slightly pink. at 30º. Insert in each tube a rubber stopper having two holes,
one for the tip of a burette and the other for a short glass tube
B. Triturate 0.25 g with 10 ml of water and adjust to pH 8.0 by
through which passes a thread operating a glass stirring coil.
the addition of 1 M sodium hydroxide using cresol red
Stir the contents of the tube until they attain the temperature
solution as indicator. Divide the resulting solution into two
of the thermostat. Prepare a solution of 0.1 g of the substance
equal portions. Boil one portion [solution (1)] and leave the
under examination in 10.0 ml of water. To tube A add 1.0 ml of
other untreated [solution (2)]. Dissolve 0.1 g of soluble starch
the solution, to tube B add 1.0 ml of the solution previously
in 100 ml of boiling water, boil for 2 minutes, cool and dilute to
boiled. Adjust and maintain the pH of the solutions in the two
150 ml with water. Add solution (1) to half the starch mucilage
tubes to 6.2 to 6.4 by the addition of 0.05 M sodium hydroxide
and solution (2) to the remainder and maintain the mixtures at
dropwise, stirring frequently. After 30 minutes, the difference
39º ± 1º for 5 minutes. To 1 ml of each mixture add 10 ml of
between the volumes of 0.05 M sodium hydroxide added to
iodinated potassium iodide solution. The liquid containing
the two tubes is not less than 1.0 ml.
solution (2) retains the colour of the solution of iodine and
the liquid containing solution (1) acquires an intense blue For amylase activity — Not less than 100 Units per g. Dissolve
colour. 0.1 g or a quantity containing 10 Units, accurately weighed, in

899
D-PANTHENOL IP 2007

sufficient buffer solution pH 6.8 to produce 1000.0 ml. Filter if Refractive index (2.4.27). 1.490 to 1.498, determined at 20º.
necessary (1 ml of the test solution should be capable of Heavy metals (2.3.13). 1.0 g dissolved in 25 ml of water complies
digesting about 10 mg of dry soluble maize or corn starch). with the limit test for heavy metals, Method A (20 ppm).
Into each of six stoppered test-tubes add 5.0 ml of starch
substrate without touching the sides of the test-tube. Place Assay. Weigh accurately about 0.5 g and carry out Method A
the test-tubes in a water-bath maintained at 40º ± 0.1º. When for the determination of nitrogen (2.3.30).
the temperature of the solution in the tubes has reached 40º, 1 ml of 0.05 M sulphuric acid is equivalent to 0.001401 g of N.
add 0.35 ml, 0.4 ml, 0.45 ml, 0.5 ml, 0.55 ml and 0.6 ml of the test
solution to each of the test-tubes marked 1 to 6 respectively Storage. Store protected from moisture.
and record the time of addition. Mix thoroughly and replace
the tubes in the water-bath. After exactly 60 minutes remove
the tubes and cool rapidly in cold water. Add to each tube Paracetamol
0.05 ml of 0.02 M iodine and mix well. Note the tube containing
the lowest volume of test solution, which does not show a Acetaminophen
bluish or violet tinge (if there is doubt, warm the solution
slightly, when the colour distinction is prominent). From this O
volume calculate the number of grams of dry soluble maize or
HN CH3
corn starch digested by 1.0 g of the substance under
examination. This represents the number of Units of amylase
activity per g.
Storage. Store protected from moisture.
OH
Labelling. The label states the name of any added substance.
C8H9NO2 Mol. Wt. 151.2
Paracetamol is 4-hydroxyacetanilide.
D-Panthenol Paracetamol contains not less than 99.0 per cent and not more
Pantothenol; Dextro-pantothenyl Alcohol than 101.0 per cent of C8H9NO2, calculated on the dried basis.
Description. White crystals or a white, crystalline powder.
H OH
HO N OH Identification
H3C CH3 O H Test A may be omitted if tests B C and D are carried out. Tests
C9H19NO4 Mol. Wt. 205.3 B, C and D may be omitted if test A is carried out.
A. Determine by infrared absorption spectrophotometry (2.4.6).
D-Panthenol is (R)-2,4-dihydroxy-N-(3-hydroxypropyl)-3, 3-
Compare the spectrum with that obtained with paracetamol
dimethylbutanamide.
RS or with the reference spectrum of paracetamol.
D-Panthenol contains not less than 6.60 per cent and not more
B. Dissolve 50 mg in sufficient methanol to produce 100 ml.
than 6.95 per cent of nitrogen, N.
To 1 ml of this solution add 0.5 ml of 0.1 M hydrochloric acid
Description. A clear, colourless or slightly yellow, viscous and dilute to 100 ml with methanol. Protect the resulting
liquid; odourless. solution from bright light and immediately measure the
absorbance at the maximum at about 249 nm; absorbance at
Identification 249 nm, about 0.44 (2.4.7).
Boil 50 mg with 5 ml of 0.1 M sodium hydroxide for 1 minute, C. Boil 0.1 g in 1 ml of hydrochloric acid for 3 minutes, add
cool and add 5 ml of 1 M hydrochloric acid and 0.1 ml of ferric 10 ml of water and cool; no precipitate is produced. Add
chloride test solution; a deep yellow colour is produced. 0.05 ml of 0.0167 M potassium dichromate; a violet colour
develops which does not turn red.
Tests
D. Gives the reaction of acetyl groups (2.3.1).
pH (2.4.24). Not more than 10.5, determined in a 5.0 per cent
w/v solution in carbon dioxide-free water. Tests
Specific optical rotation (2.4.22). +28.2º to +30.2º, determined 4-Aminophenol. Dissolve 0.5 g in sufficient methanol (50 per
at 20º in a 5.0 per cent w/v solution. cent) to produce 10 ml. Add 0.2 ml of freshly prepared alkaline

900
IP 2007 PARACETAMOL SYRUP

sodium nitroprusside solution, mix and allow to stand for 1 ml of 0.1 M ceric ammonium sulphate is equivalent to
30 minutes. Any blue colour in the solution is not more intense 0.00756 g of C8H9No2.
than that in 10 ml of a solution prepared at the same time and Storage. Store protected from light and moisture.
in the same manner containing 0.5 g of 4-aminophenol-free
paracetamol and 0.5 ml of a 0.005 per cent w/v solution of
4-aminophenol in methanol (50 per cent) (50 ppm).
Related substances. Determine by thin layer chromatograpy Paracetamol Syrup
(2.4.17), coating the plate with silica gel GF254. Paracetamol Oral Solution; Acetaminophen Syrup
Mobile phase. A mixture of 65 volumes of chloroform, Paracetamol Syrup is a solution of Paracetamol in a suitable
25 volumes of acetone and 10 volumes of toluene. flavoured vehicle.
Test solution (a). Transfer 1 g of the substance under Paracetamol Syrup contains not less than 95.0 per cent and
examination, finely powdered, to a ground-glass stoppered not more than 105.0 per cent w/v solution of the stated amount
15-ml centrifuge tube, add 5 ml of peroxide-free ether, shake of paracetamol, C8H9NO2.
mechanically for 30 minutes and centrifuge at 1000 rpm for 15
minutes or until a clear supernatant liquid is obtained. Identification
Test solution (b). Dilute 1 ml of the test solution to 10 ml with Determine by thin-layer chromatography (2.4.17), coating the
ethanol (95 per cent). plate with silica gel GF254.
Reference solution (a). A 0.005 per cent w/v solution of Mobile phase. A mixture of 65 volumes of chloroform,
4-chloroacetanilide in ethanol (95 per cent). 25 volumes of acetone, 10 volumes of toluene and 0.5 volumes
of glacial acetic acid.
Reference solution (b). Dissolve 0.25 g of 4-chloroacetanilide
and 0.1 g of the substance under examination in sufficient Test solution. Dilute a volume containing 25 mg of Paracetamol
ethanol (95 per cent) to produce 100 ml. to 10 ml with methanol and filter if necessary.
Apply to the plate 200 µl of test solution (a) and 40 µl of each Reference solution. A 0.25 per cent w/v solution of
of test solution (b) and reference solutions (a) and (b). After paracetamol RS in methanol
development, dry the plate in a current of warm air and examine Apply to the plate 10 µl of each solution. After development,
in ultraviolet light at 254 nm. Any spot corresponding to dry the plate in a current of warm air, examine in ultraviolet
4-chloroacetanilide in the chromatogram obtained with test light at 254 nm. The principal spot in the chromatogram
solution (a) is not more intense than the spot in the obtained with the test solution corresponds to that in the
chromatogram obtained with reference solution (a). Any other chromatogram obtained with the reference solution.
secondary spot in the chromatogram obtained with test
solution (b) is not more intense than the spot in the Tests
chromatogram obtained with reference solution (a). The 4-Aminophenol. Determine by liquid chromatography (2.4.14).
chromatogram obtained with reference solution (b) shows two
clearly separated spots, the spot corresponding to paracetamol Test solution. Shake 5 ml of the preparation under examination
having the lower Rf value. with 15 ml of the mobile phase, dilute to 25 ml with the mobile
phase and filter if necessary.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method B (10 ppm). Reference solution. A 0.0025 per cent w/v solution of
4-aminophenol in the mobile phase.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Chromatographic system
Loss on drying (2.4.19). Not more than 0.5 per cent, determined – a stainless steel column 20 cm x 4.6 mm, packed with
on 1.0 g by drying in an oven at 105º. octadecylsilane bonded to porous silica (10 µm),
– mobile phase: 0.01 M sodium butanesulphonate in a
Assay. Weigh accurately about 0.5 g, dissolve in a mixture of
mixture of 85 volumes of water, 15 volumes of methanol
10 ml of water and 50 ml of 1 M sulphuric acid. Boil under a
and 0.4 volume of formic acid,
reflux condenser for 1 hour, cool and dilute to 100.0 ml with
– flow rate. of 2 ml per minute,
water. To 20.0 ml of the solution add 40 ml of water, 40 g of
– spectrophotometer set at 272 nm,
water in the form of ice, 15 ml of 2 M hydrochloric acid and
– a 20 µl loop injector.
0.1 ml of ferroin solution and titrate with 0.1 M ceric
ammonium sulphate until a yellow colour is produced. Carry In the chromatogram obtained with the test solution the area
out a blank titration. of any peak corresponding to 4-aminophenol is not greater

901
PARACETAMOL TABLETS IP 2007

than the area of the peak in the chromatogram obtained with Test solution. Shake a quantity of the powdered tablets
the reference solution. In the chromatogram obtained with the containing 1 g of Paracetamol with 15 ml of methanol, dilute to
test solution peaks with a long retention time may occur due 100 ml with water and filter.
to preservatives in the preparations.
Reference solution. A 0.001 per cent w/v solution of
Other tests. Complies with the tests stated under Oral Liquids. 4-aminophenol in methanol (15 per cent).
Assay. Determine by liquid chromatography (2.4.14). Chromatographic system
Test solution. Mix an accurately weighed quantity of the – a stainless steel column 20 cm x 4.6 mm, packed with
preparation under examination containing 25 mg of octadecylsilane bonded to porous silica (10 µm),
Paracetamol in 100 ml of the mobile phase, dilute to 200.0 ml – mobile phase: 0.01 M sodium butanesulphonate in a
with the mobile phase and filter if necessary. mixture of 85 volumes of water, 15 volumes of methanol
and 0.4 volume of formic acid,
Reference Solution. A 0.0125 per cent w/v solution of – flow rate. 2 ml per minute,
paracetamol RS in the mobile phase. – spectrophotometer set at 272 nm,
Chromatographic system – a 20 µl loop injector.
– a stainless steel column 20 cm x 4.6 mm, packed with In the chromatogram obtained with the test solution the area
octadecylsilane bonded to porous silica (10 µm), of any peak corresponding to 4-aminophenol is not greater
– mobile phase: 0.01 M sodium butanesulphonate in a than the area of the peak in the chromatogram obtained with
mixture of 85 volumes of water, 15 the reference solution. In the chromatogram obtained with the
– volumes of methanol and 0.4 volume of formic acid, test solution peaks with a long retention times may occur due
– flow rate. of 2 ml per minute, to excipients.
– spectrophotometer set at 243 nm,
– a 20 µl loop injector. Related substances. Determine by thin layer chromatograpy
(2.4.17), coating the plate with silica gel F254.
Determine the weight per ml of the syrup (2.4.29), and calculate
the percentage content of C8H9NO2, weight in volume. Mobile phase. A mixture of 65 volumes of chloroform,
25 volumes of acetone and 10 volumes of toluene.
Storage. Store protected from light and moisture.
Test solution (a). Transfer a quantity of the powdered tablets
containing 1 g of Paracetamol to a ground-glass stoppered
15-ml centrifuge tube, add 5 ml of peroxide-free ether, shake
Paracetamol Tablets mechanically for 30 minutes, centrifuge at 1000 rpm for
Acetaminophen Tablets 15 minutes or until a clear supernatant liquid is obtained and
use the supernatant liquid.
Paracetamol Tablets contain not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of Test solution (b). Dilute 1 ml of the test solution to 10 ml with
paracetamol, C8H9NO2. ethanol (95 per cent).
Reference solution (a). A 0.005 per cent w/v solution of
Identification
4-chloroacetanilide in ethanol (95 per cent).
Extract a quantity of the powdered tablets containing 0.5 g of
Reference solution (b). Dissolve 0.25 g of 4-chloroacetanilide
Paracetamol with 20 ml of acetone, filter, evaporate the filtrate
and 0.1 g of the substance under examination in sufficient
to dryness and dry at 105º. The residue complies with the
ethanol (95 per cent) to produce 100 ml.
following tests.
Apply to the plate 200 µl of test solution (a) and 40 µl of each
A. Determine by infrared absorption spectrophotometry (2.4.6).
of test solution (b) and reference solutions (a) and (b). After
Compare the spectrum with that obtained with paracetamol
development, dry the plate in a current of warm air and examine
RS or with the reference spectrum of paracetamol.
in ultraviolet light at 254 nm. Any spot corresponding to
B. Boil 0.1 g in 1 ml of hydrochloric acid for 3 minutes, add 4-chloroacetanilide in the chromatogram obtained with test
10 ml of water and cool; no precipitate is produced. Add solution (a) is not more intense than the spot in the
0.05 ml of 0.0167 M potassium dichromate; a violet colour chromatogram obtained with reference solution (a). Any other
develops which does not turn red. secondary spot in the chromatogram obtained with test
solution (b) is not more intense than the spot in the
Tests
chromatogram obtained with reference solution (a). The
4-Aminophenol. Determine by liquid chromatography (2.4.14). chromatogram obtained with reference solution (b) shows two

902
IP 2007 LIQUID PARAFFIN

clearly separated spots, the spot corresponding to paracetamol Liquid Paraffin


having the lower Rf value.
White Mineral Oil; Liquid Petrolatum
Dissolution (2.5.2).
Liquid Paraffin is a purified mixture of liquid hydrocarbons
Apparatus. No 1
obtained from petroleum to which not more than 10 ppm of
Medium. 900 ml of phosphate buffer pH 5.8
tocopherol or of butylated hydroxytoluene may be added.
Speed and time. 50 rpm and 30 minutes.
Description. A transparent, colourless, oily liquid, free from
Withdraw a suitable volume of the medium and filter and dilute fluorescence by daylight; odourless or almost odourless.
a suitable volume of the filtrate with the same solvent. Measure
the absorbance of the resulting solution at the maximum at Tests
about 243 nm (2.4.7). Similarly measure the absorbance of a
solution of known concentration of paracetamol RS. Calculate Weight per ml (2.4.29). 0.860 g to 0.904 g.
the content of C8H9NO2. Dynamic viscosity (2.4.28). 110 mPas to 230 mPas, determined
at 20º ± 1º by Method B.
D. Not less than 80 per cent of the stated amount of C8H9NO2.
Acidity or alkalinity. To 10.0 g add 20 ml of boiling water, heat
Other tests. Complies with the tests stated under Tablets.
in a water-bath for 5 minutes, shake vigorously for 1 minute,
Assay. Weigh and powder 20 tablets. Weigh accurately a cool, allow to separate and filter the aqueous layer. To 10 ml of
quantity of the powder containing about 0.15 g of Paracetamol, the filtrate add 0.1 ml of phenolphthalein solution. The
add 50 ml of 0.1 M sodium hydroxide, dilute with 100 ml of solution is colourless and not more than 0.1 ml of 0.1 M sodium
water, shake for 15 minutes and add sufficient water to produce hydroxide is required to change the colour of the solution to
200.0 ml. Mix, filter and dilute 10.0 ml of the filtrate to 100.0 ml pink.
with water. To 10.0 ml of the resulting solution add 10 ml of Light absorption. When examined in the range 240 nm to
0.1 M sodium hydroxide, dilute to 100.0 ml with water and 280 nm (2.4.7), a 2.0 per cent w/v solution in 2,2,4-trimethyl-
mix. Measure the absorbance of the resulting solution at the pentane shows an absorption of not more than 0.1.
maximum at about 257 nm (2.4.7). Calculate the content of
C8H9NO2 taking 715 as the specific absorbance at 257 nm. Readily carbonisable substances. Place 5 ml in a dry, heat-
resistant glass-stoppered test-tube (125 mm x 18 mm)
Storage. Store protected from light and moisture. previously rinsed with chromic acid solution, then with water
and dried. Add 5 ml of nitrogen-free sulphuric acid
(containing 94.5 per cent to 95.5 per cent w/w of H2SO4), insert
the stopper and shake as vigorously as possible in the
Hard Paraffin longitudinal direction of the tube for 5 seconds. Loosen the
stopper, immediately place the tube in a bath of boiling water,
Hard Paraffin is a purified mixture of solid hydrocarbons supporting it so as to prevent contact of the tube with the
obtained from petroleum or from shale oil. bottom or side of the bath and heat for 10 minutes. At the end
Description. A white or colourless, translucent mass, of the second, fourth, sixth, and eighth minutes, remove the
frequently showing a crystalline structure; odourless even tube from the bath and shake as vigorously as possible in the
when freshly cut; slightly greasy to the touch. Burns with a longitudinal direction of the tube for 5 seconds. At the end of
luminous flame. When melted, the liquid is free from 10 minutes from the time the tube was placed in the bath
fluorescence by daylight. remove the tube and allow to stand for 10 minutes. The lower
acid layer is not more intensely coloured than a mixture of 3 ml
Tests of FCS, 1.5 ml of CCS and 0.5 ml of CSS (2.4.1), overlaid with
5 ml of liquid paraffin. If the sulphuric acid remains dispersed
Acidity or alkalinity. To 10.0 g add 20 ml of boiling water, heat
in the molten paraffin, the colour of the emulsion is not darker
in a water-bath for 5 minutes, shake vigorously for 1 minute,
than that of the standard mixture when shaken vigorously.
cool, allow to separate and filter the aqueous layer. To 10 ml of
the filtrate add 0.1 ml of phenolphthalein solution. The Solid paraffins. Place a suitable quantity, previously dried by
solution is colourless and not more than 0.1 ml of 0.1 M sodium heating at 100º for 2 hours and cooled in a desiccator over
hydroxide is required to change the colour of the solution to sulphuric acid, in a glass cylindrical vessel having an internal
pink. diameter of approximately 25 mm. Close the vessel and immerse
in a mixture of ice and water; after 4 hours the liquid is
Congealing range (2.4.10). 50º to 65º.
sufficiently clear that a black line, 0.5 mm in width, held vertically
Sulphated ash (2.3.18). Not more than 0.1 per cent. behind the vessel is easily seen.

903
LIGHT LIQUID PARAFFIN IP 2007

Sulphur compounds. Mix 4 ml with 2 ml of ethanol (95 per dispersed in the molten paraffin, the colour of the emulsion is
cent), and 2 drops of a clear, saturated solution of lead not darker than that of the standard mixture when shaken
monoxide in sodium hydroxide solution and heat at 70º for vigorously.
10 minutes with frequent shaking; the mixture remains Solid paraffins. Place a suitable quantity, previously dried by
colourless. heating at 100º for 2 hours and cooled in a desiccator over
Storage. Store protected from light. sulphuric acid, in a glass cylindrical vessel having an internal
diameter of approximately 25 mm. Close the vessel and immerse
in a mixture of ice and water; after 4 hours the liquid is
sufficiently clear that a black line, 0.5 mm in width, held vertically
Light Liquid Paraffin behind the vessel is easily seen.
Light Mineral Oil; Light Liquid Petrolatum Sulphur compounds. Mix 4 ml with 2 ml of ethanol (95 per
cent), and 2 drops of a clear, saturated solution of lead
Light Liquid Paraffin is a purified mixture of liquid saturated
monoxide in sodium hydroxide solution and heat at 70º for
hydrocarbons obtained from petroleum. It may contain a
10 minutes with frequent shaking; the mixture remains
suitable stabiliser.
colourless.
Description. A transparent, colourless, oily liquid, free from
Storage. Store protected from light.
fluorescence by daylight; almost odourless when cold.

Tests
Weight per ml (2.4.29). 0.820 g to 0.880 g. Liquid Paraffin Emulsion
Dynamic viscosity (2.4.28). 25 mPa s to 80 mPa s, determined Liquid Paraffin Oral Emulsion
at 20º ± 1º by Method B. Liquid Paraffin Emulsion is an oral emulsion of Liquid Paraffin
Acidity or alkalinity. To 10.0 g add 20 ml of boiling water, heat in Purified Water.
in a water-bath for 5 minutes, shake vigorously for 1 minute, Liquid Paraffin Emulsion contains not less than 44.0 per cent
cool, allow to separate and filter the aqueous layer. To 10 ml of and not more than 49.0 per cent w/w of liquid paraffin.
the filtrate add 0.1 ml of phenolphthalein solution. The
solution is colourless and not more than 0.1 ml of 0.1 M sodium Tests
hydroxide is required to change the colour of the solution to
pink. Other tests. Complies with the tests stated under Oral Liquids.
Light absorption. When examined in the range 240 nm to 280 Assay. Weigh accurately about 5.0 g, add 10 ml of water, extract
nm (2.4.7), a 2.0 per cent w/v solution in 2,2,4-trimethylpentane with two quantities, each of 40 ml, of a mixture of 2 volumes of
shows an absorption of not more than 0.1. ethanol (95 per cent), 3 volumes of light petroleum (40º to
60º) and 3 volumes of ether and then with 30 ml of a mixture of
Readily carbonisable substances. Place 5 ml in a dry, heat- equal volumes of light petroleum (40º to 60º) and ether. Wash
resistant glass-stoppered test-tube (125 mm x 18 mm) the combined extracts with 15 ml of 0.5 M sodium hydroxide
previously rinsed with chromic acid solution, then with water and then with 15 ml of water, evaporate the solvent, add 5 ml
and dried. Add 5 ml of nitrogen-free sulphuric acid of acetone and evaporate again. Repeat the addition and
(containing 94.5 per cent to 95.5 per cent w/w of H2SO4), insert evaporation of acetone until the residue is free from water,
the stopper and shake as vigorously as possible in the dry at 105º for 15 minutes and weigh.
longitudinal direction of the tube for 5 seconds. Loosen the
stopper, immediately place the tube in a bath of boiling water, Storage. Store protected from moisture.
supporting it so as to prevent contact of the tube with the
bottom or side of the bath and heat for 10 minutes. At the end
of the second, fourth, sixth, and eighth minutes, remove the
tube from the bath and shake as vigorously ssas possible in
White Soft Paraffin
the longitudinal direction of the tube for 5 seconds. At the White Petroleum Jelly
end of 10 minutes from the time the tube was placed in the
White Soft Paraffin is a purified, semi-solid mixture of
bath remove the tube and allow to stand for 10 minutes. The
hydrocarbons obtained from petroleum and bleached.
lower acid layer is not more intensely coloured than a mixture
of 3 ml of FCS, 1.5 ml of CCS and 0.5 ml of CSS (2.4.1), overlaid Description. A white, translucent, soft unctuous mass,
with 5 ml of liquid paraffin. If the sulphuric acid remains retaining these characteristics on storage and when melted

904
IP 2007 YELLOW SOFT PARAFFIN

and allowed to cool without stirring; not more than slightly and read the total penetration from the scale. Make three or
fluorescent by daylight, even melted; odourless when rubbed more trials, each so spaced that there is no overlapping of the
on the skin. areas of penetration. Where the penetration exceeds 20 mm,
use a separate container of the test substance for each trial.
Tests Read the penetration to the nearest 0.1 mm. Calculate the
average of the three or more readings and conduct further
Melting range (2.4.21). 38º to 56º, determined by Method IV. trials to a total of 10 if the individual results differ from the
Acidity or alkalinity. To 10.0 g add 20 ml of boiling water, heat average by more than ± 3 per cent. The final average of the
in a water-bath for 5 minutes, shake vigorously for 1 minute, trials is not less than 10.0 mm and not more than 30.0 mm
cool, allow to separate and filter the aqueous layer. To 10 ml of indicating a consistency value between 100 and 300.
the filtrate add 0.1 ml of phenolphthalein solution. The Sulphated ash (2.3.18). Not more than 0.1 per cent.
solution is colourless and not more than 0.1 ml of 0.1 M sodium
hydroxide is required to change the colour of the solution to Storage. Store protected from light and moisture.
pink.
Light absorption (2.4.7). Absorbance of a 0.05 per cent w/v
solution in 2,2,4-trimethylpentane at about 290 nm, not more Yellow Soft Paraffin
than 0.5.
Yellow Petroleum Jelly
Fixed oils, fats and resin. Digest 10 g with 50 ml of sodium
hydroxide solution at 100º for 30 minutes and allow the Yellow Soft Paraffin is a purified, semi-solid mixture of
aqueous layer to separate. On acidifying the aqueous layer hydrocarbons obtained from petroleum.
with dilute sulphuric acid, no precipitate or oily matter is Description. A pale yellow to yellow, translucent, soft
produced. unctuous mass, retaining these characteristics on storage and
Foreign organic matter. Volatilises when heated, without when melted and allowed to cool without stirring; not more
emitting an acrid odour. than slightly fluorescent by daylight, even melted; odourless
when rubbed on the skin.
Consistency. 100 to 300, determined by the following method.
Apparatus. The apparatus is essentially in agreement with IS Tests
4887:1968 and comprises a penetrometer fitted with a polished Melting range (2.4.21). 38º to 56º, determined by Method IV.
cone-shaped metal plunger weighing 150 g having a detachable
steel tip of the following dimensions. The tip of the cone has Light absorption (2.4.7). Absorbance of a 0.05 per cent w/v
an angle of 30º, the point being truncated to a diameter of solution in 2,2,4-trimethylpentane at about 290 nm, not more
0.38 ± 0.08 mm, the base of the tip is 8.38 ± 0.13 mm in diameter than 0.75.
and the length of the tip is 15 ± 0.25 mm. The remaining portion Acidity or alkalinity. To 10.0 g add 20 ml of boiling water, heat
of the cone has an angle of 90º, is 28 to 29 mm in height, and in a water-bath for 5 minutes, shake vigorously for 1 minute,
has a maximum diameter of 65.1 mm at the base. The containers cool, allow to separate and filter the aqueous layer. To 10 ml of
of the test are flat-bottomed metal or glass cylinders that are the filtrate add 0.1 ml of phenolphthalein solution. The
102 ± 6 mm in diameter and not less than 60 mm in height. solution is colourless and not more than 0.1 ml of 0.1 M sodium
Procedure. Melt a sufficient quantity at a temperature below hydroxide is required to change the colour of the solution to
85º and pour into one or more of the containers filling to within pink.
6 mm of the rim. Cool to 25º± 2.5º over a period of not less than Fixed oils, fats and resin. Digest 10 g with 50 ml of sodium
16 hours, protected from drafts. Two hours before the test, hydroxide solution at 100º for 30 minutes and allow the
place the containers in a water-bath at 25º ± 0.5º. If the room aqueous layer to separate. On acidifying the aqueous layer
temperature is below 23.5º or above 26.5º, adjust the with dilute sulphuric acid, no precipitate or oily matter is
temperature of the cone to 25º ± 0.5º by placing it in a water- produced.
bath.
Foreign organic matter. Volatilises when heated, without
Without disturbing the surface of the substance under emitting an acrid odour.
examination, place the container on the penetrometer table,
Consistency. 100 to 300, determined by the following method.
and lower the cone until the tip just touches the top surface of
the test substance at a spot 25 mm to 38 mm from the edge of Apparatus. The apparatus is essentially in agreement with IS
the container. Adjust the zero setting and quickly release the 4887.1968 and comprises a penetrometer fitted with a polished
plunger, then hold it free for 5 seconds. Secure the plunger cone-shaped metal plunger weighing 150 g having a detachable

905
PARAFFIN OINTMENT IP 2007

steel tip of the following dimensions. The tip of the cone has Paraldehyde
an angle of 30º, the point being truncated to a diameter of
0.38 ± 0.08 mm, the base of the tip is 8.38 ± 0.13 mm in diameter H3C O CH3
and the length of the tip is 15 ± 0.25 mm. The remaining portion
of the cone has an angle of 90º, is 28 to 29 mm in height, and O O
has a maximum diameter of 65.1 mm at the base. The containers
of the test are flat-bottomed metal or glass cylinders that are CH3
102 ± 6 mm in diameter and not less than 60 mm in height.
C6H12O3 Mol. Wt. 132.7
Procedure. Melt a sufficient quantity at a temperature below
Paraldehyde is 2,4,6-trimethyl-1,3,5-trioxane, the cyclic trimer
85º and pour into one or more of the containers filling to within
of acetaldehyde. It may contain a suitable amount of
6 mm of the rim. Cool to 25º± 2.5º over a period of not less than
antioxidant.
16 hours, protected from drafts. Two hours before the test,
place the containers in a water-bath at 25º ± 0.5º. If the room Description. A colourless or slightly yellow, transparent liquid;
temperature is below 23.5º or above 26.5º, adjust the odour, strong and characteristic. Solidifies at low temperature
temperature of the cone to 25º ± 0.5º by placing it in a water- to form a crystalline mass.
bath.
Identification
Without disturbing the surface of the substance under
examination, place the container on the penetrometer table, A. Heat 5 ml with 0.1 ml of 1 M sulphuric acid; acetaldehyde,
and lower the cone until the tip just touches the top surface of recognisable by its odour, is evolved.
the test substance at a spot 25 mm to 38 mm from the edge of
B. To 5 ml of a 10 per cent v/v solution add 5 ml of ammoniacal
the container. Adjust the zero setting and quickly release the
silver nitrate solution in a test-tube and heat on a water-bath;
plunger, then hold it free for 5 seconds. Secure the plunger
metallic silver is deposited as a mirror on the sides of the tube.
and read the total penetration from the scale. Make three or
more trials, each so spaced that there is no overlapping of the C. A 10 per cent w/v solution in carbon dioxide-free water is
areas of penetration. Where the penetration exceeds 20 mm, clear (2.4.1), but becomes turbid on warming.
use a separate container of the test substance for each trial.
Read the penetration to the nearest 0.1 mm. Calculate the Tests
average of the three or more readings and conduct further Congealing range (2.4.10). 10º to 13º.
trials to a total of 10 if the individual results differ from the
average by more than ± 3 per cent. The final average of the Distillation range (2.4.8). Not more than 10 per cent distils
trials is not less than 10.0 mm and not more than 30.0 mm below 123º and not less than 95 per cent distils below 126º.
indicating a consistency value between 100 and 300. Refractive index (2.4.27). 1.403 to 1.406.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Relative density (2.4.29). 0.991 to 0.996.
Storage. Store protected from light and moisture. Acetaldehyde. Shake 5 ml with a mixture of 5 ml of ethanol
(60 per cent), 5 ml of hydroxylamine hydrochloride reagent
in ethanol (60 per cent) and 2 drops of methyl orange solution
and titrate with 0.5 M sodium hydroxide to full yellow colour;
Paraffin Ointment not more than 0.8 ml of 0.5 M sodium hydroxide is required.
White Beeswax 20 g Acidity. Mix 5 ml with 45 ml of carbon dioxide-free water and
Hard Paraffin 30 g titrate with 0.1 M sodium hydroxide using phenolphthalein
Cetostearyl Alcohol 50 g solution as indicator; not more than 1.5 ml is required.
White Soft Paraffin* 900 g Chlorides. To 5 ml of a 1 per cent v/v solution add one drop of
*May be replaced by Yellow Soft Paraffin if other medicaments nitric acid and three drops of silver nitrate solution; no
to be incorporated are coloured. opalescence is produced immediately.
Mix the ingredients, heat gently with stirring until Sulphates. To 5 ml of a 1 per cent v/v solution add one drop of
homogeneous and stir until cold. hydrochloric acid and three drops of barium chloride
solution; no turbidity is produced.
Tests Peroxides. In a stoppered vessel, dissolve 5 ml in sufficient of
Paraffin Ointment complies with the tests stated under recently boiled and cooled water to produce 50 ml, add 5 ml of
Ointments. dilute sulphuric acid and 10 ml of potassium iodide solution.

906
IP 2007 PENICILLAMINE

Close the flask and set aside in the dark for 15 minutes. Titrate and expose to iodine vapour for 5 to 10 minutes. The principal
with 0.1 M sodium thiosulphate using starch solution as spot in the chromatogram obtained with the test solution
indicator; set aside for 5 minutes and, if necessary, complete corresponds to that in the chromatogram obtained with the
the titration. Not more than 2.0 ml of 0.1 M sodium reference solution.
thiosulphate is required. B. Dissolve 0.5 g in a mixture of 0.5 ml of hydrochloric acid
Non-volatile matter. Heat 5 ml in a small dish on a water-bath and 4 ml of warm acetone, cool in ice and scratch the inside of
and dry at 105º for 1 hour; the residue weighs not more than 3 the tube with a glass rod to initiate crystallisation; a white
mg (0.06 per cent w/v). precipitate is produced. Filter under vacuum, wash the
precipitate with acetone and dry with suction. A 1 per cent
Storage. Store protected from moisture, in complete darkness
w/v solution of the dried material is dextrorotatory.
and at a temperature of 8º to 15º. If solidified, the whole of the
contents of the container should be liquified by warming C. To 4 ml of a 1 per cent w/v solution add 2 ml of
before use. phosphotungstic acid solution and heat nearly to boiling; a
blue colour is produced.
NOTE — Do not use Paraldehyde if it has a brownish colour
or an odour of acetic acid. Avoid contact with rubber and D. In the test for Penicillamine disulphide, the principal peak
plastics. in the chromatogram obtained with test solution (b)
Labelling. The label states (1) the nature and the proportion corresponds to the peak due to penicillamine in the
of any antioxidant added; (2) that it may decompose on chromatogram obtained with reference solution (a).
standing to form potentially harmful substances. Tests
Appearance of solution. A 10.0 per cent w/v solution in carbon
dioxide-free water (solution A) is clear (2.4.1), and not more
Penicillamine intensely coloured than degree 6 of the appropriate range of
reference solutions (2.4.1).
D-Penicillamine
pH (2.4.24). 4.5 to 5.5, determined in a 1.0 per cent w/v solution.
CH3 O Specific optical rotation (2.4.22). –61.0º to –65.0º, determined
H3 C
OH in a 5.0 per cent w/v solution in 1 M sodium hydroxide.
HS
H NH2 Heavy metals (2.3.13). 10 ml of solution A, complies with the
limit test for heavy metals, Method D (20 ppm). Use lead
C5H11NO2S Mol. Wt. 149.2
standard solution (2 ppm Pb) to prepare the standard.
Penicillamine is 3-mercapto-D-valine.
Mercuric salts. Determine by atomic absorption
Penicillamine contains not less than 98.0 per cent and not spectrophotometry (2.4.2), using a solution prepared in the
more than 101.0 per cent of C5H11NO2S, calculated on the dried following manner. To 1.0 g of the substance under examination
basis. add 10 ml of water and 0.15 ml of perchloric acid and swirl
Description. A white or almost white, crystalline powder. until dissolution is complete. Add 1 ml of ammonium
pyrrolidinedithiocarbamate solution that has been washed
Identification three times immediately before use, each time with an equal
volume of 4-methyl-2-pentanone. Mix, add 2 ml of 4-methyl-
Test A may be omitted if test B, C and D carried out. Test D 2-pentanone, shake for 1 minute, dilute to 25 ml with water,
may be omitted if test A, B and C are carried out. allow the layers to separate and use the 4-methyl-2-pentanone
A. Determine by thin layer chromatography (2.4.17), coating layer. Measure the absorbance at 254 nm using a mercury
the plate with silica gel G. hollow-cathode lamp and an air-acetylene flame and setting
the zero using a 4-methyl-2-pentanone layer obtained by
Mobile phase. A mixture of 40 volumes of 1-butanol,
repeating the procedure described above but omitting the
10 volumes of glacial acetic acid and 10 volumes of water.
substance under examination. For the standard solution
Test solution. Dissolve 0.25 g of the substance under dissolve 0.108 g of yellow mercuric oxide in the minimum
examination in 100 ml of water. volume of 2 M hydrochloric acid, add sufficient water to
Reference solution. A 0.25 per cent w/v solution of produce 1000.0 ml and treat suitable volumes in the same manner
penicillamine RS in water. as the solution of the substance under examination (10 ppm).
Apply to the plate 2 µl of each solution. Allow the mobile Penicillamine disulphide. Determine by liquid
phase to rise 10 cm. Dry the plate at 105º for 5 to 10 minutes chromatography (2.4.14).

907
PENICILLAMINE IP 2007

Test solution (a). Dissolve 40 mg of the substance under ether and shake vigorously for 1 minute. Repeat the extraction
examination in 5 ml of the mobile phase, add 1 ml of a 0.0025 and combine the ether layers. Add 8 ml of phosphate buffer
per cent w/v solution of sulphanilamide (internal standard) pH 2.5, shake for 1 minute, allow to settle and separate the
in the mobile phase and dilute to 10 ml with the mobile phase. ether layer quantitatively, taking care to eliminate the aqueous
phase completely. (Penicillin is unstable at pH 2.5; carry out
Test solution (b). Dissolve 40 mg of the substance under
the operations at this pH within 6 to 7 minutes). Add 8 ml of
examination in the mobile phase and dilute to 10 ml with the
phosphate buffer pH 6.0, shake for 5 minutes, allow to settle,
same solvent.
separate the aqueous layer and check that the pH is 6.0. For
Reference solution (a). A 0.4 per cent w/v solution of solution (2) add 20 µl of penicillinase solution to 2 ml of
penicillamine RS in the mobile phase. solution (1) and incubate at 37º for 1 hour. For solution (3)
dissolve 5 mg of benzylpenicillin sodium in 500 ml of
Reference solution (b). Add 1 ml of test solution (a) to 1 ml of
phosphate buffer pH 6.0 and dilute 0.25 ml of this solution to
a 0.04 per cent w/v solution of penicillamine disulphide RS in
200 ml with phosphate buffer pH 2.5. Carry out the extraction
the mobile phase and dilute to 10 ml with the mobile phase.
procedure described under solution (1) using 8 ml of this
Chromatographic system solution and beginning at the words “add 8 ml of ether...”. For
– a stainless steel column 25 cm x 5 mm, packed with solution (4) add 20 ml of penicillinase solution to 2 ml of
octylsilane bonded to porous silica (5 to 10 µm), solution (3) and incubate at 37º for 1 hour. Prepare solution (5)
– mobile phase: an equal volume of 0.2 per cent w/v in the same manner as solution (1) but omitting the substance
solution of methanesulphonic acid and 0.01 per cent under examination.
w/v solution of disodium edetate, Maintain the dishes at 30º for at least 24 hours. Measure the
– flow rate. 2 ml per minute, diameters of the zones of inhibition to within 0.1 mm. The test
– spectrophotometer set at 220 nm, is not valid unless solution (3) gives a clear zone of inhibition
– a 20 µl loop injector. and solutions (4) and (5) give no zones of inhibition. If solution
In the chromatogram obtained with test solution (a) the ratio (1) gives a zone of inhibition it is caused by penicillin provided
of the area of any peak corresponding to penicillamine solution (2) gives no zone of inhibition. If this is the case, the
disulphide to the area of the peak due to the internal standard average diameter of the zones of inhibition given by solution
is not greater than the corresponding ratio in the chromatogram (1) for the five Petri dishes is less than that given by solution
obtained with reference solution (b). (3) (0.1 ppm).
Penicillin. Carry out the following procedure in a penicillin- Nutrient medium
free atmosphere and with equipment reserved for the test. Peptone 5 g
Sterilise the equipment at 180º for 3 hours and the buffer
Yeast extract 1.5 g
solutions at 121º for 20 minutes before use.
Meat extract 1.5 g
Liquefy a suitable nutrient medium such as that described Sodium chloride 3.5 g
below and inoculate at a suitable temperature with a culture of
Agar 15 g
Micrococcus flavus (ATCC 9341) to give 5 x 104 micro-
organisms per ml or a quantity necessary to obtain the required Distilled water 1000 ml
sensitivity and formation of clearly defined inhibition zones Adjust the pH to 6.0
of suitable diameter. Immediately pour the inoculated medium Penillic acid. Absorbance of a 0.2 per cent w/v solution at
into five Petri dishes (10 cm in diameter) to give uniform layers about 268 nm, not more than 0.07 (2.4.7) (about 0.5 per cent).
2 to 5 mm in depth. Alternatively, the medium may consist of
two layers, only the upper layer being inoculated. Store the Sulphated ash (2.3.18). Not more than 0.1 per cent.
dishes so that no appreciable growth or death of micro- Loss on drying (2.4.19). Not more than 0.5 per cent, determined
organisms occurs before use and so that the surface of the on 1.0 g by drying in an oven at 60º over phosphorus pentoxide
medium is dry at the time of use. In each dish, place five at a pressure not exceeding 0.7 kPa.
stainless steel hollow cylinders (6 mm in diameter) on the
Assay. Dissolve 0.1 g in 30 ml of anhydrous glacial acetic
surface of the medium evenly spaced on a circle with a radius
acid. Titrate with 0.1 M perchloric acid, determining the end-
of about 25 mm and concentric with the dish. For each dish,
point potentiometrically (2.4.25). Carry out a blank titration.
place in separate cylinders 0.15 ml of each of the following
five solutions. 1 ml of 0.1 M perchloric acid is equivalent to 0.01492 g of
C5H11NO2S.
For solution (1) dissolve 1.0 g of the substance under
examination in 8 ml of phosphate buffer pH 2.5, add 8 ml of Storage. Store protected from moisture.

908
IP 2007 DILUTED PENTAERYTHRITOL TETRANITRATE

Penicillamine Tablets In the chromatogram obtained with the test solution the area
of any peak corresponding to penicillamine disulphide is not
D-Penicillamine Tablets greater than the area of the principal peak in the chromatogram
Penicillamine Tablets contain not less than 95.0 per cent and obtained with the reference solution (1.0 per cent).
not more than 105.0 per cent of the stated amount of Other tests. Complies with the tests stated under Tablets.
penicillamine, C5H11NO2S. The tablets are coated.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Identification quantity of the powder containing 0.1 g of Penicillamine,
dissolve as completely as possible in 50 ml of water and filter.
A. Shake a quantity of the powdered tablets containing 20 mg Add to the filtrate 5 ml of 1 M sodium hydroxide and 0.2 ml of
of Penicillamine with 4 ml of water and filter. Add to the filtrate a 0.1 per cent w/v solution of dithizone in ethanol (95 per
2 ml of phosphotungstic acid solution and allow to stand for cent) and titrate with 0.02 M mercuric nitrate.
5 minutes; a blue colour is produced.
1 ml of 0.02 M mercuric nitrate is equivalent to 0.005968 g of
B. Dissolve a quantity of the powdered tablets containing C5H11NO2S.
10 mg of Penicillamine in 5 ml of water and add 0.3 ml of 5 M
Storage. Store protected from moisture.
sodium hydroxide and 20 mg of ninhydrin; an intense blue or
violet-blue colour is produced immediately.

Tests
Mercuric salts. Disperse a quantity of the powdered tablets
Diluted Pentaerythritol Tetranitrate
containing 1 g of Penicillamine in 10 ml of water in a stoppered
flask, add 0.2 ml of 9 M perchloric acid and swirl to dissolve. O2 N NO2
O O
Add 1 ml of ammonium pyrrolidinedithiocarbamate solution,
mix, add 2 ml of 4-methyl-2-pentanone, shake for 1 minute and
O2N O O
add sufficient water to produce 25 ml. Determine by atomic NO2
absorption spectrophotometry (2.4.2), using a mercury hollow-
cathode lamp and an air-acetylene flame and setting the zero C5H8N4O12 Mol. Wt. 316.1
using a 4-methyl-2-pentanone layer obtained by repeating
the procedure described above but omitting the substance Diluted Pentaerythritol Tetranitrate is a dry mixture of 2,2-
under examination, measuring at 254 nm. Use mercury solution bis(hydroxymethyl)- propane1,3-diol tetranitrate with
AAS, suitably diluted with water, for the standard solutions, Lactose or Mannitol or a mixture of Lactose and Starch or
adjusted to contain the same concentrations of 9 M perchloric any other suitable inert excipients which permit safe
acid, ammonium pyrrolidinedithiocarbamate solution and handling.
4-methyl-2-pentanone as the solution under examination Diluted Pentaerythritol Tetranitrate contains not less than
(40 ppm). 95.0 per cent and not more than 105.0 per cent of the stated
Penicillamine disulphide. Determine by liquid amount of pentaerythritol tetranitrate, C5H8N4O12.
chromatography (2.4.14). Description. A white or almost white, powder; odour, faint
Test solution. Shake quantity of the powdered tablets and mild.
containing 40 mg of Penicillamine with 10 ml of the mobile
phase, filter and use the filtrate. Identification
Reference solution. A 0.004 per cent w/v solution of A. Transfer a quantity containing 10 mg of pentaerythritol
penicillamine disulphide RS in the mobile phase. tetranitrate to a medium porosity sintered-glass filter, add 5 ml
Chromatographic system of dry acetone and collect the filtrate. Repeat with two further
– a stainless steel column 25 cm x 5 mm, packed with quantities, each of 5 ml, of dry acetone and evaporate the
octylsilane bonded to porous silica (5 to 10 µm), combined filtrate at a temperature not exceeding 60º, with the
– mobile phase: an equal volume of 0.2 per cent w/v aid of a gentle current of air, and dry the residue at 60º for
solution of methanesulphonic acid and 0.01 per cent 4 hours; the residue melts at 138º to 142º (2.4.21).
w/v solution of disodium edetate, B. Suspend 10 mg of the residue obtained in test A in a mixture
– flow rate. 2 ml per minute, of 2 ml of sulphuric acid and 1 ml of water; cool and carefully
– spectrophotometer set at 220 nm, overlay with 3 ml of ferrous sulphate solution; a reddish brown
– a 20 µl loop injector. colour is produced at the interface of the two liquids.

909
PENTAERYTHRITOL TETRANITRATE TABLETS IP 2007

Tests of dry acetone and collect the filtrate. Repeat with two further
quantities, each of 5 ml, of dry acetone and evaporate the
Heavy metals (2.3.13). 2.0 g complies with limit test for heavy combined filtrate at a temperature not exceeding 60º, with the
metals, Method B (10 ppm). aid of a gentle current of air, and dry the residue at 60º for
Assay. Weigh accurately a quantity containing about 50 mg 4 hours; the residue melts at 138º to 142º (2.4.21).
of pentaerythritol tetranitrate and transfer to a 100-ml
volumetric flask with the aid of about 30 ml of acetone. Add Tests
sufficient acetone to produce 50 ml and warm on a water-bath
Uniformity of content. (For tablets containing 10 mg or less)
at a temperature not exceeding 60º and boil gently, with
— Comply with the test stated under Tablets.
occasional swirling, for 5 minutes. Cool, dilute to volume with
acetone and mix. Transfer a portion of the mixture to a glass- Crush one tablet and transfer to a 50-ml volumetric flask with
stoppered centrifuge tube and centrifuge at 1500 rpm for the aid of 15 ml of acetone. Add sufficient acetone to produce
5 minutes. Transfer 1.0 ml of the supernatant solution to a 25 ml, heat the mixture on a water-bath at a temperature not
100-ml volumetric flask and evaporate at 35º with the aid of a exceeding 60º and boil gently, with occasional swirling, for
current of air to dryness. To the residue add 1.0 ml of glacial 5 minutes. Cool, dilute to volume with acetone and mix.
acetic acid and swirl to dissolve. Add 2 ml of Transfer a portion of the mixture to a glass-stoppered
phenoldisulphonic acid solution, mix and allow to stand for centrifuge tube and centrifuge at 1500 rpm for 5 minutes.
5 minutes. Add 25 ml of water and 10 ml of strong ammonia Transfer 2.5 ml of the supernatant solution to a 100-ml
solution, cool, dilute to volume with water and mix. Measure volumetric flask and evaporate at 35º with the aid of a current
the absorbance of the resulting solution at the maximum at of air to dryness. To the residue add 1.0 ml of glacial acetic
about 409 nm (2.4.7), using water as the blank. acid and swirl to dissolve. Add 2 ml of phenoldisulphonic
acid solution, mix and allow to stand for 5 minutes. Add 25 ml
Weigh accurately 0.13 g of potassium nitrate, previously dried
of water and 10 ml of strong ammonia solution, cool, dilute to
at 105º for 4 hours, dissolve in 3 ml of water, dilute with
volume with water and mix. Measure the absorbance of the
sufficient glacial acetic acid to produce 200.0 ml and mix
resulting solution at the maximum at about 409 nm (2.4.7),
well. Using 1.0 ml of this solution repeat the procedure
using water as the blank.
beginning at the words “Add 2 ml of phenoldisulphonic acid
solution,.......”. Weigh accurately 0.130 g of potassium nitrate, previously
dried at 105º for 4 hours, dissolve in 3 ml of water, dilute with
Calculate the content of C5H8N4O12 from the values of the
sufficient glacial acetic acid to produce 200.0 ml and mix
absorbances so obtained.
well. Using 1.0 ml of this solution repeat the procedure
1 ml of the potassium nitrate solution is equivalent to beginning at the words “Add 2 ml of phenoldisulphonic acid
0.000503 g of C5H8N4O12. solution,.......”.Calculate the content of C5H8N4O12 in the tablet.
Storage. Store protected from light and moisture. 1 ml of the potassium nitrate solution is equivalent to 0.000503
NOTE — Undiluted pentaerythritol tetranitrate is a powerful g of C5H8N4O12.
explosive. It can be exploded with percussion or excessive Other tests. Complies with the tests stated under Tablets.
heat. Great care and appropriate precautions should be
taken in handling and only exceedingly small amounts should Assay. Weigh and powder 20 tablets. Weigh accurately a
be isolated. quantity of the powder containing about 50 mg of
pentaerythritol tetranitrate and transfer to a 100-ml volumetric
Labelling. The label states the percentage content of
flask with the aid of about 30 ml of acetone. Add sufficient
pentaerythritol tetranitrate.
acetone to produce 50 ml and warm on a water-bath at a
temperature not exceeding 60º and boil gently, with occasional
swirling, for 5 minutes. Cool, dilute to volume with acetone
Pentaerythritol Tetranitrate Tablets and mix. Transfer a portion of the mixture to a glass-stoppered
centrifuge tube and centrifuge at 1500 rpm for 5 minutes.
Pentaerythritol Tetranitrate Tablets contain not less than Transfer 1.0 ml of the supernatant solution to a 100-ml
90.0 per cent and not more than 110.0 per cent of the stated volumetric flask and evaporate at 35º with the aid of a current
amount of pentaerythritol tetranitrate, C5H8N4O12. of air to dryness. To the residue add 1.0 ml of glacial acetic
acid and swirl to dissolve. Add 2 ml of phenoldisulphonic
Identification acid solution, mix and allow to stand for 5 minutes. Add 25 ml
A. Transfer a quantity containing 10 mg of pentaerythritol of water and 10 ml of strong ammonia solution, cool, dilute to
tetranitrate to a medium porosity sintered-glass filter, add 5 ml volume with water and mix. Measure the absorbance of the

910
IP 2007 PENTAMIDINE INJECTION

resulting solution at the maximum at about 409 nm (2.4.7), Tests


using water as the blank.
pH (2.4.24). 4.5 to 6.5, determined in a 5.0 per cent w/v solution.
Weigh accurately 0.13 g of potassium nitrate, previously dried
Related substances. Determine by thin-layer chromatography
at 105º for 4 hours, dissolve in 3 ml of water, dilute with
(2.4.17), coating the plate with silica gel GF254.
sufficient glacial acetic acid to produce 200.0 ml and mix
well. Using Mobile phase. The upper layer obtained by shaking together
1.0 ml of this solution repeat the procedure beginning at the 50 volumes of water, 40 volumes of 1-butanol and 10 volumes
words “Add 2 ml of phenoldisulphonic acid solution,.......”. of glacial acetic acid.
Calculate the content of C5H8N4O12 from the values of the Test solution. Dissolve 0.5 g of the substance under
absorbances so obtained. examination in 10 ml of methanol.
1 ml of the potassium nitrate solution is equivalent to Reference solution. A 0.025 per cent w/v solution of the
0.000503 g of C5H8N4O12. substance under examination in 100 ml of methanol.

Storage. Store protected from light and moisture. Activate the plate by heating at 105º for 1 hour, apply to the
plate 10 µl of each solution. After development, dry the plate
Labelling. The label states the strength in terms of the in air and examine in ultraviolet light at 254 nm. Any secondary
equivalent amount of pentaerythritol tetranitrate. spot in the chromatogram obtained with the test solution is
not more intense than the spot in the chromatogram obtained
with the reference solution.
Pentamidine Isethionate Ammonium isethionate. To 1.0 g in a test-tube (about 4 cm in
diameter) add 10 ml of water and 20 ml of 1 M sodium
O O hydroxide. Immediately attach a bung carrying a splash head
SO3H and an aspirator tube (about 5 mm in diameter). Connect the
H2N NH2 . HO
2 splash head to two test-tubes in series, each containing 20 ml
NH NH of 0.01 M sulphuric acid. Heat the tube containing the
substance under examination in a water-bath at 45º to 50º and,
C19H24N4O2,2C2H6O4S Mol. Wt. 592.7 maintaining this temperature, draw a current of air, previously
Pentamidine Isethionate is 4,4′-[pentane-1,5- passed through 1 M sulphuric acid, through the liquids in a
diylbis(oxy)]dibenzamidine di(2-hydroxyethanesulphonate). series of tubes for 3 hours at such a rate that the bubbles are
just too rapid to count. Titrate the combined solutions from
Pentamidine Isethionate contains not less than 98.5 per cent
the two absorption tubes with 0.02 M sodium hydroxide using
and not more than 101.0 per cent of C19H24N4O2, 2C2H6O4S,
methyl red-methylene blue solution as indicator; not less than
calculated on the dried basis.
36.5 ml of 0.02 M sodium hydroxide is required.
Description. A white or almost white powder or crystals;
Sulphated ash (2.3.18). Not more than 0.1 per cent.
odourless or almost odourless; hygroscopic.
Loss on drying (2.4.19). Not more than 4.0 per cent, determined
Identification on 1.0 g by drying in an oven at 105º.
A. Determine by infrared absorption spectrophotometry (2.4.6). Assay. Dissolve 0.25 g in 50 ml of dimethylformamide. Titrate
Compare the spectrum with that obtained with pentamidine with 0.1 M tetrabutylammonium hydroxide, determining the
isethionate RS or with the reference spectrum of pentamidine end-point potentiometrically (2.4.25). Carry out a blank titration.
isethionate. 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
B. When examined in the range 230 nm to 360 nm (2.4.7), a 0.02963 g of C19H24N4O2,2C2H6O4S.
0.001 per cent w/v solution in 0.01 M hydrochloric acid shows Storage. Store protected from moisture.
an absorption maximum only at about 262 nm; absorbance at
about 262 nm, about 0.46).
C. To 10 ml of a 0.05 per cent w/v solution add 1 ml of a 0.1 per Pentamidine Injection
cent w/v solution of glyoxal sodium bisulphite and 1 ml of a
solution prepared by dissolving 4 g of boric acid in a mixture Pentamidine Isethionate Injection
of 27 ml of 1 M sodium hydroxide and sufficient water to Pentamidine Injection is a sterile material consisting of
produce 100 ml. Heat on a water-bath for 10 minutes; a magenta Pentamidine Isethionate with or without buffering agents and
colour is produced. other excipients. It is filled in a sealed container.

911
PENTAZOCINE IP 2007

The injection is constituted by dissolving the contents of the not more intense than the spot in the chromatogram obtained
sealed container in the requisite amount of sterile Water for with the reference solution.
Injections, immediately before use. Ammonium isethionate. To 1.0 g in a test-tube (about 4 cm in
The constituted solution complies with the requirements for diameter) add 10 ml of water and 20 ml of 1 M sodium
Clarity of solution and Particulate matter stated under hydroxide. Immediately attach a bung carrying a splash head
Parenteral Preparations (Injections). and an aspirator tube (about 5 mm in diameter). Connect the
splash head to two test-tubes in series, each containing 20 ml
Storage. The constituted solution should be used immediately
of 0.01 M sulphuric acid. Heat the tube containing the
after preparation but, in any case, within the period
substance under examination in a water-bath at 45º to 50º and,
recommended by the manufacturer.
maintaining this temperature, draw a current of air, previously
Pentamidine Injection contains not less than 95.0 per cent and passed through 1 M sulphuric acid, through the liquids in a
not more than 105.0 per cent of the stated amount of series of tubes for 3 hours at such a rate that the bubbles are
pentamidine isethionate, C19H24N4O2, 2C2H6O4S. just too rapid to count. Titrate the combined solutions from
The contents of the sealed container comply with the the two absorption tubes with 0.02 M sodium hydroxide using
requirements stated under Parenteral Preparations methyl red-methylene blue solution as indicator; not less than
(Powders for Injection) and with the following requirements. 36.5 ml of 0.02 M sodium hydroxide is required.
Assay. Dissolve 0.25 g of the mixed contents of 10 containers
Identification in 50 ml of dimethylformamide. Titrate with 0.1 M
tetrabutylammonium hydroxide, determining the end-point
A. Determine by infrared absorption spectrophotometry (2.4.6).
potentiometrically (2.4.25). Carry out a blank titration.
Compare the spectrum with that obtained with pentamidine
isethionate RS or with the reference spectrum of pentamidine 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
isethionate. 0.02963 g of C19H24N4O2, 2C2H6O4S.
B. When examined in the range 230 nm to 360 nm (2.4.7), a Storage. Store in single dose containers.
0.001 per cent w/v solution in 0.01 M hydrochloric acid shows
an absorption maximum only at about 262 nm; absorbance at
about 262 nm, about 0.46.
Pentazocine
C. To 10 ml of a 0.05 per cent w/v solution add 1 ml of a 0.1 per
cent w/v solution of glyoxal sodium bisulphite and 1 ml of a
solution prepared by dissolving 4 g of boric acid in a mixture HO
of 27 ml of 1 M sodium hydroxide and sufficient water to
produce 100 ml. Heat on a water-bath for 10 minutes; a magenta
colour is produced.
H3C N CH3
Tests CH3
CH3
pH (2.4.24). 4.5 to 6.5, determined in a 5.0 per cent w/v solution.
Related substances. Determine by thin-layer chromatography C19H27NO Mol. Wt. 285.4
(2.4.17), coating the plate with silica gel GF254. Pentazocine is (2RS,6RS,11RS)-6,11-dimethyl-3-(3-methylbut-
Mobile phase. The upper layer obtained by shaking together 2-enyl)-1,2,3,4,5,6-hexahydro-2,6-methano-3-benzazocin-8-ol.
50 volumes of water, 40 volumes of 1-butanol and 10 volumes Pentazocine contains not less than 98.0 per cent and not more
of glacial acetic acid. than 101.0 per cent of C19H27NO, calculated on the dried basis.
Test solution. Dissolve 0.5 g of the substance under Description. A white or pale cream powder.
examination in 10 ml of methanol.
Identification
Reference solution. A 0.025 per cent w/v solution of the
substance under examination in 100 ml of methanol. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with pentazocine
Activate the plate by heating at 105º for 1 hour, apply to the
RS or with the reference spectrum of pentazocine.
plate 10 µl of each solution. After development, dry the plate
in air and examine in ultraviolet light at 254 nm. Any secondary B. To 1 mg in a porcelain crucible add 0.5 ml of a solution of
spot in the chromatogram obtained with the test solution is sulphuric acid containing 1 per cent w/v solution of

912
IP 2007 PENTAZOCINE HYDROCHLORIDE

ammonium molybdate; an intense blue colour is produced Pentazocine Hydrochloride


which changes to bluish green, green and finally, on standing,
yellow.
HO
C. Dissolve 5 mg in 5 ml of sulphuric acid, add 0.05 ml of ferric
chloride solution and mix; a yellow colour is produced which
deepens slightly in intensity on warming. On the addition of , HCl
0.05 ml of nitric acid the yellow colour is unchanged. N CH3
H3C
Tests CH3
CH3
Light absorption (2.4.7). To 0.1 g add 20 ml of water and 10 ml
of 1 M hydrochloric acid, shake to dissolve and add sufficient C19H27NO,HCl Mol. Wt. 321.9
water to produce 100 ml. Dilute 10 ml to 100 ml with water. The
absorbance of the resulting solution at the maximum at about Pentazocine Hydrochloride is (2RS,6RS,11RS)-6,11-
278 nm, 0.67 to 0.71. dimethyl-3-(3-methylbut-2-enyl)-1,2,3,4,5,6-hexahydro-2,6-
methano-3-benzazocin-8-ol hydrochloride.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel HF254. Pentazocine Hydrochloride contains not less than 98.0 per
cent and not more than 101.0 per cent of C19H27NO,HCl,
Mobile phase. A mixture of 94 volumes of chloroform, calculated on the dried basis.
3 volumes of 2-propylamine and 3 volumes of methanol.
Description. A white or pale cream-coloured, crystalline
Test solution. Dissolve 0.2 g of the substance under powder; odourless. The material exhibits polymorphism.
examination in 10 ml of chloroform.
Reference solution (a). A 0.02 per cent w/v solution of the Identification
substance under examination in chloroform.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution (b). A 0.01 per cent w/v solution of the Compare the spectrum with that obtained with pentazocine
substance under examination in chloroform. hydrochloride RS or with the reference spectrum of
Reference solution (c). A 0.005 per cent w/v solution of the pentazocine hydrochloride.
substance under examination in chloroform. B. To 1 mg in a porcelain crucible add 0.5 ml of a solution of
Apply to the plate 10 µl of each solution. After development, sulphuric acid containing 1 per cent w/v solution of
dry the plate in air and examine in ultraviolet light at 254 nm. ammonium molybdate; an intense blue colour is produced
Heat the plate at 105º for 15 minutes, allow to cool, expose to which changes to bluish green, green and finally, on standing,
iodine vapour and re-examine in ultraviolet light at 254 nm. yellow.
Any secondary spot in the chromatogram obtained with the C. Gives reaction A of chlorides (2.3.1).
test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a); not more Tests
than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b) and not pH (2.4.24). 4.0 to 6.0, determined in a 1.0 per cent w/v solution.
more than four such spots are more intense than the spot in Light absorption (2.4.7). Dissolve 0.1 g in 10 ml of 1 M
the chromatogram obtained with reference solution (c). hydrochloric acid and add sufficient water to produce
Sulphated ash (2.3.18). Not more than 0.1 per cent. 100 ml; dilute 10 ml to 100 ml with water. Absorbance of the
resulting solution at the maximum at about 278 nm, 0.59 to
Loss on drying (2.4.19). Not more than 1.0 per cent, determined 0.63.
on 1.0 g by drying in an oven at 60º at a pressure not exceeding
0.7 kPa for 4 hours. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel HF254.
Assay. Weigh accurately about 0.6 g, dissolve in 50 ml of
anhydrous glacial acetic acid and titrate with 0.1 M perchloric Mobile phase. A mixture of 94 volumes of chloroform,
acid, using crystal violet solution as indicator. Carry out a 3 volumes of 2-propylamine and 3 volumes of methanol.
blank titration. Test solution. Dissolve 0.2 g of the substance under
1 ml of 0.1 M perchloric acid is equivalent to 0.02854 g of examination in 10 ml of chloroform.
C19H27NO. Reference solution (a). A 0.02 per cent w/v solution of the
Storage. Store protected from light and moisture. substance under examination in chloroform.

913
PENTAZOCINE TABLETS IP 2007

Reference solution (b). A 0.01 per cent w/v solution of the B. To a quantity of the powdered tablets containing 50 mg of
substance under examination in chloroform. Pentazocine Hydrochloride add 70 ml of water, shake for
Reference solution (c). A 0.005 per cent w/v solution of the 15 minutes, add sufficient water to produce 100 ml and filter.
substance under examination in chloroform. To 10 ml of the filtrate add 10 ml of 1 M sodium hydroxide and
sufficient water to produce 100 ml. When examined in the
Apply to the plate 10 µl of each solution. After development, range 230 nm to 360 nm (2.4.7), the resulting solution shows
dry the plate in air and examine in ultraviolet light at 254 nm. absorption maxima at about 238 nm and 298 nm.
Heat the plate at 105º for 15 minutes, allow to cool, expose to
C. Shake a quantity of the powdered tablets containing 25 mg
iodine vapour and re-examine in ultraviolet light at 254 nm.
of Pentazocine Hydrochloride with 5 ml of water and 0.5 ml of
Any secondary spot in the chromatogram obtained with the
2 M nitric acid for 1 minute and filter. The filtrate gives reaction
test solution is not more intense than the spot in the
A of chlorides (2.3.1).
chromatogram obtained with reference solution (a); not more
than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b) and not
Tests
more than four such spots are more intense than the spot in Related substances. Determine by thin-layer chromatography
the chromatogram obtained with reference solution (c). (2.4.17), coating the plate with silica gel HF254.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Mobile phase. A mixture of 94 volumes of chloroform,
Loss on drying (2.4.19). Not more than 1.0 per cent, determined 3 volumes of 2-propylamine and 3 volumes of methanol.
on 1.0 g by drying in an oven at 100º at a pressure not exceeding Test solution. Shake a quantity of the powdered tablets
0.7 kPa. containing 0.2 g of Pentazocine Hydrochloride with 10 ml of
Assay. Weigh accurately about 0.5 g, dissolve in 50 ml of 0.1 M methanolic ammonia for 10 minutes, centrifuge and
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric use the supernatant liquid.
acid, using crystal violet solution as indicator. Carry out a Reference solution (a). Dilute 1 volume of test solution to
blank titration. 100 volumes with the same solvent.
1 ml of 0.1 M perchloric acid is equivalent to 0.03219 g of Reference solution (b). Dilute 1 volume of test solution to
C19H27NO,HCl. 200 volumes with the same solvent.
Storage. Store protected from light and moisture. Reference solution (c). Dilute 1 volume of test solution to
400 volumes with the same solvent.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Pentazocine Tablets Heat the plate at 105º for 15 minutes, allow to cool, expose to
Pentazocine Hydrochloride Tablets iodine vapour and re-examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the
Pentazocine Tablets contain not less than 92.5 per cent and test solution is not more intense than the spot in the
not more than 107.5 per cent of the stated amount of chromatogram obtained with reference solution (a), not more
pentazocine hydrochloride, C19H27NO,HCl. than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b) and not
Identification
more than four such spots are more intense than the spot in
A. Shake a quantity of the powdered tablets containing 0.1 g the chromatogram obtained with reference solution (c).
of Pentazocine Hydrochloride with 10 ml of water, filter, add Other tests. Complies with the tests stated under Tablets.
1 ml of 1 M sodium hydroxide and shake the resulting solution
with 20 ml of chloroform. Wash the chloroform extract with Assay. Weigh and powder 20 tablets. Weigh accurately a
5 ml of water, dry over anhydrous sodium sulphate and filter. quantity of the powder containing about 25 mg of Pentazocine
Evaporate the chloroform using a rotary evaporator and dry Hydrochloride, shake with 100 ml of water for 15 minutes, add
the oily residue at a temperature not exceeding 25º at a pressure 2.5 ml of 1 M hydrochloric acid and sufficient water to
of 2 kPa for 1 hour. produce 250.0 ml and filter. Measure the absorbance of the
filtrate at the maximum at about 278 nm (2.4.7). Calculate the
On the residue, determine by infrared absorption
content of C19H27NO,HCl taking 61.2 as the specific absorbance
spectrophotometry (2.4.6). Compare the spectrum with that
at 278 nm.
obtained with pentazocine RS or with the reference spectrum
of pentazocine. Storage. Store protected from light and moisture.

914
IP 2007 PENTAZOCINE INJECTION

Pentazocine Lactate Reference solution (b). A 0.01 per cent w/v solution of the
substance under examination in chloroform.
HO Reference solution (c). A 0.005 per cent w/v solution of the
substance under examination in chloroform.
Apply to the plate 25 µl of each solution. After development,
COOH
, dry the plate in air and examine in ultraviolet light at 254 nm.
H3C N CH3 Heat the plate at 105º for 15 minutes, allow to cool, expose to
H3C OH
CH3 iodine vapour and re-examine in ultraviolet light at 254 nm.
CH3 Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the
C19H27NO,C3H6O3 Mol. Wt. 375.4 chromatogram obtained with reference solution (a); not more
than one such spot is more intense than the spot in the
Pentazocine Lactate is (2RS,6RS,11RS)-6,11-dimethyl-3-(3-
chromatogram obtained with reference solution (b) and not
methylbut-2-enyl)-1,2,3,4,5,6-hexahydro-2,6-methano-3-
more than four such spots are more intense than the spot in
benzazocin-8-ol lactate.
the chromatogram obtained with reference solution (c).
Pentazocine Lactate contains not less than 98.5 per cent and
Sulphated ash (2.3.18). Not more than 0.1 per cent.
not more than 101.0 per cent of C19H27NO,C3H6O3, calculated
on the dried basis. Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 60º at a pressure not exceeding
Description. A white or pale cream-coloured, crystalline
0.7 kPa for 16 hours.
powder; odourless.
Assay. Weigh accurately about 0.75 g of the substance under
Identification examination, dissolve in 50 ml of anhydrous glacial acetic
acid. Titrate with 0.1 M perchloric acid, using crystal violet
A. Determine by infrared absorption spectrophotometry (2.4.6).
solution as indicator. Carry out a blank titration.
Compare the spectrum with that obtained with pentazocine
lactate RS or with the reference spectrum of pentazocine 1 ml of 0.1 M perchloric acid is equivalent to 0.03754 g of
lactate. C19H27NO,C3H6O3.
B. To 1 mg in a porcelain crucible add 0.5 ml of a solution of Storage. Store protected from light and moisture.
sulphuric acid containing 1 per cent w/v solution of
ammonium molybdate; an intense blue colour is produced
which changes to bluish green, green and finally, on standing,
yellow.
Pentazocine Injection
C. Gives reaction A of lactates (2.3.1).
Pentazocine Lactate Injection
Tests Pentazocine Injection is a sterile solution in Water for
pH (2.4.24). 5.5 to 6.5, determined in a 1.0 per cent w/v solution. Injections of either Pentazocine Lactate or pentazocine lactate
prepared by the interaction of Pentazocine and Lactic Acid.
Light absorption (2.4.7). Dissolve 0.1 g in 10 ml of 1 M
hydrochloric acid and add sufficient water to produce Pentazocine Injection contains not less than 95.0 per cent and
100 ml; dilute 10 ml to 100 ml with water. Absorbance of the not more than 105.0 per cent of the stated amount of
resulting solution at the maximum at about 278 nm, 0.50 to pentazocine, C19H27NO.
0.54. Description. A clear, colourless or almost colourless liquid.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel HF254. Identification
Mobile phase. A mixture of 94 volumes of chloroform, A. To a volume containing 90 mg of pentazocine add 5 ml of
3 volumes of 2-propylamine and 3 volumes of methanol. 0.1 M sodium hydroxide and shake the resulting solution
with 5 ml of chloroform. Wash the chloroform extract with 2 ml
Test solution. Dissolve 0.2 g of the substance under
of water, dry over anhydrous sodium sulphate and filter.
examination in 10 ml of chloroform.
Evaporate the chloroform without applying heat and dry the
Reference solution (a). A 0.02 per cent w/v solution of the oily residue at a temperature not exceeding 25º and at a pressure
substance under examination in chloroform. of 2 kPa for 1 hour.

915
PENTOBARBITONE SODIUM IP 2007

On the residue, determine by infrared absorption 100.0 ml. To 5.0 ml add 1 ml of 1 M hydrochloric acid and
spectrophotometry (2.4.6). Compare the spectrum with that sufficient water to produce 100.0 ml. Measure the absorbance
obtained with pentazocine RS or with the reference spectrum of the resulting solution at the maximum at about 278 nm (2.4.7).
of pentazocine (form B). Calculate the content of C19H27NO, taking 69 as the specific
absorbance at 278 nm.
B. Dilute a volume containing 30 mg of pentazocine to 100 ml
with water. To 10 ml add 10 ml of 1 M sodium hydroxide and Storage. Store protected from light.
sufficient water to produce 100 ml. When examined in the
Labelling. The label states the strength in terms of the
range 230 nm to 360 nm (2.4.7), the solution shows absorption
equivalent amount of pentazocine in a suitable dose-volume.
maxima, at about 238 nm and 298 nm.
C. To a volume containing 30 mg of pentazocine add 2 ml of
0.1 M sodium hydroxide, extract with 2 ml of chloroform and
evaporate 0.1 ml of the chloroform extract to dryness in a
porcelain crucible. Add to the residue 0.5 ml of a 1 per cent
Pentobarbitone Sodium
w/v solution of ammonium molybdate in sulphuric acid; an Soluble Pentobarbitone; Pentobarbital Sodium
intense blue colour is produced which changes to bluish green,
green and finally, on standing, yellow.
H
O N ONa
Tests
H3C
pH (2.4.24). 4.0 to 5.0. N

Related substances. Determine by thin-layer chromatography H3C H3C O


(2.4.17), coating the plate with silica gel HF254.
Mobile phase. A mixture of 94 volumes of chloroform, C11H17N2NaO3 Mol. Wt. 248.3
3 volumes of 2-propylamine and 3 volumes of methanol.
Pentobarbitone Sodium is sodium 5-ethyl-5-[(1RS)-1-
Test solution. Dilute a volume of the injection with sufficient methylbutyl]barbiturate.
ethanol (95 per cent) to produce a solution containing
2.0 per cent w/v solution of pentazocine. Pentobarbitone Sodium contains not less than 99.0 per cent
and not more than 101.5 per cent of C11H17N2NaO3, calculated
Reference solution (a). Dilute 1 volume of the test solution to on the dried basis.
100 volumes with ethanol (95 per cent).
Description. A white, crystalline powder or granules;
Reference solution (b). Dilute 1 volume of the test solution to hygroscopic.
200 volumes with ethanol (95 per cent).
Reference solution (c). Dilute 1 volume of the test solution to Identification
400 volumes with ethanol (95 per cent). A. To 10 ml of a 10 per cent w/v solution add 5 ml of 2 M acetic
Apply to the plate 10 µl of each solution. After development, acid; a white, crystalline precipitate is produced. Filter, wash
dry the plate in air and examine in ultraviolet light at 254 nm. the precipitate with water and dry at 105º. Determine the
Heat the plate at 105º for 15 minutes, allow to cool, expose to melting point of the dried precipitate (2.4.21). Mix equal parts
iodine vapour and re-examine in ultraviolet light at 254 nm. of the dried precipitate and phenobarbitone RS and determine
Any secondary spot in the chromatogram obtained with the the melting point (2.4.21). The difference between the melting
test solution is not more intense than the spot in the points (which are about 131º) is not greater than 2º.
chromatogram obtained with reference solution (a), not more B. Complies with the test for identification of barbiturates
than one such spot is more intense than the spot in the (2.3.1), using the dried precipitate obtained in test A for
chromatogram obtained with reference solution (b) and not preparing the test solution.
more than four such spots are more intense than the spot in
the chromatogram obtained with reference solution (c). C. To 10 mg add 10 mg of vanillin and 2 ml of sulphuric acid,
mix and heat on a water-bath for 2 minutes; a reddish-brown
Other tests. Complies with the tests stated under Parenteral colour is produced. Cool and add 5 ml of ethanol; the colour
Preparations (Injections). changes to violet and then blue.
Assay. To an accurately measured volume containing about D. Ignite 1 g; the residue gives reaction A of sodium salts
0.15 g of pentazocine add sufficient water to produce (2.3.1).

916
IP 2007 PENTOBARBITONE TABLETS

Tests Pentobarbitone Tablets


Appearance of solution. Prepare freshly a 10.0 per cent w/v Pentobarbitone Sodium Tablets; Pentobarbital Sodium
solution in carbon dioxide-free water (solution A). Solution Tablets
A is clear (2.4.1).
Pentobarbitone Tablets contain not less than 92.5 per cent
pH (2.4.24). 9.6 to 11.0, determined in solution A. and not more than 107.5 per cent of the stated amount of
Related substances. Complies with the test for related pentobarbitone sodium, C11H17N2NaO3.
substances in barbiturates (2.3.4), but applying 10 µl of each
of the following solutions. Identification
Test solution. A 2.0 per cent w/v solution of the substance A. Shake a quantity of the powdered tablets containing 0.1 g
under examination. of Pentobarbitone Sodium with 10 ml of a 10 per cent w/v
solution of pyridine and filter. Add to the filtrate 1 ml of cupric
Reference solution. A 0.01 per cent w/v solution of the
sulphate with pyridine solution and set aside for 10 minutes;
substance under examination.
a reddish violet precipitate is produced.
Do not ignore any spot remaining on the line of application.
B. Shake a quantity of the powdered tablets containing 0.1 g
Free pentobarbitone. Not more than 3.5 per cent, determined of Pentobarbitone Sodium with 10 ml of water and filter. To
by the following method. Weigh accurately about 2.0 g and the filtrate add 2 ml of hydrochloric acid; a white precipitate
dissolve in 75 ml of dimethylformamide, heating gently if is produced (distinction from pentobarbitone).
necessary. Add 0.25 ml of a 1 per cent w/v solution of thymol C. The residue obtained in the Assay melts at 127º to
blue in dimethylformamide and titrate with 0.1 M sodium 130º (2.4.21).
methoxide to a blue end-point. Carry out a blank titration.
D. The powdered tablets, when moistened with hydrochloric
1 ml of 0.1 M sodium methoxide is equivalent to 0.02263 g of acid and introduced on a platinum wire into a flame, impart a
pentobarbitone. yellow colour to the flame.
Isomer. Dissolve 0.3 g in 5 ml of a 5 per cent w/v solution of
anhydrous sodium carbonate and add 0.3 g of 4-nitrobenzyl Tests
bromide dissolved in 10 ml of ethanol (95 per cent). Heat Isomer. Dissolve a quantity of the powdered tablets containing
under a reflux condenser for 30 minutes, cool to 25º scratch 0.3 g of Pentobarbitone Sodium in 5 ml of a 5 per cent w/v
the side of the vessel with a glass rod if necessary to induce solution of anhydrous sodium carbonate and add 0.3 g of
crystallisation and filter. Wash the residue with five quantities, 4-nitrobenzyl bromide dissolved in 10 ml of ethanol (95 per
each of 5 ml, of water. Transfer the residue as completely as cent). Heat under a reflux condenser for 30 minutes, cool to
possible to a small flask, add 25 ml of ethanol (95 per cent) 25º scratch the side of the vessel with a glass rod if necessary
and heat under a reflux condenser for 10 minutes; the solid to induce crystallisation and filter. Wash the residue with five
dissolves completely. Cool to 25º and scratch the side of the quantities, each of 5 ml, of water. Transfer the residue as
flask with a glass rod to induce crystallisation. Filter, wash the completely as possible to a small flask, add 25 ml of ethanol
residue with two quantities, each of 5 ml, of water and dry at (95 per cent) and heat under a reflux condenser for 10 minutes;
105º for 30 minutes. The dried residue melts at 136º to 148º filter the hot solution. Cool to 25º and scratch the side of the
(2.4.21). flask with a glass rod to induce crystallisation. Filter, wash the
Heavy metals (2.3.13). 0.67 g complies with the limit test for residue with two quantities, each of 5 ml, of water and dry at
heavy metals, Method B (30 ppm). 105º for 30 minutes. The dried residue melts at 136º to 148º
(2.4.21).
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
on 1.0 g by drying in an oven at 105º. Other tests. Complies with the tests stated under Tablets.
Assay. Weigh accurately about 0.4 g, dissolve in 25 ml of a Assay. Weigh and powder 20 tablets. Weigh accurately a
12.75 per cent w/v solution of silver nitrate in pyridine and quantity of the powder containing about 0.3 g of
titrate with 0.1 M ethanolic sodium hydroxide using 0.5 ml of Pentobarbitone Sodium, dissolve as completely as possible
thymolphthalein solution as indicator, until a pure blue colour in 10 ml of a 2 per cent w/v solution of sodium hydroxide,
is obtained. Carry out a blank titration. saturate with sodium chloride, acidify with hydrochloric acid
and extract with successive quantities, each of 15 ml, of ether
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to until complete extraction is effected. Wash the combined
0.02483 g of C11H17N2NaO3. extracts with two quantities, each of 2 ml, of water and extract
Storage. Store protected from moisture. the combined washings with 10 ml of ether. Add the ether to

917
PEPSIN IP 2007

the main ether layer, filter and wash the filter with ether. 65 ml of 1 M hydrochloric acid to 1000 ml with water, continue
Evaporate the solvent and dry the residue to constant weight the trituration, dilute to 1000.0 ml with the acidified water and
at 105º. shake for 15 minutes. Prepare coagulated egg albumin by
1 g of residue is equivalent to 1.097 g of C11H17N2NaO3. boiling fresh hen-eggs in water for 15 minutes, cooling rapidly
to room temperature by immersion in cold water, separating
Storage. Store protected from moisture. the whites and rubbing through a no. 44 sieve. Reject the first
portion that passes through the sieve and triturate 15.0 g of
freshly prepared coagulated egg albumin with 50 ml of the
Pepsin acidified water ensuring that the particles of egg albumin are
thoroughly disintegrated, add a further 50 ml of the acidified
Pepsin is obtained from the gastric mucosa of pigs, cattle or water and keep in a water-bath at 51º ± 1º for 15 minutes. Add
sheep. It contains gastric proteinases that are active in an 20.0 ml of the prepared solution of the substance under
acid medium, pH 1 to 5. It may contain a suitable diluent such examination and digest at 51º ± 1º for 4 hours, shaking at
as Lactose. intervals of 15 minutes. Centrifuge and decant off most of the
Pepsin has an activity equivalent to its ability to digest not clear supernatant liquid, wash the remainder into a 10-ml
less than 3000 times its weight of coagulated egg albumin graduated cylinder and allow to stand for 30 minutes. The
when determined by the method given under Assay. volume of the undissolved albumin is not more than 2 ml.
Description. A white or light buff-coloured, crystalline or Storage. Store protected from moisture.
amorphous powder or translucent scales; odour, faint and
meaty but not rancid; hygroscopic.

Identification Peritoneal Dialysis Solutions


A. Place 1 ml of congo red fibrin on a filter paper and wash Intraperitoneal Dialysis Fluids
until a colourless filtrate is obtained with a solution prepared
by diluting 30 ml of 1 M hydrochloric acid to 1000 ml with Peritoneal Dialysis Solutions are sterile preparations for
water and adjusting the pH 1.5 to 1.7. Perforate the filter paper intraperitoneal use containing electrolytes with a concentration
and wash the congo red fibrin through it with 20 ml of the close to the electrolytic composition of plasma. They contain
same hydrochloric acid solution. Shake this suspension before dextrose in varying concentrations and/or other suitable
use. Dissolve about 10 mg of the substance under examination osmotic agents. They do not contain antioxidants such as
in 2 ml of the hydrochloric acid solution and adjust the pH 1.5 metabisulphite salts.
to 1.7. Place 4 ml of the congo red fibrin suspension in each of Peritoneal Dialysis Solutions contain not less than 97.5 per
two tubes. To one of the tubes add 1 ml of the solution of the cent and not more than 102.5 per cent of the stated amount of
substance under examination and to the other tube add 1 ml of sodium, Na, not less than 95.0 per cent and not more than
water (control solution). Mix the contents of each tube and 105.0 per cent of the stated amounts of potassium, K, calcium,
place in a water-bath at 25º with gentle shaking for 15 minutes; Ca, magnesium, Mg, chloride, Cl, acetate, C2H3O2, lactate,
the control solution is colourless and the solution of the C3H5O3, sodium bicarbonate, NaHCO3, bicarbonate, HCO3 and
substance under examination is violet blue. dextrose, C6H12O6.
B. The proteolytic activity of a solution in water is destroyed Description. Clear, colourless or faintly straw-coloured
at once by boiling. It is destroyed by warming for 10 minutes solutions.
at 40º at a pH of 8.0.
Identification
Tests
A. To 5 ml of the solution under examination, add 2 ml of
Microbial contamination (2.2.9). 1 g is free from Escherichia
dilute sodium hydroxide solution and 0.05 ml of potassium
coli and 10 g is free from salmonellae.
cupri-tartrate solution; the solution remains blue and clear.
Sulphated ash (2.3.18). Not more than 5.0 per cent. Heat to boiling; a copious red precipitate is formed.
Loss on drying (2.4.19). Not more than 5.0 per cent, determined B. 20 ml gives reactions of chlorides, sodium salts, potassium
on 1.0 g by drying in an oven at 60º at a pressure not exceeding salts and calcium salts (2.3.1).
0.7 kPa for 4 hours.
C. To 5 ml add 1 ml of hydrochloric acid in a test-tube fitted
Assay. Weigh accurately 0.25 g, triturate with 1.0 g of sodium with a stopper and a bent tube, heat and collect a few ml of the
chloride, add slowly acidified water prepared by diluting distillate. The distillate gives reaction C of acetates (2.3.1).

918
IP 2007 PERITONEAL DIALYSIS SOLUTIONS

D. To 0.1 ml of titan yellow solution add 10 ml of water, 2 ml of Pyrogens (2.2.8). Solutions for which a validated test for
the solution under examination and 1 ml of 1 M sodium bacterial endotoxins cannot be carried out, comply with the
hydroxide; a pink colour is produced if magnesium salts are test for pyrogens, injecting 10 ml of the solution per kg of the
present. rabbit’s body weight.
E. Lactates and bicarbonates are identified together with the Sterility (2.2.11). Complies with the test for sterility.
Assay for lactate and bicarbonate. Assay. For sodium — Dilute suitably with water and determine
by Method A for flame photometry (2.4.4), or by Method A for
Tests atomic absorption spectrophotometry (2.4.2), measuring at
589 nm and using sodium solution FP, or sodium solution
Appearance of solution. The solution under examination is
AAS respectively, suitably diluted with water for the standard
clear (2.4.1), and not more intensely coloured than reference
solutions.
solution YS4 (2.4.1).
For potassium — Dilute suitably with water and determine
pH (2.4.24). 4.5 to 6.5. If the solution contains bicarbonate, 6.5 by Method A for flame photometry (2.4.4), or by Method A for
to 8.0. atomic absorption spectrophotometry (2.4.2), measuring at
5-Hydroxymethylfurfural and Related substances. Dilute a 767 nm and using potassium solution FP or potassium solution
volume containing 1.0 g of Dextrose to 250.0 ml with water AAS respectively, suitably diluted with water for the standard
and measure the absorbance of the resulting solution (2.4.7) solutions.
at the maximum at about 284 nm; absorbance at about 284 nm, For calcium — Dilute suitably with water and determine by
not more than 0.25. Method A for flame photometry (2.4.4), or by Method A for
Aluminium. Adjust the pH of 400 ml of the solution under atomic absorption spectrophotometry (2.4.2), measuring at
examination to pH 6.0 and add 10 ml of acetate buffer pH 6.0. 422.7 nm and using calcium solution FP or calcium solution
Extract the resulting solution with successive quantities of AAS respectively, suitably diluted with water for the standard
20, 20 and 10 ml of a 0.5 per cent w/v solution of solutions.
8-hydroxyquinoline in chloroform and dilute the combined For magnesium — To 50.0 ml add 50 ml of water and 5 ml of
extracts to 50.0 ml with chloroform. Use as the blank a mixture strong ammonia-ammonium chloride solution and titrate with
of 10 ml of acetate buffer pH 6.0 and 100 ml of water treated in 0.005 M disodium edetate using 50 mg of eriochrome black
the same manner and as the standard solution a mixture of T mixture as indicator.
2.0 ml of aluminium standard solution (2 ppm Al), 10 ml of 1 ml of 0.005 M disodium edetate is equivalent to 0.1215 mg
acetate buffer pH 6.0 and 90 ml of water treated in the same of Mg.
manner. Measure the fluorescence of the test solution (I1), of
For total chloride — Dilute an accurately measured volume
the standard solution (I2) and of the blank (I3), (2.4.5), using an
containing about 60 mg of chloride to 50.0 ml with water. Add
excitation wavelength of 392 nm and a secondary filter with a
25.0 ml of 0.1 M silver nitrate and 2 ml of nitric acid. Filter,
transmission band centred at 518 nm, or a monochromator set
wash the precipitate with water slightly acidified with nitric
to transmit at this wavelength. The fluorescence of the test
acid and titrate the excess of silver nitrate with 0.1 M
solution (I1 – I3) is not greater than that of the standard solution
ammonium thiocyanate using ferric ammonium sulphate
(I2 – I3).
solution as indicator until a reddish yellow colour is produced.
Particulate contamination (2.5.9). Carry out the test using Carry out a blank titration.
50 ml of the solution under examination. 1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total
The preparation meets the requirements of the test if it contains chloride, calculated as Cl.
particles within the maximum limits shown below. For acetate (if present) — Determine by liquid
chromatography (2.4.14).
Particle size in µm Maximum number
(Equal to or larger than) of particles per ml Test solution. Dilute an accurately measured volume of the
preparation under examination quantitatively with water to
10 25 obtain a solution containing about 1.0 mg of acetate per ml.
25 3 Reference solution. Dissolve an accurately weighed quantity
of sodium acetate in water to obtain a solution having a known
Other tests. Complies with the tests stated under Parenteral concentration of about 0.12 per cent w/v of Sodium Acetate.
Preparations (Injections).
Chromatographic system
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin – a stainless steel column 30 cm x 7.8 mm, packed with a
Unit per ml. strong cation-exchange resin consisting of sulphonated

919
PERITONEAL DIALYSIS SOLUTIONS IP 2007

cross-linked styrene-divinylbenzene copolymer in the 1 ml of 0.1 M hydrochloric acid is equivalent to 8.40 mg of


hydrogen form (7 µm), NaHCO3.
– mobile phase: filtered and degassed 0.1 M sulphuric For lactate and bicarbonate — Determine by liquid
acid, chromatography (2.4.14).
– flow rate. 0.8 ml per minute,
Test solution. Use the preparation under examination.
– column temperature. 60º,
– spectrophotometer set at 210 nm, Reference solutions. Dissolve accurately weighed quantities
– a 20 µl loop injector. of lactates and bicarbonates in order to obtain solutions having
concentrations of about 90 per cent, 100 per cent and 110 per
Inject the reference solution and record the chromatograms. cent of the claim in 100 ml of water.
The test is not valid unless the tailing factor is not more than
2.0 and the relative standard deviation for replicate injections Chromatographic system
is not more than 2.0 per cent. – a stainless steel column 30 cm x 7.8 mm, packed with a
cation-exchange resin (9 µm),
Inject separately the test and standard solutions and record – column. temperature 85º,
the chromatograms. Measure the responses for the major peak – mobile phase: filtered and degassed 0.005 M sulphuric
and calculate the content of acetate in the preparation under acid,
examination. – flow rate. 0.6 ml per minute,
For lactate (if present) — Determine by liquid chromatography – differential refractometer detector,
(2.4.14). – a 20 µl loop injector.
Test solution. Use the preparation under examination. Inject separately the test solution and the reference solutions
in duplicate, and record the chromatograms in the prescribed
Reference solution(a). Dissolve an accurately weighed conditions. The peaks elute in the following order, lactates,
quantity of sodium lactate RS in water to obtain a solution then bicarbonates. Determine the concentration of lactates
having a known concentration of about 2 mg per ml. and bicarbonates in the test solution by interpolating the peak
Reference solution (b). Prepare a solution in water containing area for lactate and the peak height for bicarbonate from the
about 3 mg of anhydrous sodium acetate and 3 mg of sodium linear regression curve obtained with the solutions prepared
lactate RS per ml. as reference solutions.
Chromatographic system For dextrose — Transfer a volume of the preparation under
– a stainless steel column 10 cm x 4.6 mm, packed with examination containing about 25 mg of Dextrose to a 250-ml
octadecylsilane chemically bonded to porous silica or conical flask with a ground-glass neck and add 25.0 ml of
ceramic microparticles 3 to 10 µm, cupri-citric solution. Add a few grains of pumice, fit a reflux
– mobile phase: a filtered and degassed solution in water condenser, heat so that boiling occurs within 2 minutes and
containing about 1 ml of formic acid and 1 ml of boil for exactly 10 minutes. Cool and add 3 g of potassium
dicyclohexylamine per litre, iodide dissolved in 3 ml of water. Carefully add, in small
– flow rate. 1 ml per minute, amounts, 25 ml of a 25 per cent w/w solution of sulphuric acid.
– spectrophotometer set at 210 nm, Titrate with 0.1 M sodium thiosulphate using starch solution
– a 20 µl loop injector. as indicator. Carry out a blank titration using 25 ml of water.
Calculate the content of anhydrous dextrose, C6H12O6, from
Inject separately reference solutions (a) and (b), and record
the following Table.
the chromatograms. The test is not valid unless the resolution
between the peaks due to acetate and lactate is not less than Volume of Anhydrous dextrose
2.0, the tailing factor for the analyte peak is not more than 2.0 0.1 M sodium thiosulphate
and the relative standard deviation for replicate injections is consumed (ml) (mg)
not more than 2.0 per cent. 8 19.8
Inject separately the test solution and reference solution (a), 9 22.4
and record the chromatograms. Measure the responses for 10 25.0
the major peak and calculate the content of lactate in the 11 27.6
preparation under examination. 12 30.3
For sodium bicarbonate — Titrate with 0.1 M hydrochloric 13 33.0
acid a volume of the preparation under examination containing 14 35.7
about 0.1 g of sodium bicarbonate, determining the end-point 15 38.5
potentiometrically (2.4.25). 16 41.3

920
IP 2007 PETHIDINE HYDROCHLORIDE

Storage. Peritoneal Dialysis Solutions are supplied in rigid or B. To 5 ml of a 1 per cent w/v solution add a few drops of
semi-rigid plastic containers, in flexible plastic containers fitted potassium mercuri-iodide solution; a cream-coloured
with a special connecting device (these are generally filled to precipitate is produced.
a volume below their nominal capacity and presented in closed
C. Dissolve 5 mg in 0.5 ml of water and add 0.1 ml of
protective envelopes) or in glass containers. Store at a
formaldehyde solution and 2 ml of sulphuric acid; an orange-
temperature not exceeding 30º.
red colour is produced.
CAUTION — Exposure to temperatures below 4º may cause
D. Gives reaction A of chlorides (2.3.1).
crystallisation and separation of solid particles rendering
the preparation unsuitable for use.
Tests
Labelling. The label states (1) the formula of the solution for
peritoneal dialysis, expressed in grams per litre and in millimoles Appearance of solution. A 2.0 per cent w/v solution in carbon
per litre; (2) the total osmolar concentration in mOsmol per dioxide-free water is clear (2.4.1), and colourless (2.4.1).
litre; (3) the nominal volume of the solution in the container; Acidity or alkalinity. To 10 ml of a 2.0 per cent w/v solution in
(4) that the solution is free from bacterial endotoxins, or where carbon dioxide-free water add 0.2 ml of methyl red solution
applicable, that it is apyrogenic; (5) that the solution is not to and 0.2 ml of 0.01 M sodium hydroxide; the solution is yellow.
be used for intravenous infusion; (6) that any unused portion Add 0.3 ml of 0.01 M hydrochloric acid; the solution is red.
of the solution is to be discarded; (7) that the solution
containing visible particles should not be used; (8) the storage Related substances. Determine by thin-layer chromatography
conditions. (2.4.17), coating the plate with kieselguhr G.
Mobile phase. The upper layer obtained by shaking together
100 volumes of light petroleum (50º to 70º), 8 volumes of
2-phenoxyethanol and 1 volume of diethylamine.
Pethidine Hydrochloride
Test solution. Dissolve 0.1 g in 5 ml of water, add 0.5 ml of
Meperidine Hydrochloride 10 M sodium hydroxide and 2 ml of ether and shake; allow the
layers to separate and use the upper layer.
CH3 Reference solution. Dilute 0.5 ml of the test solution to 50 ml
N with ether.
Impregnate the dry plate by placing it in a closed tank
O CH3 , HCl containing a mixture of 90 volumes of acetone and 10 volumes
of 2-phenoxyethanol so that the plate dips about 5 mm beneath
O
the surface of the liquid, allowing the impregnating solvent to
ascend at least 15 cm, removing the plate from the tank and
drying in a current of air. Use immediately, with the flow of the
C15H21NO2,HCl Mol. Wt. 283.8 mobile phase in the same direction as the impregnation.
Pethidine Hydrochloride is ethyl 1-methyl-4- Apply to the plate 5 µl of each solution. Allow the mobile
phenylpiperidine-4-carboxylate hydrochloride. phase to rise 12 cm. Dry the plate in air for 10 minutes, return
the plate to the tank and repeat the development. Remove the
Pethidine Hydrochloride contains not less than 99.0 per cent
plate, allow it to dry in air for 10 minutes and spray with a
and not more than 101.0 per cent of C15H21NO2,HCl, calculated
0.2 per cent w/v solution of 2,7-dichlorofluorescein in
on the dried basis.
methanol. Allow to stand for 5 minutes and spray with water
Description. Colourless crystals or a white, crystalline powder. until the background is white to pale yellow. Examine in
daylight. The chromatograms show red or orange spots. Any
Identification secondary spot in the chromatogram obtained with the test
solution is not more intense than the spot in the chromatogram
Test A may be omitted if tests B, C and D are carried out. Tests
obtained with the reference solution. Examine without delay
B and C may be omitted if tests A and D are carried out.
in ultraviolet light at 365 nm. The chromatograms show spots
A. Determine by infrared absorption spectrophotometry (2.4.6). with intense yellow fluorescence. Any secondary spot in the
Compare the spectrum with that obtained with pethidine chromatogram obtained with the test solution is not more
hydrochloride RS or with the reference spectrum of pethidine intense than the spot in the chromatogram obtained with the
hydrochloride. reference solution.

921
PETHIDINE INJECTIONS IP 2007

Sulphated ash (2.3.18). Not more than 0.1 per cent. necessary to 5 ml with water, with 0.5 ml of 5 M sodium
Loss on drying (2.4.19). Not more than 0.5 per cent, determined hydroxide and 2 ml of ether.
on 1.0 g by drying in an oven at 105º. Reference solution. Dilute 0.5 ml of the test solution to 50 ml
with ether.
Assay. Weigh accurately about 0.5 g, dissolve in 30 ml of
anhydrous glacial acetic acid, add 12 ml of mercuric acetate Impregnate the dry plate by placing it in a closed tank
solution. Titrate with 0.1 M perchloric acid, using crystal containing a mixture of 90 volumes of acetone and 10 volumes
violet solution as indicator. Carry out a blank titration. of 2-phenoxyethanol so that the plate dips about 5 mm beneath
the surface of the liquid, allowing the impregnating solvent to
1 ml of 0.1 M perchloric acid is equivalent to 0.02838 g of
ascend at least 15 cm, removing the plate from the tank and
C15H21NO2,HCl.
drying in a current of air. Use immediately, with the flow of the
Storage. Store protected from light and moisture. mobile phase in the same direction as the impregnation.
Apply to the plate 5 µl of each solution. Allow the mobile
phase to rise 12 cm. Dry the plate in air for 10 minutes, return
Pethidine Injection the plate to the tank and repeat the development. Remove the
plate, allow it to dry in air for 10 minutes and spray with a
Pethidine Hydrochloride Injection; Meperidine 0.2 per cent w/v solution of 2,7-dichlorofluorescein in
Hydrochloride Injection methanol. Allow to stand for 5 minutes and spray with water
Pethidine Injection is a sterile solution of Pethidine until the background is white to pale yellow. Examine in
Hydrochloride in Water for Injections. daylight. The chromatograms show red or orange spots. Any
secondary spot in the chromatogram obtained with the test
Pethidine Injection contains not less than 95.0 per cent and solution is not more intense than the spot in the chromatogram
not more than 105.0 per cent of pethidine hydrochloride, obtained with the reference solution. Examine without delay
C15H21NO2,HCl. in ultraviolet light at 365 nm. The chromatograms show spots
with intense yellow fluorescence. Any secondary spot in the
Identification
chromatogram obtained with the test solution is not more
A. To a volume containing 50 mg of Pethidine Hydrochloride intense than the spot in the chromatogram obtained with the
add sufficient 1 M sodium hydroxide to make strongly alkaline reference solution.
to litmus paper and extract with two quantities, each of 10 ml, Other tests. Complies with the tests stated under Parenteral
of chloroform. Wash the combined extracts with 5 ml of water, Preparations (Injections).
dry over anhydrous sodium sulphate, filter and evaporate the
Assay. Dilute an accurately measured volume containing about
filtrate to dryness. Remove the last traces of chloroform by
0.1 g of Pethidine Hydrochloride with 40 ml of water, add 1 ml
drying the residual oil at 60º at a pressure not exceeding
of 5 M sodium hydroxide and extract immediately with
0.7 kPa.
successive quantities of 25, 10 and 10 ml of chloroform. Wash
On the oily residue, determine by infrared absorption each extract with the same 15 ml of water, filter into a dry flask
spectrophotometry (2.4.6). Compare the spectrum with that and combine the extracts (which should be clear and free from
obtained with pethidine hydrochloride RS or with the droplets of water). Titrate with 0.02 M perchloric acid, using
reference spectrum of pethidine. 0.15 ml of 1-naphtholbenzein solution as indicator. Carry out
B. To 0.5 ml add 0.1 ml of formaldehyde solution and 2 ml of a blank titration.
sulphuric acid; an orange-red colour is produced. 1 ml of 0.02 M perchloric acid is equivalent to 0.005676 g of
C. Gives the reactions of chlorides (2.3.1). C15H21NO2,HCl.
Storage. Store protected from light.
Tests
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with kieselguhr G. Pethidine Tablets
Mobile phase. The upper layer obtained by shaking together Pethidine Hydrochloride Tablets; Meperidine
100 volumes of light petroleum (50º to 70º), 8 volumes of Hydrochloride Tablets
2-phenoxyethanol and 1 volume of diethylamine. Pethidine Tablets contain not less than 92.5 per cent and not
Test solution. The upper layer obtained by shaking a volume more than 107.5 per cent of the stated amount of pethidine
containing 0.1 g of Pethidine Hydrochloride, diluted if hydrochloride, C15H21NO2,HCl.

922
IP 2007 PHENINDAMINE TARTRATE

Identification intense than the spot in the chromatogram obtained with the
reference solution.
A. Shake a quantity of the powdered tablets containing 50 mg
of Pethidine Hydrochloride with 20 ml of chloroform, filter, Other tests. Comply with the tests stated under Tablets.
evaporate the filtrate to dryness and dry the residue at a Assay. Weigh and powder 20 tablets. Weigh accurately a
pressure of 2 kPa. quantity of the powder containing about 0.3 g of Pethidine
On the residue, determine by infrared absorption Hydrochloride in 40 ml of water, add 2 ml of 5 M sodium
spectrophotometry (2.4.6). Compare the spectrum with that hydroxide and extract immediately with successive quantities
obtained with pethidine hydrochloride RS or with the of 25, 10 and 10 ml of chloroform. Wash each extract with the
reference spectrum of pethidine hydrochloride. same 15 ml of water and filter into a dry flask. Combine the
extracts (which should be clear and free from droplets of water).
B. Shake a quantity of the powdered tablets containing 0.2 g Titrate with 0.05 M perchloric acid, using 0.15 ml of
of Pethidine Hydrochloride with 20 ml of water and filter. To 1-naphtholbenzein solution as indicator. Carry out a blank
5 ml of the filtrate add 10 ml of picric acid solution. The crystals titration.
so obtained, after washing with water and drying, melt at
about 190º (2.4.21). 1 ml of 0.05 M perchloric acid is equivalent to 0.01419 g of
C15H21NO2,HCl.
Tests Storage. Store protected from light and moisture.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with kieselguhr G.
Mobile phase. The upper layer obtained by shaking together Phenindamine Tartrate
100 volumes of light petroleum (50º to 70º), 8 volumes of
2-phenoxyethanol and 1 volume of diethylamine.
Test solution. The upper layer obtained by shaking a quantity
of the powdered tablets containing 0.1 g of Pethidine
Hydrochloride with 5 ml of water, filtering, shaking the filtrate H OH
with 0.5 ml of 5 M sodium hydroxide and 2 ml of ether and , COOH
allowing the layers to separate. N CH3 HOOC
H OH
Reference solution. Dilute 0.5 ml of the test solution to 50 ml
with ether.
C19H19N,C4H6O6 Mol. Wt. 411.5
Impregnate the dry plate by placing it in a closed tank
Phenindamine Tartrate is (RS)-2,3,4,9-tetrahydro-2-methyl-9-
containing a mixture of 90 volumes of acetone and 10 volumes
phenyl-1H-indeno[2,1-c]pyridine (2R,3R)-tartrate.
of 2-phenoxyethanol so that the plate dips about 5 mm beneath
the surface of the liquid, allowing the impregnating solvent to Phenindamine Tartrate contains not less than 98.5 per cent
ascend at least 15 cm, removing the plate from the tank and and not more than 101.0 per cent of C19H19N, C4H6O6, calculated
drying in a current of air. Use immediately, with the flow of the on the dried basis.
mobile phase in the same direction as the impregnation. Description. A white or almost white voluminous powder;
Apply to the plate 5 µl of each solution. Allow the mobile odourless or almost odourless.
phase to rise 12 cm. Dry the plate in air for 10 minutes, return
the plate to the tank and repeat the development. Remove the Identification
plate, allow it to dry in air for 10 minutes and spray with a A. When examined in the range 230 nm to 360 nm (2.4.7), a
0.2 per cent w/v solution of 2,7-dichlorofluorescein in 0.004 per cent w/v solution shows an absorption maximum
methanol. Allow to stand for 5 minutes and spray with water only at about 259 nm; absorbance at about 259 nm, about 0.88.
until the background is white to pale yellow. Examine in
daylight. The chromatograms show red or orange spots. Any B. Dissolve 25 mg in 5 ml of sulphuric acid; an orange-brown
secondary spot in the chromatogram obtained with the test colour is produced which is discharged when the solution is
solution is not more intense than the spot in the chromatogram carefully diluted with 20 ml of water.
obtained with the reference solution. Examine without delay C. Dissolve 0.5 g in 15 ml of hot water, add a slight excess of
in ultraviolet light at 365 nm. The chromatograms show spots 5 M sodium hydroxide, filter and neutralise the filtrate to litmus
with intense yellow fluorescence. Any secondary spot in the paper with 2 M hydrochloric acid. The solution gives reaction
chromatogram obtained with the test solution is not more B of tartrates (2.3.1).

923
PHENINDAMINE TABLETS IP 2007

D. Melting range (2.4.21). 160º to 162º, when heated to 163º it accurately measured quantity of the filtrate containing about
re-solidifies and melts again at about 168º, with decomposition. 0.2 g of Phenindamine Tartrate add 10 ml of 5 M sodium
hydroxide, and extract with 25 ml and then with three quantities,
Tests each of 10 ml, of chloroform, washing each extract with the
same 15 ml of water and filtering in a dry flask. Combine the
pH (2.4.24). 3.4 to 3.9, determined in a 1.0 per cent w/v solution.
extracts (which should be clear and free from droplets of
Sulphated ash (2.3.18). Not more than 0.1 per cent. water).Titrate with 0.02 M perchloric acid, using oracet blue
Loss on drying (2.4.19). Not more than 1.0 per cent, determined B solution as indicator. Carry out a blank titration.
on 1.0 g by drying in an oven at 105º. 1 ml of 0.02 M perchloric acid is equivalent to 0.008229 g of
Assay. Weigh accurately about 0.4 g, dissolve in 40 ml of C19H19N, C4H6O6.
warm water, cool, add 10 ml of dilute sodium carbonate Storage. Store protected from light and moisture.
solution and extract with successive quantities of 25, 10 and
10 ml of chloroform, washing each extract with the same 15 ml
of water and filtering into a dry flask. Combine the extracts
(which should be clear and free from droplets of water). Titrate
Phenindione
with 0.05 M perchloric acid, using oracet blue B solution as O
indicator. Carry out a blank titration.
1 ml of 0.05 M perchloric acid is equivalent to 0.02057 g of
C19H19N, C4H6O6.
Storage. Store protected from light and moisture. O
C15H10O2 Mol. Wt. 222.2
Phenindione is 2-phenylindane-1,3-dione.
Phenindamine Tablets Phenindione contains not less than 98.0 per cent and not
Phenindamine Tartrate Tablets more than 100.5 per cent of C15H10O2, calculated on the dried
basis.
Phenindamine Tablets contain not less than 92.5 per cent and
Description. Soft, white or creamy-white crystals; almost
not more than 107.5 per cent of the stated amount of
odourless.
phenindamine tartrate, C19H19N, C4H6O6. The tablets are coated.
Identification
Identification
A. Determine by infrared absorption spectrophotometry (2.4.6).
A. Shake a quantity of the powdered tablets containing 20 mg Compare the spectrum with that obtained with phenindione
of Phenindamine Tartrate with 100 ml of water, dilute 10 ml to RS.
100 ml with water and filter; absorbance of the filtrate at the
maximum at about 259 nm, about 0.44 (2.4.7). B. Dissolve 0.1 g in 30 ml of ethanol (95 per cent) with the aid
of heat, cool and add sufficient ethanol (95 per cent) to
B. To a portion of the finely powdered tablets containing about produce 50 ml. Dilute 10 ml of this solution to 250 ml with
50 mg of Phenindamine Tartrate, add 5 ml of water and 3 ml of 0.1 M sodium hydroxide and further dilute 5 ml to 100 ml with
dilute ammonia solution and extract the liberated base with 0.1 M sodium hydroxide. When examined in the range 230 nm
two successive quantities, each of 10 ml, of ether. Wash the and 360 nm (2.4.7), the solution shows absorption maxima at
combined ether extracts with 2 ml of water and evaporate to about 278 nm and at about 330 nm; absorbance at about
dryness. Dissolve 25 mg of the residue in 5 ml of sulphuric 278 nm, about 0.55 and at about 330 nm, about 0.16.
acid; an orange-brown colour is produced which is discharged
when the solution is carefully diluted with 20 ml of water. C. To 1 g add 50 ml of ethanol (95 per cent) and 0.5 ml of
aniline, heat gently under a reflux condenser for 3 hours, cool
Tests in ice and filter. The residue, after washing with 2 ml of ethanol
(95 per cent) and recrystallisation from chloroform, melts at
Other tests. Complies with the tests stated under Tablets. about 225º (2.4.21).
Assay. Weigh and digest 20 tablets with 70 ml of water and
Tests
5 ml of dilute hydrochloric acid until completely disintegrated,
filter and wash the residue with 20 ml of water. Dilute the Related substances. Determine by thin-layer chromatography
combined filtrate and washings to 100.0 ml with water. To an (2.4.17), coating the plate with silica gel GF254.

924
IP 2007 PHENINDIONE TABLETS

Mobile phase. A 0.02 per cent w/v solution of butylated A. Determine by infrared absorption spectrophotometry (2.4.6).
hydroxytoluene in a mixture of 80 volumes of toluene, Compare the spectrum with that obtained with phenindione
20 volumes of ethyl acetate and 4 volumes of glacial acetic RS.
acid. B. Dissolve 0.1 g in 30 ml of ethanol (95 per cent) with the aid
Test solution. Dissolve 0.1 g of the substance under of heat, cool and add sufficient ethanol (95 per cent) to
examination in 10 ml of dichloromethane. produce 50 ml. Dilute 10 ml of this solution to 250 ml with
0.1 M sodium hydroxide and further dilute 5 ml to 100 ml with
Reference solution (a). A 0.02 per cent w/v solution of the 0.1 M sodium hydroxide. When examined in the range 230 nm
substance under examination in dichloromethane. to 360 nm (2.4.7), the solution shows absorption maxima at
Reference solution (b). A 0.005 per cent w/v solution of the about 278 nm and 330 nm; absorbance at about 278. about
substance under examination in dichloromethane. 0.55 and at about 330 nm, about 0.16.

Apply to the plate 10 µl of each solution. Allow the mobile C. To 50 mg add 1 ml of sulphuric acid; a deep blue to violet
phase to rise 4 cm. Dry the plate in warm air and examine in solution is produced. On dilution with water the solution
ultraviolet light at 254 nm. Any secondary spot in the becomes colourless and a white precipitate is produced.
chromatogram obtained with the test solution is not more
Tests
intense than the spot in the chromatogram obtained with
reference solution (a) and not more than one such spot is Related substances. Determine by thin-layer chromatography
more intense than the spot in the chromatogram obtained (2.4.17), coating the plate with silica gel GF254.
with reference solution (b).
Mobile phase. A 0.02 per cent w/v solution of butylated
Sulphated ash (2.3.18). Not more than 0.1 per cent. hydroxytoluene in a mixture of 80 volumes of toluene,
20 volumes of ethyl acetate and 4 volumes of glacial acetic
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
acid.
on 1.0 g by drying in oven at 105º for 2 hours.
Test solution. Shake a quantity of the powdered tablets
Assay. Weigh accurately about 0.3 g, add 50 ml of ethanol
containing 50 mg of Phenindione with 15 ml of
(95 per cent) and warm until solution is effected. Cool to
dichloromethane, filter, evaporate the filtrate to dryness and
room temperature, add 10 ml of a 10 per cent v/v solution of
dissolve the residue in 5 ml of dichloromethane.
bromine in ethanol (95 per cent) and allow to stand for
10 minutes, shaking occasionally. Add 1 g of 2-naphthol and Reference solution (a). Dilute 1 volume of the test solution to
shake until the colour of the bromine is discharged. Remove 50 volumes with dichloromethane.
any vapour of bromine in the flask with a current of air, add Reference solution (b). Dilute 1 volume of the test solution to
50 ml of water and 10 ml of dilute potassium iodide solution 200 volumes with dichloromethane.
and titrate the liberated iodine with 0.1 M sodium thiosulphate
using starch solution as indicator. Apply to the plate 10 µl of each solution. Allow the mobile
phase to rise 4 cm. Dry the plate in warm air and examine in
1 ml of 0.1 M sodium thiosulphate is equivalent to 0.01111 g ultraviolet light at 254 nm. Any secondary spot in the
of C15H10O2. chromatogram obtained with the test solution is not more
Storage. Store protected from moisture. intense than the spot in the chromatogram obtained with
reference solution (a) and not more than one such spot is
more intense than the spot in the chromatogram obtained
with reference solution (b).
Phenindione Tablets Uniformity of content. (For tablets containing 50 mg or less)
— Comply with the test stated under Tablets.
Phenindione Tablets contain not less than 92.5 per cent and
not more than 107.5 per cent of the stated amount of Place one tablet in 50 ml of 0.1 M sodium hydroxide, dissolve
phenindione, C15H10O2. completely by shaking gently, add a further 100 ml of 0.1 M
sodium hydroxide and shake for 1 hour. Dilute to 250.0 ml with
Identification 0.1 M sodium hydroxide, filter and dilute a portion of the
filtrate with sufficient 0.1 M sodium hydroxide to produce a
Shake a quantity of the powdered tablets containing 0.2 g of solution containing 4 µg of Phenindione per ml. Measure the
Phenindione with 50 ml of chloroform, filter and evaporate the absorbance of the resulting solution at the maximum at about
filtrate to dryness. Recrystallise the residue from ethanol 278 nm (2.4.7). Calculate the content of C15H10O2 taking 1310
(95 per cent). The crystals complies with the following tests. as the specific absorbance at 278 nm.

925
PHENIRAMINE MALEATE IP 2007

Other tests. Complies with the tests stated under Tablets. add 5 ml of water; a yellow colour is produced. To 2 ml of the
Assay. Weigh and powder 20 Tablets. Weigh accurately a solution add 3 ml of a 50 per cent w/v solution of ammonium
quantity of the powder containing about 50 mg of Phenindione acetate, previously cooled in ice; a pink colour is produced
and shake with 150 ml of 0.1 M sodium hydroxide for 1 hour, which persists for at least 10 minutes in the cooled solution.
add sufficient 0.1 M sodium hydroxide to produce 250.0 ml, Tests
filter and dilute 5.0 ml of the filtrate to 250.0 ml with 0.1 M
sodium hydroxide. Measure the absorbance of the resulting pH (2.4.24). 4.5 to 5.5, determined in a 1.0 per cent w/v solution.
solution at the maximum at about 278 nm (2.4.7). Calculate the
Related substances. Determine by thin-layer chromatography
content of C15H10O2 taking 1310 as the specific absorbance at
(2.4.17), coating the plate with silica gel G.
278 nm.
Mobile phase. A mixture of 50 volumes of cyclohexane,
Storage. Store protected from moisture.
40 volumes of chloroform and 10 volumes of diethylamine.
Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml of methanol.
Pheniramine Maleate
Reference solution (a). A 0.02 per cent w/v solution of the
substance under examination in methanol.
Reference solution (b). A 0.004 per cent w/v solution of the
substance under examination in methanol.
COOH
N CH3 , Apply to the plate 10 µl of each solution. After development,
N
dry the plate in air, spray with dilute potassium
CH3 COOH
iodobismuthate solution. Any secondary spot in the
chromatogram obtained with the test solution is not more
C16H20N2,C4H4O4 Mol. Wt. 356.4 intense than the spot in the chromatogram obtained with
Pheniramine Maleate is (3RS)-N,N-dimethyl-3-phenyl-3- reference solution (a) and not more than one such spot is
(pyridin-2-yl)propan-1-amine hydrogen maleate. more intense than the spot in the chromatogram obtained
with reference solution (b).
Pheniramine Maleate contains not less than 99.0 per cent and
not more than 101.0 per cent of C16H20N2,C4H4O4, calculated Heavy metals (2.3.13). Dissolve 1.0 g in 10 ml of water and add
on the dried basis. 2 ml of acetic acid and sufficient water to produce 25 ml. The
resulting solution complies with the limit test for heavy metals,
Description. A white or almost white, crystalline powder;
Method A (20 ppm).
odourless or almost odourless.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Identification
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
A. Determine by infrared absorption spectrophotometry (2.4.6). on 1.0 g by drying in an oven at 60º at a pressure not exceeding
Compare the spectrum with that obtained with pheniramine 0.7 kPa.
maleate RS or with the reference spectrum of pheniramine Assay. Weigh accurately about 0.4 g, dissolve in 20 ml of
maleate. anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
B. When examined in the range 230 nm to 360 nm (2.4.7), a acid, using 1- naphtholbenzein solution as indicator. Carry
0.002 per cent w/v solution in 0.1 M hydrochloric acid shows out a blank titration.
an inflection at about 262 nm; absorbance at about 265 nm, 1 ml of 0.1 M perchloric acid is equivalent to 0.01782 g of
about 0.42. C16H20N2,C4H4O4.
C. Dissolve 0.25 g in 5 ml of water, add 2 ml of strong ammonia Storage. Store protected from light and moisture.
solution and extract with three quantities, each of 5 ml, of
chloroform. Evaporate the aqueous extract to dryness, add
0.2 ml of 1 M sulphuric acid and 5 ml of water, extract with
four quantities, each of 25 ml, of ether and evaporate the Pheniramine Injection
combined ether extracts to dryness in a current of warm air. To
Pheniramine Maleate Injection
the residue add 50 mg of resorcinol and 1 ml of sulphuric
acid, heat in a water-bath for 2 minutes, shake well, heat in a Pheniramine Injection is a sterile solution of Pheniramine
water-bath for a further 30 minutes and cool in ice. Carefully Maleate in Water for Injections.

926
IP 2007 PHENIRAMINE TABLETS

Pheniramine Injection contains not less than 90.0 per cent and Pheniramine Tablets
not more than 110.0 per cent of the stated amount of
pheniramine maleate, C16H20N2, C4H4O4. Pheniramine Maleate Tablets
Identification Pheniramine Tablets contain not less than 92.5 per cent and
not more than 107.5 per cent of the stated amount of
Determine by thin-layer chromatography (2.4.17), coating the pheniramine maleate, C16H20N2, C4H4O4.
plate with silica gel GF254.
Mobile phase. A mixture of 50 volumes of cyclohexane,
Identification
40 volumes of chloroform and 10 volumes of diethylamine. Boil a quantity of the powdered tablets containing about 0.5 g
Test solution. Evaporate an appropriate volume of the injection of Pheniramine Maleate with 150 ml of acetone under a reflux
to dryness in a current of nitrogen using the minimum amount condenser for about 45 minutes. Filter and evaporate the filtrate
of heat, dissolve the residue in sufficient chloroform to to dryness on a water-bath. The residue complies with the
produce a solution containing 2.0 per cent w/v solution of following tests.
Pheniramine Maleate and centrifuge. A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution. A 2.0 per cent w/v solution of pheniramine Compare the spectrum with that obtained with pheniramine
maleate RS in chloroform. maleate RS or with the reference spectrum of pheniramine
Apply to the plate 10 µl of each solution. After development, maleate.
dry the plate in air and examine in ultraviolet light at 254 nm. B. When examined in the range 230 nm to 360 nm (2.4.7), a
The two principal spots in the chromatogram obtained with 0.002 per cent w/v solution in 0.1 M hydrochloric acid shows
the test solution correspond to those in the chromatogram an inflection at about 262 nm; absorbance at about 265 nm,
obtained with the reference solution. Spray the plate with about 0.42.
dilute potassium iodobismuthate solution. The principal spot C. Dissolve 0.25 g in 5 ml of water, add 2 ml of strong ammonia
in the chromatogram obtained with the test solution solution and extract with three quantities, each of 5 ml, of
corresponds to that in the chromatogram obtained with the chloroform. Evaporate the aqueous extract to dryness, add
reference solution. 0.2 ml of 1 M sulphuric acid and 5 ml of water, extract with
Tests four quantities, each of 25 ml, of ether and evaporate the
combined ether extracts to dryness in a current of warm air. To
pH (2.4.24). 4.5 to 5.5. the residue add 50 mg of resorcinol and 1 ml of sulphuric
Related substances. Determine by the method described under acid, heat in a water-bath for 2 minutes, shake well, heat in a
the Identification test using as the reference solution a water-bath for a further 30 minutes and cool in ice. Carefully
solution prepared by diluting 1 volume of the test solution to add 5 ml of water; a yellow colour is produced. To 2 ml of the
500 volumes with chloroform. Any secondary spot in the solution add 3 ml of a 50 per cent w/v solution of ammonium
chromatogram obtained with the test solution is not more acetate, previously cooled in ice; a pink colour is produced
intense than the spot in the chromatogram obtained with the which persists for at least 10 minutes in the cooled solution.
reference solution.
Tests
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections). Related substances. Determine by thin-layer chromatography
Assay. To an accurately measured volume containing about (2.4.17), coating the plate with silica gel G.
0.11 g of Pheniramine Maleate add sufficient water to produce Mobile phase. A mixture of 50 volumes of cyclohexane,
50.0 ml and mix well. To 20.0 ml add sufficient 1 M sodium 40 volumes of chloroform and 10 volumes of diethylamine.
hydroxide to make the solution just alkaline to litmus paper, Test solution. Shake a quantity of the powdered tablets
add 2 ml in excess and extract with two quantities, each of containing 20 mg of Pheniramine Maleate with 10 ml of
50 ml, of ether. Wash each ether extract in succession with 20, methanol, centrifuge and use the supernatant liquid.
20 and 5 ml of 0.1 M hydrochloric acid, dilute the combined
extracts to 100.0 ml with 0.1 M hydrochloric acid and mix. Reference solution (a). Dilute 1 volume of the test solution to
Dilute 5.0 ml to 100.0 ml with 0.1 M hydrochloric acid and 100 volumes with methanol.
measure the absorbance of the resulting solution at the Reference solution (b). Dilute 1 volume of reference solution
maximum at about 265 nm (2.4.7), using 0.1 M hydrochloric (a) to 20 volumes with methanol.
acid as the blank. Calculate the content of C16H20N2, C4H4O4 Apply to the plate 10 µl of each solution. After development,
taking 210 as the specific absorbance at 265 nm. dry the plate in air, spray with dilute potassium
Storage. Store protected from light. iodobismuthate solution. Any secondary spot in the

927
PHENOBARBITONE IP 2007

chromatogram obtained with the test solution is not more difference between the melting points, which are about 175º,
intense than the spot in the chromatogram obtained with is not greater than 2º.
reference solution (a) and not more than one such spot is C. Complies with the test for identification of barbiturates
more intense than the spot in the chromatogram obtained (2.3.2).
with reference solution (b).
D. Dissolve about 20 mg in 5 ml of ethanol, add a drop of
Other tests. Complies with the tests stated under Tablets. cobalt chloride solution and a drop of dilute ammonia
Assay. Weigh and powder 20 tablets. Weigh accurately a solution; a violet colour is produced.
quantity of the powder containing about 45 mg of Pheniramine E. Gives the reaction of non-nitrogen substituted barbiturates
Maleate, shake with 20 ml of 0.1 M hydrochloric acid, centrifuge (2.3.1).
and transfer the supernatant liquid to a 100-ml volumetric flask.
Repeat the extraction with three further quantities, each of 20 Tests
ml, of 0.1 M hydrochloric acid. Combine the extracts and add
Appearance of solution. A 10.0 per cent w/v solution in a
sufficient 0.1 M hydrochloric acid to produce 100.0 ml. Mix
mixture of 20 volumes of 2 M sodium hydroxide and 30 volumes
and dilute 5.0 ml to 100.0 ml with 0.1 M hydrochloric acid;
of water is clear (2.4.1), and not more intensely coloured than
measure the absorbance of the resulting solution at the
reference solution YS6 (2.4.1).
maximum at about 265 nm (2.4.7), using 0.1 M hydrochloric
acid as the blank. Calculate the content of C16H20N2,C4H4O4 Acidity. Mix 1.0 g with 50 ml of water, boil for 2 minutes, allow
taking 210 as the specific absorbance at 265 nm. to cool, filter and adjust the volume to 50 ml. To 10 ml of the
filtrate add 0.15 ml of methyl red solution; not more than 0.1 ml
Storage. Store protected from light and moisture.
of 0.1 M sodium hydroxide is required to change the colour of
the solution from orange-yellow to pure yellow.
Related substances. Complies with the test for related
Phenobarbitone substances in barbiturates (2.3.4).
Phenobarbital Sulphated ash (2.3.18). Not more than 0.1 per cent.

H Loss on drying (2.4.19). Not more than 1.0 per cent w/w,
O N O determined on 1.0 g by drying in an oven at 105º for 2 hours.
H3C Assay. Weigh accurately about 0.1 g, dissolve in 5 ml of
NH pyridine, add 0.25 ml of thymolphthalein solution and 10 ml
of silver nitrate-pyridine reagent and titrate with 0.1 M
O
ethanolic sodium hydroxide until a pure blue colour is
obtained. Repeat the operation without the substance under
C12H12N2O3 Mol. Wt. 232.2 examination. The difference between the titrations represents
Phenobarbitone is 5-ethyl-5-phenylbarbituric acid. the amount of sodium hydroxide required.
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
Phenobarbitone contains not less than 99.0 per cent and not
0.01161 g of C12H12N2O3.
more than 101.0 per cent of C12H12N2O3, calculated on the
dried basis. Storage. Store protected from moisture.
Description. Colourless crystals or a white, crystalline powder;
odourless.
Phenobarbitone Sodium
Identification Phenobarbital Sodium; Soluble Phenobarbitone; Soluble
Test A may be omitted if tests B, C, D and E are carried out. Phenobarbital
Tests C, D and E may be omitted if tests A and B are carried
out. H
O N ONa
A. Determine by infrared absorption spectrophotometry (2.4.6). H3C
Compare the spectrum with that obtained with phenobarbitone N
RS or with the reference spectrum of phenobarbitone.
O
B. Determine the melting point (2.4.21) of the substance under
examination and of a mixture of equal quantities of the
substance under examination and phenobarbitone RS. The C12H11N2NaO3 Mol. Wt. 254.2

928
IP 2007 PHENOBARBITONE INJECTION

Phenobarbital Sodium; Soluble Phenobarbitone; Soluble Test solution. A 1 per cent w/v solution of the substance
Phenobarbital. under examination in ethanol (50 per cent).
Phenobarbitone Sodium contains not less than 99.0 per cent Reference solution. A 0.005 per cent w/v solution of the
and not more than 101.0 per cent of C12H11N2NaO3, calculated substance under examination in ethanol (50 per cent).
on the dried basis.
Loss on drying (2.4.19). Not more than 7.0 per cent, determined
Description. A white powder or crystalline granules or flaky on 0.5 g by drying in an oven at 150º for 4 hours.
crystals; hygroscopic.
Assay. Weigh accurately about 0.15 g, dissolve in 2 ml of
Identification water and add 8 ml of 0.05 M sulphuric acid. Heat to boiling
and cool. Add 30 ml of methanol and shake until dissolution
Test A may be omitted if tests B, C, D, E and F are carried out. is complete. Titrate with 0.1 M sodium hydroxide, determining
Tests C, D and E may be omitted if tests A, B and F are carried the end-point potentiometrically (2.4.25). After the first
out. inflection, stop the addition of the sodium hydroxide, add
A. Dissolve 0.2 g in 20 ml of ethanol (50 per cent), acidify 10 ml of pyridine, mix and continue the titration until the
with dilute hydrochloric acid and extract with 50 ml of ether. second inflection is reached. The difference between the
Wash the ether layer with 10 ml of water, dry over anhydrous volumes represents the amount of sodium hydroxide
sodium sulphate and filter. Evaporate the filtrate to dryness required.
and dry the residue at 105º.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02542 g of
On the residue, determine by infrared absorption C12H11N2NaO3.
spectrophotometry (2.4.6). Compare the spectrum with that
Storage. Store protected from moisture.
obtained with phenobarbitone RS or with the reference
spectrum of phenobarbitone.
B. Determine the melting point (2.4.21), of the residue obtained
in test A and of a mixture of equal quantities of the residue and
phenobarbitone RS. The difference between the melting
Phenobarbitone Injection
points, which are about 175º, is not greater than 2º. Phenobarbital Sodium Injection; Phenobarbitone Sodium
C. Complies with the test for identification of barbiturates Injection; Soluble Phenobarbitone Injection
(2.3.2), but using the following solutions. Phenobarbitone Injection is a sterile solution of
Test solution. A 0.1 per cent w/v solution of the substance Phenobarbitone Sodium in a mixture of nine volumes of
under examination in ethanol (50 per cent). Propylene Glycol and one volume of Water for Injections.
Reference solution. A 0.09 per cent w/v solution of Phenobarbitone Injection contains not less than 95.0 per cent
phenobarbitone RS in ethanol (50 per cent). and not more than 105.0 per cent of the stated amount of
phenobarbitone sodium, C12H11N2NaO3.
D. 1 g dissolves completely in 20 ml of ethanol (90 per cent)
(distinction from barbitone sodium).
Identification
E. Gives the reaction of non-nitrogen substituted barbiturates
(2.3.1). To a volume containing 1 g of Phenobarbitone Sodium add
15 ml of water if necessary, make slightly acidic with 1 M
F. Ignite about 0.1 g; the residue gives the reactions of sodium sulphuric acid and filter. The residue, after washing with water
salts (2.3.1). and drying at 105º, complies with the following tests.
Tests A. Determine by infrared absorption spectrophotometry (2.4.6).
Appearance of solution. A 10.0 per cent w/v solution in ethanol Compare the spectrum with that obtained with phenobarbitone
(50 per cent) is clear (2.4.1), and not more intensely coloured RS or with the reference spectrum of phenobarbitone.
than reference solution YS7 (2.4.1). B. Dissolve 50 mg in 2 ml of a 0.2 per cent w/v solution of
pH (2.4.24). Not more than 10.2, determined in a 10.0 per cent cobalt acetate in methanol, warm, add 50 mg of powdered
w/v solution. borax and heat to boiling; a bluish violet colour is produced.

Related substances. Complies with the test for related Tests


substances in barbiturates (2.3.4), but using the following
solutions. pH (2.4.24).10.0 to 11.0.

929
PHENOBARBITONE SODIUM TABLETSS IP 2007

Other tests. Complies with the tests stated under Parenteral Tests
Preparations (Injections).
Other tests. Complies with the tests stated under Tablets.
Assay. Weigh accurately about 2.0 g, add 30 ml of water and
3 g of sodium carbonate, stir to dissolve and titrate with Assay. Weigh and powder 20 tablets. Weigh accurately a
0.1 M silver nitrate until a distinct turbidity is observed when quantity of the powder containing about 0.3 g of
viewed against a black background, the solution being stirred Phenobarbitone Sodium, dissolve as completely as possible
vigorously throughout the titration. in 10 ml of a 2 per cent w/v solution of sodium hydroxide,
saturate with sodium chloride, acidify with hydrochloric acid
1 ml of 0.1 M silver nitrate is equivalent to 0.02542 g of and extract with successive quantities, each of 15 ml, of ether
C12H11N2NaO3. until complete extraction is effected. Wash the combined
Determine the weight per ml of the injection (2.4.29) and extracts with two quantities, each of 2 ml, of water and extract
calculate the percentage weight in volume of C12H11N2NaO3. the combined washings with 10 ml of ether. Add the ether to
the main ether layer and dry the residue to constant weight at
Storage. Store in single dose containers. 105º.
Labelling. The label states that the injection should not be 1 g of the residue is equivalent to 1.095 g of C12H11N2NaO3.
used if the solution is discoloured or if it contains a precipitate.
Storage. Store protected from moisture.

Phenobarbitone Sodium Tablets Phenobarbitone Tablets


Phenobarbital Sodium Tablets; Soluble Phenobarbitone Phenobarbital Tablets
Tablets; Soluble Phenobarbital Tablets Phenobarbitone Tablets contain not less than 92.5 per cent
and not more than 107.5 per cent of the stated amount of
Phenobarbitone Sodium Tablets contain not less than
phenobarbitone, C12H12N2O3.
92.5 per cent and not more than 107.5 per cent of the stated
amount of phenobarbitone sodium, C12H11N2NaO3.
Identification
Identification Extract a quantity of the powdered tablets containing about
0.5 g of Phenobarbitone with 50 ml of ether, filter through
A. Heat 0.1 g of the residue obtained in Assay on a water-bath
anhydrous sodium sulphate and evaporate the ether to
with 15 ml of ethanol (25 per cent) until dissolved, filter while
dryness on a water-bath. The residue complies with the
hot and allow to cool. Filter through a sintered-glass crucible,
following tests.
wash with a small quantity of ethanol (25 per cent) and dry at
105º. Heat in a sealed tube at 105º for 1 hour. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with phenobarbitone
On the residue, determine by infrared absorption
RS or with the reference spectrum of phenobarbitone.
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with phenobarbitone RS or with the reference B. Dissolve 50 mg in 2 ml of a 0.2 per cent w/v solution of
spectrum of phenobarbitone. cobaltous acetate in methanol, warm, add 50 mg of powdered
borax and heat to boiling; a bluish violet colour is produced.
B. The residue obtained in test A melts at about 175º (2.4.21).
C. Dissolve 50 mg of the residue obtained in Assay in 2 ml of Tests
a 0.2 per cent w/v solution of cobaltous acetate in methanol,
warm, add 50 mg of powdered borax and heat to boiling; a Disintegration (2.5.1). 30 minutes
bluish violet colour is produced. Other tests. Complies with the tests stated under Tablets.
D. Triturate a quantity of the powdered tablets containing Assay. Weigh and powder 20 tablets. Weigh accurately a
0.2 g of Phenobarbitone Sodium with 5 ml of water and filter; quantity of the powder containing about 0.3 g of
the filtrate is alkaline to litmus solution and yields a white Phenobarbitone and extract in a continuous extraction
precipitate on the addition of dilute hydrochloric acid. apparatus (2.1.8) with ether until complete extraction is
effected. Remove the ether and dry the residue, which is
E. The powdered tablets, when moistened with hydrochloric
C12H12N2O3, to constant weight at 105º.
acid and introduced on a platinum wire into a flame, impart a
yellow colour to the flame. Storage. Store protected from moisture.

930
IP 2007 PHENOLPHTHALEIN

Phenol under examination. The difference between the titrations


represents the amount of bromine required.
Carbolic acid
1 ml of 0.05 M bromine is equivalent to 0.001569 g of C6H6O.

OH Storage. Store protected from light and moisture.

Phenolphthalein
C6H6O Mol. Wt. 94.1
O
Phenol contains not less than 99.0 per cent and not more than
100.5 per cent of C6H6O. O
Description. Colourless or faintly pink or faintly yellowish
crystals or crystalline masses; odour, characteristic;
deliquescent. OH

Identification HO
A. 0.5 g dissolves completely in 2 ml of strong ammonia
solution. Dilute this solution to about 100 ml with water and C20H14O4 Mol. Wt. 318.3
to 2 ml of the resulting solution add 0.05 ml of sodium Phenolphthalein is 3,3-bis(4-hydroxyphenyl)phthalide.
hypochlorite solution (3 per cent Cl); a blue colour develops
Phenolphthalein contains not less than 98.0 per cent and not
which becomes progressively more intense.
more than 102.0 per cent of C20H14O4, calculated on the dried
B. Dissolve 1.0 g in sufficient water to produce 15 ml (solution basis.
A) and to 1 ml add 10 ml of water and 0.1 ml of ferric chloride
Description. A white or yellowish white, crystalline or
solution; a violet colour is produced which disappears on the
amorphous powder; odourless or almost odourless.
addition of 5 ml of 2-propanol.
C. To 1 ml of solution A add 10 ml of water and 1 ml of bromine Identification
solution; a pale yellow precipitate is produced.
A. Dissolves in dilute solutions of alkali hydroxides and in
Tests hot solutions of alkali carbonates forming a red solution which
is decolorised by dilute acids.
Appearance of solution. Solution A is clear (2.4.1), and not
B. Melting range (2.4.21). 258º to 263º.
more intensely coloured than reference solution BS6 (2.4.1).
Acidity. To 2 ml of solution A add 0.05 ml of methyl orange Tests
solution; the solution is yellow.
Heavy metals (2.3.13). Heat 5 g with 50 ml of dilute
Freezing point (2.4.11). Not less than 39.5º. hydrochloric acid on a water-bath for 5 min and filter.
Non-volatile matter. Not more than 0.05 per cent, when 5.0 g is Evaporate the filtrate almost to dryness and dissolve the
volatilised on a water-bath and dried to constant weight at residue in 50 ml of water. 12 ml of this solution complies with
105º. the limit test for heavy metals, Method D (20 ppm). Use 10 ml
of lead standard solution (2 ppm Pb) to prepare the standard.
Assay. Weigh accurately about 0.5 g and dissolve in sufficient
water to produce 250.0 ml. Transfer 25.0 ml to a ground-glass- Fluoran. 0.5 g dissolves completely in a mixture of 4 ml of 1 M
stoppered flask, add 50.0 ml of 0.05 M bromine and 5 ml of sodium hydroxide and 50 ml of water.
hydrochloric acid, stopper, allow to stand for 30 minutes, Sulphated ash (2.3.18). Not more than 0.1 per cent.
swirling occasionally, and allow to stand for a further
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
15 minutes. Add 5 ml of a 20 per cent w/v solution of potassium
on 1.0 g by drying in an oven at 105º.
iodide, shake and titrate with 0.1 M sodium thiosulphate until
a faint yellow colour remains. Add 0.5 ml of starch solution Assay. Weigh accurately about 0.1 g, dissolve in 100.0 ml of
and 10 ml of chloroform and continue the titration with ethanol (95 per cent), dilute 5.0 ml of this solution to 50.0 ml
vigorous shaking. Repeat the operation without the substance with ethanol (95 per cent) and evaporate 5.0 ml of the resulting

931
PHENOXYMETHYLPENICILLIN POTASSIUM IP 2007

solution to dryness on a water-bath. Dissolve the residue in Phenoxyacetic acid. (Not more than 0.5 per cent).
sufficient glycine buffer pH 11.3 to produce 100.0 ml, mix and Determine by liquid chromatography (2.4.14).
immediately measure the absorbance of the resulting solution
at the maximum at about 555 nm (2.4.7). Calculate the content Diluent. pH 6.6.phosphate buffer.
of C20H14O4 taking 1055 as the specific absorbance at 555 nm. Test solution. Weigh accurately a suitable quantity of the
Storage. Store protected from moisture. substance under examination, dissolve in the diluent and dilute
to obtain a solution having a known concentration of about
20.0 mg per ml. (Use this solution on the day prepared).
Reference solution. Weigh accurately a suitable quantity of
Phenoxymethylpenicillin Potassium phenoxyacetic acid, dissolve in the diluent and dilute to obtain
Penicillin V Potassium a solution having a known concentration of about 0.1 mg per
ml.
H COOK Chromatographic system
O
O N CH3 – a stainless steel column 25 cm x 4.6 mm, packed with
O octadecylsilane chemically bonded to porous silica
N S CH3 (5 µm),
H H H – mobile phase: a mixture of 65 volumes of water,
35 volumes of acetonitrile and 1 volume of glacial acetic
acid,
C16H17KN2O5S Mol. Wt. 388.5
– flow rate. 1 ml per minute,
Phenoxymethylpenicillin Potassium is potassium (6R)-6-(2- – spectrophotometer set at 254 nm,
phenoxyacetamido)penicillinate, produced by the growth of – a 20 µl loop injector.
certain strains of Penicillium notatum or related organisms
Inject the reference solution. The tailing factor is not more
on a culture medium containing an appropriate precursor, or
than 1.5 and the relative standard deviation for replicate
obtained by any other means.
injections is not more than 2.0 per cent.
Phenoxymethylpenicillin Potassium contains not less than
Inject alternately the test solution and the reference solution.
86.0 per cent of total penicillins C16H17N2O5S, calculated on
the anhydrous basis. Calculate the content of phenoxyacetic acid.
Description. A white, crystalline powder. Limit of p-hydroxyphenoxymethylpenicillin. Not more than
5.0 per cent.
Identification Using the chromatograms obtained with the test solution in
Test A may be omitted if tests B and C are carried out. Test B the Assay, calculate the content of p-hydroxyphenoxymethyl-
may be omitted if tests A and C are carried out. penicillin from the peak response of p-hydroxyphenoxy-
methylpenicillin and the sum of the peak responses of
A. Determine by infrared absorption spectrophotometry (2.4.6). p-hydroxyphenoxymethylpenicillin and phenoxymethyl-
Compare the spectrum with that obtained with penicillin.
phenoxymethylpenicillin potassium RS.
Water (2.3.43). Not more than 1.0 per cent, determined on
B. Gives reaction B of penicillins and cephalosporins (2.3.1). 1.0 g.
C. Gives reaction A of potassium salts (2.3.1). Assay. Determine by liquid chromatography (2.4.14).

Tests Test solution. Dissolve about 125 mg of the substance under


examination in the mobile phase and dilute to 50.0 ml with the
pH (2.4.24). 5.5 to 7.5, determined in a 0.5 per cent w/v solution. mobile phase.
Specific optical rotation (2.4.22). +215.0º to +230.0º, determined Reference solution (a). Dissolve an accurately weighed
in a 1.0 per cent w/v solution in carbon dioxide-free water. quantity of phenoxymethylpenicillin potassium RS in the
mobile phase and dilute to obtain a solution having a known
Light absorption (2.4.7). Absorbance of a 0.1 per cent w/v
concentration of about 2.5 mg per ml.
solution in 0.1 M sodium hydroxide at the maximum at about
306 nm, not more than 0.33. Absorbance of a 0.02 per cent Reference solution (b). A solution in the mobile phase
w/v solution in 0.1M sodium hydroxide at the maximum at containing 0.25 per cent w/v each of benzylpenicillin potassium
about 274 nm, not less than 0.50. and phenoxymethylpenicillin potassium.

932
IP 2007 PHENOXYMETHYLPENICILLIN POTASSIUM TABLETS

Chromatographic system add 1.0 ml of penicillinase solution; the solution changes


– a stainless steel column 30 cm x 4 mm, packed with rapidly to red.
octadecylsilane chemically bonded to porous silica C. Ignite 0.5 g of the powdered tablets, add 5 ml of 2 M
(3 to 10 µm), hydrochloric acid, boil, cool and filter. The filtrate gives
– mobile phase: a mixture of 650 volumes of water, reaction B of potassium salts (2.3.1).
350 volumes of acetonitrile and 5.75 volumes of glacial
acetic acid, Tests
– flow rate. I ml per minute,
Dissolution (2.5.2).
– spectrophotometer set at 254 nm,
– a 10 µl loop injector. Apparatus. No 1
Medium. 900 ml of water
Inject reference solution (b).The relative retention times are
Speed and time. 50 rpm and 45 minutes.
about 0.8 for benzylpenicillin and 1.0 for phenoxymethyl-
penicillin. The column efficiency determined from the Withdraw a suitable volume of the medium and filter. Measure
phenoxymethylpenicillin peak is not less than the absorbance of the filtrate, suitably diluted if necessary, at
1800 theoretical plates and the resolution between benzyl- the maximum at about 268 nm (2.4.7). At the same time measure
penicillin and phenoxymethylpenicillin is not less than 3.0. the absorbance of a solution of known concentration of
phenoxymethylpenicillin potassium RS at the maximum at
Inject reference solution (a). The relative standard deviation
about 268 nm. Calculate the content of C16H18N2O5S, in the
for replicate injections is not more than 1.0 per cent.
medium.
Inject the test solution and reference solution (a). Record the
D. Not less than 75 per cent of the stated amount of
chromatograms and measure the responses for the
C16H18N2O5S.
phenoxymethylpenicillin peak and any p-hydroxyphenoxy-
methylpenicillin peak with a retention time of about 0.4 relative Other tests. Complies with the tests stated under Tablets.
to that of the main phenoxymethylpenicillin peak. Assay. Determine by liquid chromatography (2.4.14).
Calculate the content of C16H17N2O5S, from the sum of the Test solution. Weigh and finely powder 20 tablets. Dissolve
peak responses of the p-hydroxyphenoxymethylpenicillin and an accurately weighed quantity of the powder containing about
phenoxymethylpenicillin peaks in the chromatograms obtained 0.25 g of phenoxymethylpenicillin in the mobile phase by
with the test solution and reference solution (a). shaking for 5 minutes and dilute to 100.0 ml with the mobile
Storage. Store protected from moisture. phase. Filter through a 0.5 µm or finer filter and use the filtrate.
Reference solution (a). Dissolve an accurately weighed
quantity of phenoxymethylpenicillin potassium RS in the
mobile phase and dilute to obtain a solution having a known
Phenoxymethylpenicillin Potassium concentration of about 2.5 mg per ml.
Tablets Reference solution (b). A solution in the mobile phase
Phenoxymethylpenicillin Tablets; Penicillin V Potassium containing 0.25 per cent w/v each of benzylpenicillin potassium
Tablets; Penicillin V Tablets and phenoxymethylpenicillin potassium.
Chromatographic system
Phenoxymethylpenicillin Potassium Tablets contain not less
– a stainless steel column 30 cm x 4 mm, packed with
than 92.5 per cent and not more than 107.5 per cent of the
octadecylsilane chemically bonded to porous silica,
stated amount of phenoxymethylpenicillin, C16H18N2O5S.
– mobile phase: a mixture of 650 volumes of water,
Identification 350 volumes of acetonitrile and 5.75 volumes of glacial
acetic acid,
A. Shake a quantity of the powdered tablets containing 80 mg – flow rate. 1 ml per minute,
of phenoxymethylpenicillin with water, dilute to 250 ml with – spectrophotometer set at 254 nm,
water and filter. When examined between 230 and 360 nm – a 10 µl loop injector.
(2.4.7), the filtrate shows absorption maxima at about 268 nm
Inject reference solution (b).The relative retention times are
and 274 nm and a minimum at about 272 nm.
about 0.8 for benzylpenicillin and 1.0 for phenoxymethyl-
B. Shake a quantity of the powdered tablets containing 10 mg penicillin. The column efficiency determined from the
of phenoxymethylpenicillin with 10 ml of water, filter and add phenoxymethylpenicillin peak is not less than 1800 theoretical
0.5 ml of neutral red solution. Add sufficient 0.01 M sodium plates and the resolution between the benzylpenicillin and
hydroxide to produce a permanent orange colour and then phenoxymethylpenicillin peaks is not less than 3.0.

933
PHENOTOLAMINE MESYLATE IP 2007

Inject reference solution (b). The relative standard deviation D. Mix 50 mg with 0.2 g of powdered sodium hydroxide, heat
for replicate injections is not more than 1.0 per cent. to fusion and continue the heating for a few seconds longer.
Inject the test solution and reference solution (a). Record the Cool, add 0.5 ml of water and a slight excess of 2 M
chromatograms and measure the responses for the hydrochloric acid and warm; sulphur dioxide is evolved, which
phenoxymethylpenicillin peak and any p-hydroxyphenoxy- turns moistened starch iodate paper blue.
methylpenicillin peak with a retention time of about 0.4 relative
to that of the main phenoxymethylpenicillin peak.
Tests

Calculate the content of C16H17N2O5S, from the sum of the Acidity or alkalinity. Dissolve 0.1 g in 10 ml of carbon dioxide-
peak responses of the p-hydroxyphenoxymethylpenicillin and free water and add 0.1 ml of methyl red solution. The solution
phenoxymethylpenicillin peaks in the chromatograms obtained is not red and not more than 0.05 ml of 0.1 M sodium hydroxide
with the test solution and reference solution (a) is required to change the colour of the solution.
Storage. Store protected from moisture. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
Labelling. The label states the strength in terms of the
equivalent amount of phenoxymethylpenicillin. Mobile phase. A mixture of 85 volumes of 2-butanone,
15 volumes of acetone and 5 volumes of strong ammonia
solution.

Phentolamine Mesylate Test solution. Dissolve 0.2 g of the substance under


examination in 10 ml of ethanol (95 per cent).
OH Reference solution. A 0.01 per cent w/v solution of the
CH3 substance under examination in ethanol (95 per cent).
Apply to the plate 10 µl of each solution. After development,
N , CH3SO3H dry the plate in air, spray with dilute potassium
N iodobismuthate solution. Any secondary spot in the
chromatogram obtained with the test solution is not more
HN intense than the spot in the chromatogram obtained with the
reference solution.
C17H19N3O,CH4O3S Mol. Wt. 377.5
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Phentolamine Mesylate is 3-[[4,5-dihydro-1H-imidazol-2-
yl)methyl](4-methylphenyl)aminophenol Loss on drying (2.4.19). Not more than 0.5 per cent, determined
methanesulphonate. on 1.0 g by drying in an oven at 105º.
Phentolamine Mesylate contains not less than 99.0 per cent Assay. Weigh accurately about 0.3 g and dissolve in 100 ml of
and not more than 100.5 per cent of C17H19N3O, CH4O3S, anhydrous 2-propanol with the aid of ultrasound if necessary.
calculated on the dried basis. Titrate with 0.1 M tetrabutylammonium hydroxide in
2-propanol. Determine the end-point potentiometrically
Identification (2.4.25), using a glass electrode and a calomel electrode
Test A may be omitted if tests B, C and D are carried out. Tests containing a saturated solution of tetramethylammonium
B, C and D may be omitted if test A is carried out. chloride in 2-propanol. Carry out a blank titration.

A. Determine by infrared absorption spectrophotometry (2.4.6). 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to


Compare the spectrum with that obtained with phentolamine 0.03775 g of C17H19N3O, CH4O3S.
mesylate RS or with the reference spectrum of phentolamine Storage. Store protected from light and moisture.
mesylate.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.002 per cent w/v solution shows an absorption maximum
only at about 278 nm; absorbance at about 278 nm, about 0.5. Phentolamine Injection
C. Dissolve 0.5 g in 5 ml of ethanol (95 per cent) and 5 ml of Phentolamine Mesylate Injection
0.1 M hydrochloric acid and add 2 ml of a 0.5 per cent w/v
solution of ammonium metavanadate; a light green precipitate Phentolamine Injection is a sterile solution of Phentolamine
is produced. Mesylate in Water for Injections containing Dextrose.

934
IP 2007 PHENYLBUTAZONE

Phentolamine Injection contains not less than 95.0 per cent B. When examined in the range 230 nm to 360 nm (2.4.7), a
and not more than 105.0 per cent of the stated amount of 0.001 per cent w/v solution in 0.01 M sodium hydroxide shows
phentolamine mesylate, C17H19N3O, CH4O3S. an absorption maximum at about 264 nm; absorbance at about
264 nm, about 0.66.
Identification
C. To 0.1 g add 1 ml of glacial acetic acid and 2 ml of
The residue obtained in the Assay melts at about 138º (2.4.21). hydrochloric acid and heat under a reflux condenser for 30
minutes. Cool, add 10 ml of water and filter. Add to the filtrate
Tests 3 ml of 0.1 M sodium nitrite; a yellow colour is produced. To
1 ml of this solution add a solution containing 10 mg of
pH (2.4.24). 3.5 to 5.0.
2-naphthol in 5 ml of sodium carbonate solution; a reddish
Other tests. Complies with the tests stated under Parenteral brown to reddish violet precipitate is produced.
Preparations (Injections).
Assay. Dilute an accurately measured volume containing about Tests
0.1 g of Phentolamine Mesylate to 40 ml with water, add 20 ml Appearance of solution (2.4.1). Dissolve 1.0 g in 20 ml of 2 M
of a 20 per cent w/v solution of trichloroacetic acid, allow to sodium hydroxide and keep the solution at 25º for 3 hours.
stand for 3 hours, filter, wash the residue with two quantities, The solution is clear.
each of 5 ml, of water and dry to constant weight at 105º.
Acidity or alkalinity. Heat to boiling 1.0 g with 50 ml of water,
1 g of the residue is equivalent to 0.8487 g of C17H19N3O, cool with shaking in a stoppered vessel and filter. To 25 ml of
CH4O3S. the filtrate add 0.5 ml of phenolphthalein solution; the
Storage. Store protected from light. solution is colourless and not more than 0.5 ml of 0.01 M
sodium hydroxide is required to change the colour of the
solution. Add 0.6 ml of 0.01 M hydrochloric acid and 0.1 ml of
methyl red solution; the solution is red or orange.
Phenylbutazone Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 50 volumes of chloroform,
40 volumes of cyclohexane and 10 volumes of glacial acetic
acid.

O N Prepare the following solutions immediately before use.


N
Test solution. Dissolve 0.2 g of the substance under
H3C examination in a solution containing 0.02 per cent w/v of
O
butylated hydroxytoluene in a mixture of equal volumes of
chloroform and ethanol and dilute to 5 ml with the same
C19H20N2O2 Mol. Wt. 308.4 solvent mixture.
Phenylbutazone is 4-butyl-1,2-diphenylpyrazolidine-3,5- Reference solution. Dilute 1 ml of the test solution to 200 ml
dione. with the same solvent mixture.
Phenylbutazone contains not less than 98.0 per cent and not Before use, spray the plate evenly with a 2 per cent w/v solution
more than 100.5 per cent of C19H20N2O2, calculated on the of sodium metabisulphite until thoroughly wet, dry the plate
dried basis. in air for 15 minutes, heat at 120º for 30 minutes and allow to
Description. A white or almost white, crystalline powder; cool. Apply separately to the plate 5 µl of each solution. After
odourless. development, dry the plate in a current of warm air for
10 minutes and examine in ultraviolet light at 254 nm. Any
Identification secondary spot in the chromatogram obtained with the test
solution is not more intense than the spot in the chromatogram
Test A may be omitted if tests B and C are carried out. Tests B
obtained with the reference solution.
and C may be omitted if test A is carried out.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
A. Determine by infrared absorption spectrophotometry (2.4.6).
heavy metals, Method B (20 ppm).
Compare the spectrum with that obtained with phenylbutazone
RS or with the reference spectrum of phenylbutazone. Sulphated ash (2.3.18). Not more than 0.1 per cent.

935
PHENYLBUTAZONE TABLETS IP 2007

Loss on drying (2.4.19). Not more than 0.5 per cent, determined Reference solution. Dilute 1 ml of the test solution to 200 ml
on 1.0 g by drying in an oven at 80º over phosphorous with the same solvent mixture.
pentoxide at a pressure of 1.5 to 2.5 kPa for 4 hours. Before use, spray the plate evenly with a 2 per cent w/v solution
Assay. Weigh accurately about 0.5 g, dissolve in 25 ml of of sodium metabisulphite until thoroughly wet, dry the plate
acetone and titrate with 0.1 M sodium hydroxide, using 0.5 ml in air for 15 minutes, heat at 120º for 30 minutes and allow to
of bromothymol blue solution as indicator and continuing cool. Apply to the plate 5 µl of each solution. After
the titration until the blue colour persists for at least development, dry the plate in a current of warm air for
15 seconds. Carry out a blank titration. 10 minutes and examine in ultraviolet light at 254 nm. Any
secondary spot in the chromatogram obtained with the test
1 ml of 0.1 M sodium hydroxide is equivalent to 0.03084 g of
solution is not more intense than the spot in the chromatogram
C19H20N2O2.
obtained with the reference solution.
Storage. Store protected from moisture.
Other tests. Complies with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.5 g of
Phenylbutazone Tablets Phenylbutazone and shake vigorously with 150 ml of 0.1 M
sodium hydroxide for 45 minutes. Add sufficient 0.1 M sodium
Phenylbutazone Tablets contain not less than 95.0 per cent hydroxide to produce 250.0 ml, mix and filter, rejecting the first
and not more than 105.0 per cent of the stated amount of 20 ml of the filtrate. To 5.0 ml of the filtrate add 50 ml of water
phenylbutazone, C19H20N2O2. The tablets are coated. and 4 ml of hydrochloric acid and extract with three quantities,
each of 30 ml, of ether. Combine the ether extracts and extract
Identification with three quantities, each of 30 ml, of 0.1 M sodium hydroxide.
Extract a quantity of the powdered tablets containing 0.2 g of Combine the aqueous extracts, pass nitrogen through the
Phenylbutazone with 40 ml of warm acetone, filter and solution to remove the residual ether and add sufficient 0.1 M
evaporate the filtrate to dryness. The residue complies with sodium hydroxide to produce 100.0 ml and mix well. To 10.0 ml
the following tests. of this solution add sufficient 0.1 M sodium hydroxide to
produce 100.0 ml and measure the absorbance of the resulting
A. When examined in the range 230 nm to 360 nm (2.4.7), a
solution at the maximum at about 264 nm (2.4.7), using 0.1 M
0.001 per cent w/v solution in 0.01 M sodium hydroxide shows
sodium hydroxide as the blank.
an absorption maximum at about 264 nm; absorbance at about
264 nm, about 0.66. Calculate the content of C19H20N2O2 from the absorbance
obtained by carrying out the Assay simultaneously on 0.5 g
B. To 0.1 g add 1 ml of glacial acetic acid and 2 ml of of phenylbutazone RS in place of the substance under
hydrochloric acid and heat under a reflux condenser for examination.
30 minutes. Cool, add 10 ml of water and filter. Add to the
filtrate 3 ml of 0.1 M sodium nitrite; a yellow colour is produced. Storage. Store protected from moisture.
To 1 ml of this solution add a solution containing 10 mg of
2-naphthol in 5 ml of sodium carbonate solution; a reddish
brown to reddish violet precipitate is produced.
Phenylephrine Hydrochloride
Tests
H OH H
Related substances. Determine by thin-layer chromatography HO N
(2.4.17), coating the plate with silica gel GF254. CH3 , HCl
Mobile phase. A mixture of 50 volumes of chloroform,
40 volumes of cyclohexane and 10 volumes of glacial acetic
C9H13NO2,HCl Mol. Wt. 203.7
acid.
Prepare the following solutions immediately before use. Phenylephrine Hydrochloride is (R)-1-(3-hydroxyphenyl)-2-
methylaminoethanol hydrochloride.
Test solution. Shake a quantity of the powdered tablets
Phenylephrine Hydrochloride contains not less than 98.5 per
containing 0.4 g of Phenylbutazone with 10 ml of a solution
cent and not more than 101.0 per cent of C9H13NO2, HCl,
containing 0.02 per cent w/v of butylated hydroxytoluene in a
calculated on the dried basis.
mixture of equal volumes of chloroform and ethanol for
15 minutes and centrifuge. Use the supernatant liquid. Description. A white or almost white, crystalline powder.

936
IP 2007 PHENYLEPHRINE INJECTION

Identification Sulphates (2.3.17). 15 ml of solution A complies with the limit


test for sulphates (500 ppm).
Test A may be omitted if tests B, C and D are carried out. Tests
B and C may be omitted if tests A and D are carried out. Phenones. To 10 ml of solution A add sufficient 0.01 M
hydrochloric acid to produce 50 ml. Absorbance of the
A. Determine by infrared absorption spectrophotometry (2.4.6). resulting solution at the maximum at about 310 nm, not more
Compare the spectrum with that obtained with phenylephrine than 0.20 (2.4.7).
hydrochloride RS or with the reference spectrum of
Sulphated ash (2.3.18). Not more than 0.1 per cent.
phenylephrine hydrochloride.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
B. Dissolve about 10 mg in 1 ml of water and add 0.05 ml of
on 1.0 g by drying in an oven at 105º.
cupric sulphate solution and 1 ml of 5 M sodium hydroxide;
a violet colour is produced. Add 1 ml of ether and shake; the Assay. Weigh accurately about 0.15 g, dissolve in a mixture of
ether layer remains colourless. 0.5 ml of 0.1 M hydrochloric acid and 80 ml of ethanol
(95 per cent) and titrate with 0.1 M ethanolic sodium
C. Dissolve 0.3 g in 3 ml of water, add 1 ml of 6 M ammonia hydroxide determining the end-point potentiometrically
and initiate crystallisation by scratching the side of the tube (2.4.25). Record the volume added between the two inflections.
with a glass rod. The melting range of the crystals, after
washing with iced water and drying at 105º for 2 hours, is 171º 1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
to 176º (2.4.21). 0.02037 g of C9H13NO2,HCl.

D. Gives reaction A of chlorides (2.3.1). Storage. Store protected from light and moisture.

Tests
Appearance of solution. Dissolve 2.0 g in 100 ml of carbon
Phenylephrine Injection
dioxide-free water prepared from distilled water (solution Phenylephrine Hydrochloride Injection
A). Solution A is clear (2.4.1), and colourless (2.4.1).
Phenylephrine Injection is a sterile solution of Phenylephrine
Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of Hydrochloride in Water for Injections.
methyl red solution and 0.2 ml of 0.01 M sodium hydroxide.
The solution is yellow and not more than 0.4 ml of 0.01 M Phenylephrine Injection contains not less than 95.0 per cent
hydrochloric acid is required to change the colour of the and not more than 105.0 per cent of the stated amount of
solution to red. phenylephrine hydrochloride, C9H13NO2,HCl.

Specific optical rotation (2.4.22). –43.0º to –47.0º, determined Identification


in solution A.
A. In the test for Related substances, the principal spot in the
Related substances. Determine by thin-layer chromatography chromatogram obtained with test solution (b) corresponds to
(2.4.17), coating the plate with silica gel H. that in the chromatogram obtained with reference solution (b).
Mobile phase. A mixture of 80 volumes of 2-propanol, B. To a volume containing 10 mg of Phenylephrine
15 volumes of 10 M ammonia and 5 volumes of chloroform. Hydrochloride add, if necessary, sufficient water to produce
Test solution. Dissolve 0.2 g of the substance under 1 ml and then add 0.05 ml of cupric sulphate solution and 1 ml
examination in 10 ml of methanol. of 5 M sodium hydroxide; a violet colour is produced. Add
1 ml of ether and shake; the ether layer remains colourless.
Reference solution (a). A 0.01 per cent w/v solution of the
substance under examination in methanol. C. Gives reaction A of chlorides (2.3.1).

Reference solution (b). A 0.004 per cent w/v solution of the Tests
substance under examination in methanol.
pH (2.4.24). 4.5 to 6.5.
Apply to the plate 5 µl of each solution. After development, Related substances. Determine by thin-layer chromatography
dry the plate in cold air, spray with ninhydrin solution, heat at (2.4.17), coating the plate with silica gel H.
105º for 10 minutes and examine in daylight. Any secondary
spot in the chromatogram obtained with the test solution is Mobile phase. A mixture of 80 volumes of 2-propanol,
not more intense than the spot in the chromatogram obtained 15 volumes of 10 M ammonia and 5 volumes of chloroform.
with reference solution (a) and not more than two such spots Test solution (a) Evaporate a volume of the injection containing
are more intense than the spot in the chromatogram obtained 20 mg of Phenylephrine Hydrochloride to dryness and dissolve
with reference solution (b). the residue in 1 ml of methanol.

937
PHENYLMERCURIC ACETATE IP 2007

Test solution (b). Dilute 1 volume of test solution (a) to a white precipitate is formed which darkens slowly on boiling
200 volumes with methanol. and allowing to stand.
Reference solution (a). Dilute 1 volume of test solution (b) to Tests
2.5 volumes with methanol.
Mercuric salts and heavy metals. Heat about 100 mg with
Reference solution (b). A 0.01 per cent w/v solution of
15 ml of water, cool and filter. To the filtrate add a few drops of
phenylephrine hydrochloride RS in methanol.
a freshly prepared 10 per cent w/v solution of sodium sulphide;
Apply to the plate 5 µl of each solution. After development, the precipitate formed shows no immediate colour.
dry the plate in cold air, spray with ninhydrin solution, heat at
Polymercurated benzene compounds. Shake 2.0 g with 100 ml
105º for 10 minutes and examine in daylight. Any secondary
of acetone. Filter, wash the residue with successive portions
spot in the chromatogram obtained with test solution (a) is
of acetone until a total of 50 ml is used, dry the residue at 105º
not more intense than the spot in the chromatogram obtained
for 1 hour and weigh. The weight of the residue does not
with test solution (b) and not more than two such spots are
exceed 30 mg (1.5 per cent).
more intense than the spot in the chromatogram obtained
with reference solution (a). Sulphated ash (2.3.18). Not more than 0.2 per cent.
Other tests. Complies with the tests stated under Parenteral Assay. Weigh accurately about 0.4 g, transfer to a 100-ml flask,
Preparations (Injections). add 15 ml of water, 5 ml of formic acid and 1 g of zinc dust and
reflux for 30 minutes. Cool, filter and wash the filter paper and
Assay. To an accurately measured volume containing about
the amalgam with water until the washings are no longer acidic
50 mg of Phenylephrine Hydrochloride add sufficient 0.5 M
to litmus. Dissolve the amalgam in 40 ml of 8 M nitric acid.
sulphuric acid to produce 100.0 ml. Dilute 10.0 ml of this
Heat on a water-bath for 3 minutes and add 0.5 g urea and
solution to 100.0 ml with 0.5 M sulphuric acid and measure
sufficient 0.02 M potassium permanganate to produce a
the absorbance of the resulting solution at the maximum at
permanent pink colour. Cool and add hydrogen peroxide
about 273 nm (2.4.7). Calculate the content of C9H13NO2,HCl
solution to decolorise the solution. Add 1 ml of ferric
taking 90 as the specific absorbance at 273 nm.
ammonium sulphate solution and titrate with 0.1 M ammonium
Storage. Store protected from light. thiocyanate. Repeat the operation without the substance under
examination. The difference between the titrations represents
the amount of ammonium thiocyanate required.
Phenylmercuric Acetate 1 ml of 0.1 M ammonium thiocyanate is equivalent to
0.01684 g of C8H8HgO2.
O
Storage. Store protected from light and moisture.
Hg O CH3

C8H8HgO2 Mol. Wt. 336.7 Phenylmercuric Nitrate


Phenylmercuric Acetate is (acetato)phenylmercury.
Phenylmercuric Nitrate is a mixture of phenylmercuric nitrate,
Phenylmercuric Acetate contains not less than 98.0 per cent C6H5HgNO3 and phenylmercuric hydroxide, C6H5HgOH.
and not more than 100.5 per cent of C8H8HgO2.
Phenylmercuric Nitrate contains not less than 62.5 per cent
Description. A white to creamy white, crystalline powder, or and not more than 63.5 per cent of mercury, Hg, calculated on
small white prisms or leaflets. the dried basis.

Identification Description. A white or pale yellow powder.

A. To 100 mg add 0.5 ml of nitric acid, warm gently until a dark Identification
brown colour is produced and dilute with water to 10 ml; the
A. To 0.1 g add 3 ml of sulphuric acid; the mixture becomes
characteristic odour of nitrobenzene is produced.
yellow and the characteristic odour of nitrobenzene is
B. To 100 mg add 0.5 ml of sulphuric acid and 1 ml of ethanol produced.
(95 per cent) and warm; the characteristic odour of ethyl
B. To 0.1 g add 45 ml of water and heat to boiling with shaking.
acetate is produced.
Cool, filter and add sufficient water to produce 50 ml (solution
C. To 5 ml of a saturated solution in water, add a few drops of A). To 1 ml of solution A add 1 ml of 2 M hydrochloric acid; a
a freshly prepared 10 per cent w/v solution of sodium sulphide; white, flocculent precipitate is produced.

938
IP 2007 PHENYTOIN SODIUM

C. To 5 ml of solution A add 8 ml of water and 0.1 ml of a Description. A white powder; odourless; somewhat
freshly prepared 10 per cent w/v solution of sodium sulphide; hygroscopic.
a white precipitate is formed which darkens slowly on boiling
and allowing to stand. Identification
D. To 5 ml of solution A add 1 ml of 2 M hydrochloric acid, Test A may be omitted if tests B, C and D are carried out. Tests
2 ml of dichloromethane and 0.2 ml of dithizone solution and B and C may be omitted if tests A and D are carried out.
shake; the lower layer is orange-yellow. A. Determine by infrared absorption spectrophotometry (2.4.6).
E. Solution A gives reaction of nitrates (2.3.1). Compare the spectrum with that obtained with phenytoin
sodium RS or with the reference spectrum of phenytoin sodium.
Tests
B. Dissolve 0.25 g in 5 ml of water and acidify with dilute
Appearance of solution. Solution A is colourless (2.4.1). hydrochloric acid; a white precipitate is produced.
Inorganic mercuric compounds. To a 10 ml of solution A add C. Dissolve 0.1 g in 10 ml of a 10 per cent w/v solution of
2 ml of potassium iodide solution and 3 ml of 2 M hydrochloric pyridine, add 1 ml of cupric sulphate with pyridine solution
acid and filter; the filtrate is colourless. Wash the precipitate and allow to stand for 10 minutes; a blue precipitate is
with 2 ml of water, combine the filtrate and washings and add produced.
2 ml of 2 M sodium hydroxide and sufficient water to produce
D. Incinerate 0.1 g; the residue after neutralisation with
20 ml. 12 ml of the solution complies with the limit test for
hydrochloric acid and addition of 2 ml of water gives the
heavy metals, Method A (2.3.13). Use lead standard solution
reactions of sodium salts (2.3.1).
(1 ppm Pb) to prepare the standard (0.1 per cent).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Tests
Loss on drying (2.4.19). Not more than 1.0 per cent, determined Appearance of solution. Suspend 1.0 g in 5 ml of water and
on 1.0 g by drying over phosphorus pentoxide at a pressure dilute to 20 ml with 0.1 M sodium hydroxide; the solution is
of 1.5 to 2.5 kPa for 24 hours. clear (2.4.1), and not more intensely coloured than reference
Assay. Weigh accurately about 0.2 g and dissolve in a mixture solution BYS6 (2.4.1).
of 90 ml of water and 10 ml of nitric acid. Add 2 ml of ferric Free phenytoin. Dissolve 0.3 g in 10 ml of a mixture of equal
ammonium sulphate solution and titrate with 0.1 M ammonium volumes of pyridine and water and add 0.5 ml of dilute
thiocyanate until a persistent reddish yellow colour is phenolphthalein solution and 3 ml of silver nitrate-pyridine
obtained. Repeat the operation without the substance under reagent. Not more than 1.0 ml of 0.1 M sodium hydroxide is
examination. The difference between the titrations represents required to change the colour of the solution to pink.
the amount of ammonium thiocyanate required.
Related substances. Determine by thin-layer chromatography
1 ml of 0.1 M ammonium thiocyanate is equivalent to 0.02006 (2.4.17), coating the plate with silica gel GF254.
g of Hg.
Mobile phase. A mixture of 45 volumes of chloroform,
Storage. Store protected from light and moisture. 45 volumes of 2-propanol and 10 volumes of strong ammonia
solution.
Test solution. Dissolve 0.4 g of the substance under
Phenytoin Sodium examination in 10 ml of methanol.
Diphenylhydantoin Sodium Reference solution (a). A 0.04 per cent w/v solution of the
substance under examination in methanol.
H
C6H5 N Reference solution (b). A 0.02 per cent w/v solution of
ONa benzophenone in methanol.
C6H5
N Apply to the plate 10 µl of each solution. After development,
O dry the plate at 80º for 5 minutes and examine in ultraviolet
C15H11N2NaO2 Mol. Wt. 274.3 light at 254 nm. Any secondary spot in the chromatogram
obtained with the test solution is not more intense than the
Phenytoin Sodium is 4-oxo-5,5-diphenyl-2-imidazolidin-2-olate spot in the chromatogram obtained with reference solution (a)
Phenytoin Sodium contains not less than 98.0 per cent and and any spot corresponding to benzophenone is not more
not more than 101.0 per cent of C15H11N2NaO2, calculated on intense than the spot in the chromatogram obtained with
the anhydrous basis. reference solution (b).

939
PHENYTOIN INJECTION IP 2007

Heavy metals (2.3.13). 1.0 g complies with the limit test for B. Dissolve 0.25 g in 5 ml of water and acidify with dilute
heavy metals, Method B (20 ppm). hydrochloric acid; a white precipitate is produced.
Water (2.3.43). Not more than 3.0 per cent, determined on 1.0 g. C. Dissolve 0.1 g in 10 ml of a 10 per cent w/v solution of
pyridine, add 1 ml of cupric sulphate with pyridine solution
Assay. Weigh accurately about 0.18 g, suspend in 2 ml of
and allow to stand for 10 minutes; a blue precipitate is
water, add 8 ml of 0.05 M sulphuric acid and heat gently for
produced.
1 minute. Add 30 ml of methanol, cool. Titrate with 0.1 M
sodium hydroxide, determining the end-point D. Incinerate 0.1 g; the residue after neutralisation with
potentiometrically (2.4.25). After the first inflection, stop the hydrochloric acid and addition of 2 ml of water gives the
addition of sodium hydroxide, add 5 ml of silver nitrate reactions of sodium salts (2.3.1).
solution in pyridine, mix and continue the titration until a
second inflection is reached. Record the volume of 0.1 M Tests
sodium hydroxide added between the two inflections.
Appearance of solution. Suspend 1.0 g in 5 ml of water and
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02743 g of dilute to 20 ml with 0.1 M sodium hydroxide; the solution is
C15H11N2NaO2. clear (2.4.1), and not more intensely coloured than reference
Storage. Store protected from moisture. solution BYS6 (2.4.1).
Completeness of solution. The contents dissolve in the
quantity of the solvent recommended on the label and give a
Phenytoin Injection clear solution.

Phenytoin Sodium Injection; Diphenylhydantoin Sodium pH (2.4.24). 10.0 to 12.0, determined in a 5.0 per cent w/v
injection solution in the stated solvent.

Phenytoin Injection is a sterile material consisting of Phenytoin Related substances. Determine by thin-layer chromatography
Sodium with or without buffering agents and other excipients. (2.4.17), coating the plate with silica gel GF254.
It is filled in a sealed container. Mobile phase. A mixture of 45 volumes of chloroform,
45 volumes of 2-propanol and 10 volumes of strong ammonia
The injection is constituted by dissolving the contents of the
solution.
sealed container in the requisite amount of sterile Water for
Injection or other suitable solvent, immediately before use. Test solution. Dissolve 0.4 g of the substance under
examination in 10 ml of methanol.
The constituted solution complies with the requirements for
Clarity of solution and Particulate matter stated under Reference solution (a). A 0.04 per cent w/v solution of the
Parenteral Preparations (Injections). substance under examination in methanol.
Storage. The constituted solution should be used immediately Reference solution (b). A 0.02 per cent w/v solution of
after preparation but, in any case, within the period benzophenone in methanol.
recommended by the manufacturer.
Apply to the plate 10 µl of each solution. After development,
Phenytoin Injection contains not less than 90.0 per cent and dry the plate at 80º for 5 minutes and examine in ultraviolet
not more than 110.0 per cent of the stated amount of phenytoin light at 254 nm. Any secondary spot in the chromatogram
sodium, C15H11N2NaO2. obtained with the test solution is not more intense than the
Description. A white powder; odourless; somewhat spot in the chromatogram obtained with reference solution (a)
hygroscopic. and any spot corresponding to benzophenone is not more
intense than the spot in the chromatogram obtained with
The contents of the sealed container comply with the reference solution (b).
requirements stated under Parenteral Preparations
(Powders for Injection) and with the following requirements. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Identification Water (2.3.43). Not more than 3.0 per cent, determined on
Test A may be omitted if tests B, C and D are carried out. Tests 1.0 g.
B and C may be omitted if tests A and D are carried out. Assay. Weigh accurately about 0.18 g of the mixed contents of
A. Determine by infrared absorption spectrophotometry (2.4.6). 10 containers, suspend in 2 ml of water, add 8 ml of 0.05 M
Compare the spectrum with that obtained with phenytoin sulphuric acid and heat gently for 1 minute. Add 30 ml of
sodium RS or with the reference spectrum of phenytoin sodium. methanol, cool. Titrate with 0.1 M sodium hydroxide,

940
IP 2007 PHOLCODINE

determining the end-point potentiometrically (2.4.25). After Reference solution. A 0.01 per cent w/v solution of
the first inflection, stop the addition of sodium hydroxide, add benzophenone in methanol.
5 ml of silver nitrate solution in pyridine, mix and continue Apply to the plate 5 µl of each solution. Allow the mobile
the titration until a second inflection is reached. Record the phase to rise 12 cm. Dry the plate in air and examine in ultraviolet
volume of 0.1 M sodium hydroxide added between the two light at 254 nm. In the chromatogram obtained with the test
inflections. solution any spot corresponding to benzophenone is not more
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02743 g of intense than the spot in the chromatogram obtained with the
C15H11N2NaO2. reference solution.
Storage. Store protected from moisture at a temperature not Other tests. Complies with the tests stated under Tablets.
exceeding 30º.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Labelling. The label states (1) the quantity of Phenytoin quantity of the powder containing about 0.25 g of Phenytoin
Sodium contained in it; (2) the directions for preparing the Sodium, shake with 40 ml of 0.01 M sodium hydroxide for
Injection. 5 minutes and add sufficient 0.01 M sodium hydroxide to
produce 50.0 ml. Centrifuge, acidify 25.0 ml of the clear liquid
with 10 ml of 0.1 M hydrochloric acid and extract successively
Phenytoin Tablets with 50, 40, 25 and 25 ml of ether. Wash the combined extracts
with 10 ml of water, evaporate to dryness and dry the residue
Phenytoin Sodium Tablets; Diphenylhydantoin Sodium at 105º. Dissolve in 50 ml of anhydrous pyridine and titrate
Tablets with 0.1 M tetrabutylammonium hydroxide, using 0.3 per cent
Phenytoin Tablets contain not less than 90.0 per cent and not w/v solution of thymol blue in pyridine as indicator and taking
more than 110.0 per cent of the stated amount of phenytoin care to prevent absorption of carbon dioxide from the
sodium, C15H11N2NaO2. The tablets are coated. atmosphere. Carry out a blank titration.
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
Identification 0.02743 g of C15H11N2NaO2.
A. Shake a quantity of the powdered tablets containing 0.1 g Storage. Store protected from moisture.
of Phenytoin Sodium with 20 ml of water, filter, acidify with
dilute hydrochloric acid and extract with 10 ml of chloroform.
Wash the chloroform extract with water, dry with anhydrous
sodium sulphate and evaporate to dryness and dry the residue Pholcodine
at 105º. The residue complies with the following test.
O
Determine by infrared absorption spectrophotometry (2.4.6). N
Compare the spectrum with that obtained with phenytoin O
sodium RS treated in the same manner or with the reference
O , H2O
spectrum of phenytoin. NCH3
B. Triturate a quantity of the powdered tablets containing 0.5 H
g of Phenytoin Sodium with 10 ml of water and filter. Acidify HO
with dilute hydrochloric acid; a white precipitate is produced.
C23H30N2O4,H2O Mol. Wt. 416.5
C. The powdered tablets, when moistened with hydrochloric
acid and introduced on a platinum wire into a flame, impart a Pholcodine is (5R,6S)-4,5-epoxy-N-methyl-3-(2-
yellow colour to the flame. morpholinoethoxy)morphin-7-en-6-ol monohydrate.
Pholcodine contains not less than 98.0 per cent and not more
Tests than 101.0 per cent of C23H30N2O4, calculated on the dried
Related substances. Determine by thin-layer chromatography basis.
(2.4.17), coating the plate with silica gel GF254. Description. A white or almost white, crystalline powder.
Mobile phase. A mixture of 75 volumes of hexane and
30 volumes of dioxan. Identification
Test solution. Shake a quantity of the powdered tablets A. Determine by infrared absorption spectrophotometry (2.4.6).
containing 0.1 g of Phenytoin Sodium with 5 ml of methanol, Compare the spectrum with that obtained with pholcodine RS
warm on a water-bath with shaking and filter. or with the reference spectrum of pholcodine.

941
PHOLCODINE LINCTUS IP 2007

B. To 10 ml of a 0.1 per cent w/v solution add 75 ml of water 1 ml of 0.1 M perchloric acid is equivalent to 0.01993 g of
and 10 ml of 1 M sodium hydroxide and dilute to 100 ml with C23H30N2O4.
water. When examined in the range 230 nm and 360 nm (2.4.7), Storage. Store protected from moisture.
the resulting solution shows an absorption maximum at about
284 nm; absorbance at about 284 nm, 0.36 to 0.39.
C. Dissolve 50 mg in 1 ml of sulphuric acid and add 0.05 ml of Pholcodine Linctus
a 10 per cent w/v solution of ammonium molybdate; a pale
blue colour is produced. Warm gently; the colour changes to Pholcodine Linctus is a solution containing 0.1 per cent w/v
deep blue. Add 0.05 ml of 2 M nitric acid; the colour changes solution of Pholcodine and 1 per cent w/v solution of Citric
to brownish red. Acid Monohydrate in a suitable flavoured vehicle.
Pholcodine Linctus contains not less than 0.090 per cent and
Tests not more than 0.110 per cent w/v of pholcodine, C23H30N2O4.
Specific optical rotation (2.4.22). –94.0º to –98.0º, determined Identification
at 20º in a 2.0 per cent w/v solution in ethanol (95 per cent).
To 20 ml add 20 ml of water, make alkaline to litmus paper with
Morphine. Dissolve 0.1 g in sufficient of 0.1 M hydrochloric
5 M ammonia, extract with two quantities, each of 20 ml, of
acid to produce 5 ml, add 2 ml of a 1 per cent w/v solution of
chloroform, washing each extract with 5 ml of water, dry the
sodium nitrite, allow to stand for 15 minutes and add 3 ml of
combined extracts with anhydrous sodium sulphate, filter and
6 M ammonia. The solution is not more intensely coloured
evaporate to dryness. If necessary, add 0.1 ml of ether and
than reference solution BS4 (2.4.1).
scratch the sides of the vessel with a glass rod to induce
Related substances. Determine by thin-layer chromatography crystallisation. The crystals, dried at a pressure not exceeding
(2.4.17), coating the plate with silica gel G. 2 kPa, comply with the following tests.
Mobile phase. A mixture of 70 volumes of ethanol (95 per A. Determine by infrared absorption spectrophotometry (2.4.6).
cent), 70 volumes of toluene, 65 volumes of acetone and Compare the spectrum with that obtained with pholcodine RS
5 volumes of strong ammonia solution. or with the reference spectrum of pholcodine.
Test solution. Dissolve 0.25 g of the substance under B. When examined in the range 230 nm and 360 nm (2.4.7), a
examination in 10 ml of chloroform. 0.01 per cent w/v solution in 0.01 M sodium hydroxide shows
an absorption maximum only at about 284 nm.
Reference solution (a). A 0.025 per cent w/v solution of the
substance under examination in chloroform. C. To a portion of the crystals add 0.05 ml of nitric acid and
mix; a yellow colour is produced.
Reference solution (b). A 0.0125 per cent w/v solution of the
D. Dissolve the remainder of the crystals in 1 ml of sulphuric
substance under examination in chloroform.
acid and add 0.05 ml of ammonium molybdate-sulphuric acid
Apply to the plate 10 µl of each solution. After development, solution; a pale blue colour is produced. Warm gently; the
dry the plate in air, spray with dilute potassium colour changes to deep blue. Add 0.05 ml of 2 M nitric acid;
iodobismuthate solution. Any secondary spot in the the colour changes to brownish red.
chromatogram obtained with the test solution is not more
intense than the spot in the chromatogram obtained with Tests
reference solution (a) and not more than one such spot of Other tests. Complies with the tests stated under Oral Liquids.
higher Rf value than the principal spot is more intense than
the spot in the chromatogram obtained with reference solution Assay. Weigh accurately about 50 g and add sufficient 5 M
(b). ammonia to make the solution alkaline to litmus paper, extract
with four quantities, each of 25 ml, of chloroform, washing
Sulphated ash (2.3.18). Not more than 0.1 per cent. each extract with the same 5 ml of water. Combine the extracts
Loss on drying (2.4.19). 3.9 to 4.5 per cent, determined on 0.5 and evaporate until the volume is reduced to 15 ml. Titrate
g by drying in an oven at 105º. with 0.02 M perchloric acid, using quinaldine red solution
as indicator. Carry out a blank titration.
Assay. Weigh accurately about 0.18 g, dissolve in 50 ml of
1 ml of 0.02 M perchloric acid is equivalent to 0.004165 g of
anhydrous glacial acetic acid, warming gently. Titrate with
C23H30N2O4,H2O.
0.1 M perchloric acid, determining the end-point
potentiometrically at the second inflection (2.4.25). Carry out Determine the weight per ml of the linctus (2.4.29), and calculate
a blank titration. the content of C23H30N2O4,H2O, weight in volume.

942
IP 2007 PHYSOSTIGMINE SALICYLATE

Storage. Store protected from light. heat on a water-bath for 5 minutes; the appearance of the
Labelling. The label states the strength in terms of the solution does not change.
equivalent amount of pholcodine. Assay. Weigh accurately about 1.0 g, add a solution of 10 g of
sodium chloride in 30 ml of water and titrate with1 M sodium
hydroxide using dilute phenolphthalein solution as indicator.
1 ml of 1 M sodium hydroxide is equivalent to 0.04900 g of
Phosphoric Acid H3PO4.
Orthophosphoric Acid; Concentrated Phosphoric Acid Storage. Store protected from moisture, in glass containers.
H3PO4 Mol. Wt. 98.0
Phosphoric acid contains not less than 84.0 per cent w/w
and not more than 90.0 per cent w/w of H3PO4. Physostigmine Salicylate
Description. A clear, colourless, syrupy liquid; corrosive. Eserine Salicylate
When kept at a low temperature it may solidify, producing a
mass of colourless crystals which do not melt until the COOH
H3C H CH3
temperature reaches 28º.
N N OH
Identification ,

A. Dilute with water; the solution is strongly acidic. CH3


O
B. Dilute 10.0 g to 150 ml with water (solution A). Neutralise O
with 2 M sodium hydroxide; the resulting solution gives the
H3C NH
reactions of phosphates (2.3.1).

Tests C15H21N3O2,C7H6O3 Mol. Wt. 413.5


Physostigmine Salicylate is (3aS,8aR)-1,2,3,3a,8,8a-
Appearance of solution. Solution A is clear (2.4.1), and
hexahydro-1,3a,8-trimethylpyrrolo[2,3-b]indol-5-yl
colourless (2.4.1).
methylcarbamate salicylate.
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
Physostigmine Salicylate contains not less than 98.5 per cent
of stannated hydrochloric acid; the resulting solution
and not more than 101.0 per cent of C15H21N3O2, C7H6O3,
complies with the limit test for arsenic (2 ppm).
calculated on the dried basis.
Heavy metals (2.3.13). Dilute 1.2 ml with 10 ml of water,
Description. A colourless or faintly yellow crystals, turning
neutralise with dilute ammonia solution, add sufficient dilute
red gradually under the action of air and light and rapidly in
acetic acid to render the solution acidic and then dilute to
presence of moisture; odourless.
25 ml with water. The resulting solution complies with the
limit test for heavy metals, Method A (10 ppm). Identification
Iron (2.3.14). 10 ml of solution A complies with the limit test for
Test A may be omitted if tests B, C and D are carried out. Test
iron (60 ppm).
C may be omitted if tests A, B and D are carried out.
Chlorides (2.3.12). 3 ml of solution A complies with the limit
A. Determine by infrared absorption spectrophotometry (2.4.6).
test for chlorides (50 ppm).
Compare the spectrum with that obtained with physostigmine
Sulphates (2.3.17). 20 ml of solution A complies with the limit salicylate RS.
test for sulphates (100 ppm).
B. In the test for Related substances, the principal spot in the
Alkali phosphates. To 1.7 g in a graduated cylinder add 6 ml of chromatogram obtained with test solution (b) corresponds to
ether and 2 ml of ethanol (95 per cent); no turbidity is that in the chromatogram obtained with reference solution (a).
produced.
C. To a 1 per cent w/v solution add 1 M sodium hydroxide; a
Aluminium and calcium. To 1.7 g add 10 ml of water and 8 ml white precipitate which turns pink is produced. The precipitate
of dilute ammonia solution; the solution remains clear. dissolves in an excess of the reagent, producing a red solution.
Hypophosphorus acid and phosphorous acid. To 5 ml of solution D. A 0.9 per cent w/v solution in carbon dioxide-free water
A add 2 ml of a 1.7 per cent w/v solution of silver nitrate and gives reaction A of salicylates (2.3.1).

943
PHYSOSTIGMINE INJECTIONS IP 2007

Tests the end-point potentiometrically (2.4.25). Carry out a blank


titration.
Appearance of solution. Dissolve 0.9 g without heating in
95 ml of carbon dioxide-free water prepared from distilled 1 ml of 0.1 M perchloric acid is equivalent to 0.04135 g of
water, and dilute to 100 ml with the same solvent (solution A). C15H21N3O2,C7H6O3.
The solution, examined immediately after preparation, is clear Storage. Store protected from light and moisture.
(2.4.1), and colourless (2.4.1).
pH (2.4.24). 5.1 to 5.9, determined in solution A immediately
after preparation.
Physostigmine Injection
Specific optical rotation (2.4.22). –90.0º to –94.0º, determined
in solution A immediately after preparation. Physostigmine Salicylate Injection; Eserine Salicylate
Injection
Eseridine. To 5 ml of solution A, examined immediately after
preparation, add a few crystals of potassium iodate and a Physostigmine Injection is a sterile solution of Physostigmine
drop of 2 M hydrochloric acid and 2 ml of chloroform and Salicylate in Water for Injections.
shake; the chloroform layer does not turn violet within 1 minute. Physostigmine Injection contains not less than 90.0 per cent
Related substances. Determine by thin-layer chromatography and not more than 110.0 per cent of the stated amount of
(2.4.17), coating the plate with silica gel G. physostigmine salicylate, C15H21N3O2, C7H6O3.

Mobile phase. A mixture of 100 volumes of cyclohexane, Identification


23 volumes of 2-propanol and 2 volumes of strong ammonia
solution. A. Warm a volume containing 3 mg of Physostigmine
Salicylate with 0.3 ml of 5 M ammonia; a yellowish red solution
Test solution (a). Dissolve 0.2 g of the substance under is produced which on evaporation gives a bluish residue.
examination in 10 ml of ethanol (95 per cent).
B. The residue obtained in test A dissolves in ethanol (95 per
Test solution (b). Dissolve 0.1 g of the substance under cent) producing a blue solution which, on the addition of
examination in 100 ml of ethanol (95 per cent). 6 M acetic acid, appears blue by transmitted light and exhibits
a red fluorescence which intensifies on dilution with water.
Reference solution (a). A 0.1 per cent w/v solution of
physostigmine salicylate RS in ethanol (95 per cent). C. The residue obtained in test A dissolves in sulphuric acid
producing a green solution which, on the gradual addition of
Reference solution (b). A 0.01 per cent w/v solution of
ethanol (95 per cent), changes to red but reverts to green
physostigmine salicylate RS in ethanol (95 per cent).
when the ethanol is evaporated.
Apply to the plate 20 µl of each solution. After development,
dry the plate in cold air, carry out a second chromatographic Tests
development in same direction, dry the plate in air and spray
pH (2.4.24). 4.0 to 6.0.
with freshly prepared acetic potassium iodobismuthate
solution and then with hydrogen peroxide solution (10 vol). Other tests. Complies with the tests stated under Parenteral
Examine the plate within 2 minutes. Any secondary spot in the Preparations (Injections).
chromatogram obtained with test solution (a) is not more Assay. Transfer an accurately measured volume containing
intense than the spot in the chromatogram obtained with 30 mg of Physostigmine Salicylate to a separator, add about
reference solution (b). 0.25 g of sodium bicarbonate and extract with six quantities,
Sulphates (2.3.17). 15 ml of solution A complies with the limit each of 15 ml, of chloroform. Filter the combined chloroform
test for sulphates (0.1 per cent). extracts through about 10 g of anhydrous sodium sulphate.
Add 25 ml of anhydrous glacial acetic acid to the filtrate.
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined Titrate with 0.1 M perchloric acid, determining the end-point
on the residue obtained in the test for Loss on drying. potentiometrically (2.4.25). Carry out a blank titration.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined 1 ml of 0.01 M perchloric acid is equivalent to 0.004135 g of
on 1.0 g by drying in an oven at 105º. C15H21N3O2, C7H6O3.
Assay. Weigh accurately about 0.35 g, dissolve in 50 ml of a Storage. Store protected from light, in single dose containers.
mixture of equal volumes of chloroform and anhydrous glacial The injection should not be used if it is more than slightly
acetic acid. Titrate with 0.1 M perchloric acid, determining discoloured.

944
IP 2007 PINDOLOL

Pilocarpine Nitrate Test solution (b). Dissolve 0.1 g of the substance under
examination in 100 ml of water.
N O O Reference solution (a). A 0.1 per cent w/v solution of
CH3 , pilocarpine nitrate RS in water.
HNO3
N Reference solution (b). A 0.03 per cent w/v solution of
H3C pilocarpine nitrate RS in water.
C11H16N2O2, HNO3 Mol. Wt. 271.3 Apply to the plate 10 µl of each solution. After development,
dry the plate at 105º for 10 minutes, cool and spray with
Pilocarpine Nitrate is (3S,4R)-3-ethyl-4-[(1-methyl-1H-
potassium iodobismuthate solution. Any secondary spot in
imidazol-5-yl)methyl]dihydrofuran-2(3H)-one nitrate.
the chromatogram obtained with test solution (a) is not more
Pilocarpine Nitrate contains not less than 98.5 per cent and intense than the spot in the chromatogram obtained with
not more than 101.0 per cent of C11H16N2O2, HNO3, calculated reference solution (b).
on the dried basis.
Other alkaloids. To a 1 per cent w/v solution add dilute
Description. Colourless crystals or a white, crystalline powder; ammonia solution; no turbidity is produced. To a 1 per cent
odourless; sensitive to light. w/v solution add a few drops of potassium dichromate
solution; no turbidity is produced.
Identification
Chlorides (2.3.12). 25 ml of a 10.0 per cent w/v solution complies
Test A may be omitted if tests B, C and D are carried out. Tests with the limit test for chlorides (100 ppm).
B and C may be omitted if tests A and D are carried out.
Iron (2.3.14). 20 ml of a 10.0 per cent w/v solution complies
A. Determine by infrared absorption spectrophotometry (2.4.6). with the limit test for iron (20 ppm).
Compare the spectrum with that obtained with pilocarpine
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined
nitrate RS.
on 0.5 g.
B. In the test for Related substances, the principal spot in the
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
chromatogram obtained with test solution (b) corresponds to
on 1.0 g by drying in an oven at 105º.
that in the chromatogram obtained with reference solution (a).
C. Dissolve about 10 mg in 2 ml of water, add 2 drops of a 5 per Assay. Weigh accurately about 0.25 g, dissolve in 30 ml of
cent w/v solution of potassium dichromate, 1 ml of hydrogen anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
peroxide solution (10 vol) and 2 ml of chloroform and shake; acid, determining the end-point potentiometrically (2.4.25).
the chloroform layer turns violet. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.02713 g of
D. Gives reaction A of nitrates (2.3.1).
C11H16N2O2, HNO3.
Tests Storage. Store protected from light and moisture.
Appearance of solution. A 5.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution YS6 (2.4.1). Pindolol
pH (2.4.24). 3.5 to 4.5, determined in a 5.0 per cent w/v solution
prepared immediately before use in carbon dioxide-free water. OH H
HN O N CH3
Specific optical rotation (2.4.22). +79.5º to +83.0º, determined
in a 5.0 per cent w/v solution in carbon dioxide-free water, CH3
prepared immediately before use.
C14H20N2O2 Mol. wt. 248.3
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G. Pindolol is (RS)-1-indol-4-yloxy-3-isopropylaminopropan-2-
ol.
Mobile phase. A mixture of 85 volumes of chloroform,
14 volumes of methanol and 1 volume of strong ammonia Pindolol contains not less than 99.0 per cent and not more
solution. than 101.0 per cent of C14H20N2O2, calculated on the dried
basis.
Test solution (a). Dissolve 0.3 g of the substance under
examination in 10 ml of water. Description. A white or almost white, crystalline powder.

945
PINDOLOL TABLETS IP 2007

Identification Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method C (20 ppm).
Test A may be omitted if tests B and C are carried out. Test B
and C may be omitted if test A is carried out. Sulphated ash (2.3.18). Not more than 0.1 per cent.

A. Determine by infrared absorption spectrophotometry (2.4.6). Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Compare the spectrum with that obtained with pindolol RS. on 1.0 g by drying in an oven at 105º.

B. When examined in the range 230 nm to 360 nm (2.4.7), a Assay. Weigh accurately about 0.2 g, dissolve in 80 ml of
0.002 per cent w/v solution in a 0.085 per cent v/v solution of methanol. Titrate with 0.1 M hydrochloric acid, determining
hydrochloric acid in methanol shows absorption maxima at the end point potentiometrically (2.4.25).
about 264 nm and at about 287 nm, and a shoulder at about 1 ml of 0.1 M hydrochloric acid is equivalent to 0.02483 g of
275 nm, absorbances at about 264 nm, 0.66 to 0.70 and at about C14H20N2O2.
287 nm, 0.34 to 0.38.
Storage. Store protected from light and moisture.
C. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution (a).

Tests
Pindolol Tablets
Pindolol Tablets contains not less than 90.0 per cent and not
Appearance of solution. A 5.0 per cent w/v solution in dilute more than 110.0 per cent of the stated amount of pindolol,
acetic acid is clear (2.4.1), and not more intensely coloured C14H20N2O2.
than reference solution BYS4 (2.4.1).
Related substances. Determine as rapidly as possible, Identification
protected from light. A. Shake a quantity of the powdered tablets containing 50 mg
Determine by thin-layer chromatography (2.4.17), coating the of Pindolol with 80 ml of ether for 30 minutes, filter and dry the
plate with silica gel GF254. extract with anhydrous sodium sulphate. Filter the extract,
remove the ether using a rotary evaporator and dry the residue
Mobile phase. A freshly prepared mixture of 50 volumes of over phosphorus pentoxide at 110º at a pressure not exceeding
ethyl acetate, 50 volumes of methanol and 4 volumes of strong 2 kPa for 1 hour. The residue complies with the following test.
ammonia solution.
Determine by infrared absorption spectrophotometry (2.4.6).
Prepare the following solutions immediately before use. Compare the spectrum with that obtained with pindolol RS.
Test solution (a). Dissolve 0.2 g of the substance under B. When examined in the range 230 nm to 360 nm (2.4.7), the
examination in 10 ml of a mixture of 99 volumes of methanol final solution obtained in the Assay shows absorption maxima
and 1 volume of anhydrous glacial acetic acid. at about 264 nm and 287 nm.
Test solution (b). Dissolve 0.2 g of the substance under C. Shake a quantity of the powdered tablets containing 20 mg
examination in 100 ml of a mixture of 99 volumes of methanol of Pindolol with 5 ml of a mixture of 99 volumes of methanol
and 1 volume of anhydrous glacial acetic acid. and 1 volume of glacial acetic acid for 45 minutes Centrifuge
Reference solution (a). A 0.2 per cent w/v solution of pindolol and dilute 1 ml of the supernatant liquid to 50 ml with the
RS in a mixture of 99 volumes of methanol and 1 volume of acetic acid-methanol mixture. To 2 ml of this solution add 1 ml
anhydrous glacial acetic acid. of dimethylaminobenzaldehyde solution; a violet-blue colour
is produced.
Reference solution (b). A 0.006 per cent w/v solution of
pindolol RS in a mixture of 99 volumes of methanol and Tests
1 volume of anhydrous glacial acetic acid.
Related substances. Determine as rapidly as possible,
Apply to the plate 5 µl of each solution. Allow the mobile protected from light.
phase to rise 10 cm. Dry the plate immediately in a current of
Determine by thin-layer chromatography (2.4.17), coating the
cold air and examine in ultraviolet at 254 nm without delay.
plate with silica gel GF254.
Any secondary spot in the chromatogram obtained with test
solution (a) is not more intense than the spot in the Mobile phase. A freshly prepared mixture of 50 volumes of
chromatogram obtained with reference solution (b) (0.3 per ethyl acetate, 50 volumes of methanol and 4 volumes of strong
cent). ammonia solution.

946
IP 2007 PIPERAZINE ADIPATE

Test solution. Shake a quantity of the powdered tablets Identification


containing 20 mg of Pindolol with 5 ml of a mixture of 99 volumes
of methanol and 1 volume of glacial acetic acid for 15 minutes, Test A may be omitted if tests B, C and D are carried out. Tests
centrifuge and apply the supernatant liquid to the plate as the B and C may be omitted if tests A and D are carried out.
last solution. A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution (a). Dilute 1 volume of the test solution to Compare the spectrum with that obtained with piperazine
10 volumes with the methanol-acetic acid mixture and further adipate RS or with the reference spectrum of piperazine
dilute 7 volumes of the solution to 100 volumes with the same adipate.
solvent mixture. B. In the test for Related substances, examine the plate after
Reference solution (b). Dilute 1 volume of the test solution to spraying with both the ninhydrin and iodine solutions. The
10 volumes with the methanol-acetic acid mixture and further principal spot in the chromatogram obtained with test solution
dilute 3 volumes of this solution to 100 volumes with the same (b) corresponds to that in the chromatogram obtained with
solvent mixture. reference solution (a).
Apply to the plate 10 µl of each solution. Allow the mobile C. Dissolve 0.1 g in 5 ml of water, add 0.5 g of sodium
phase to rise 10 cm. Dry the plate in air, spray immediately with bicarbonate, 0.5 ml of a freshly prepared 5.0 per cent w/v
dimethylaminobenzaldehyde solution and warm at 50º for solution of potassium ferricyanide and 0.1 ml of mercury,
20 minutes. Any spot with Rf value of about 0.1 in the shake vigorously for 1 minute and allow to stand for
chromatogram obtained with the test solution is not more 20 minutes; a reddish colour slowly develops.
intense than the spot in the chromatogram obtained with
reference solution (a) (0.7 per cent). Any other secondary Tests
spot in the chromatogram obtained with the test solution is Appearance of solution. A 5.0 per cent w/v solution is clear
not more intense than the spot in the chromatogram obtained (2.4.1), and not more intensely coloured than reference solution
with reference solution (b) (0.3 per cent). Ignore any spot BS8 (2.4.1).
remaining on the line of application.
pH (2.4.24). 5.0 to 6.0, determined in a 5.0 per cent w/v solution.
Other tests. Comply with the tests stated under tablets.
Related substances. Determine by thin-layer chromatography
Assay. Weigh and powder 20 tablets. Weigh accurately a (2.4.17), coating the plate with silica gel G.
quantity of the powdered tablets containing about 90 mg of
Pindolol and shake with 100.0 ml of methanol for 45 minutes. Mobile phase. A freshly prepared mixture of 80 volumes of
Centrifuge and dilute 15.0 ml of the supernatant liquid to acetone and 20 volumes of strong ammonia solution.
100.0 ml with methanol and measure the absorbance of the Test solution (a). Dissolve 1 g of the substance under
resulting solution at the maximum at about 264 nm (2.4.7). examination in 10 ml of a mixture of 3 volumes of strong
Calculate the content of C14H20N2O2 taking 338 as the specific ammonia solution and 2 volumes of ethanol.
absorbance at 264 nm.
Test solution (b). Dissolve 1 g of the substance under
Storage. Store protected from light. examination in 100 ml of the same solvent mixture.
Reference solution (a). A 1 per cent w/v solution of piperazine
adipate RS in the same solvent mixture.
Piperazine Adipate Reference solution (b). A 0.025 per cent w/v solution of
ethylenediamine in the same solvent mixture.
H
N Reference solution (c). A 0.025 per cent w/v solution of
, COOH triethylenediamine in the same solvent mixture.
HOOC
N Reference solution (d). A solution containing 0.025 per cent
H w/v of triethylenediamine and 1 per cent w/v of the substance
under examination in the same solvent mixture. .
C4H10N2, C6H10O4 Mol. Wt. 232.3
Apply to the plate 5 µl of each solution. After development,
Piperazine Adipate contains not less than 98.0 per cent and dry the plate at 105º, spray with a 0.3 per cent w/v solution of
not more than 101.0 per cent of C4H10N2, C6H10O4, calculated ninhydrin in a mixture of 3 volumes of anhydrous acetic acid
on the anhydrous basis. and 100 volumes of 1-butanol and then with a 0.15 per cent
Description. A white, crystalline powder. w/v solution of ninhydrin in ethanol and dry at 105º for 10

947
PIPERAZINE ADIPATE TABLETS IP 2007

minutes. Any secondary spot in the chromatogram obtained Tests


with test solution (a) is not more intense than the spot in the
chromatogram obtained with reference solution (b). Spray the Other tests. Comply with the tests stated under Tablets.
plate with 0.05 M iodine and allow to stand for about 10 Assay. Weigh and powder 20 tablets. Weigh accurately a
minutes. Any spot corresponding to triethylenediamine in quantity of the powder containing about 0.2 g of Piperazine
the chromatogram obtained with test solution (a) is not more Hydrate, shake with 10 ml of water for 1 hour, filter and wash
intense than the spot in the chromatogram obtained with the residue with two quantities, each of 10 ml, of water. To the
reference solution (c). The test is not valid unless the combined extract and washings add 5 ml of 1 M sulphuric
chromatogram obtained with reference solution (d) shows two acid and 50 ml of picric acid solution, bring to boil, allow to
separated spots. Ignore any spot remaining on the line of stand for 1 hour and filter through a sintered-glass crucible
application. (porosity No. 4) and wash the residue with successive
Heavy metals (2.3.13). Dissolve 1.0 g in 20 ml of water, 0.5 ml of quantities, each of 10 ml, of a mixture of equal volumes of a
0.1 M hydrochloric acid and add sufficient water to produce saturated solution of picric acid and water until the washings
25 ml. The resulting solution complies with the limit test for are free from sulphate. Wash with five quantities, each of
heavy metals, Method A (20 ppm). 10 ml, of ethanol and dry to constant weight at 105º.
1 g of the residue is equivalent to 0.3567 g of C4H10N2, 6H2O.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Storage. Store protected from moisture.
Water (2.3.43). Not more than 0.5 per cent, determined on
1.0 g. Labelling. The label states the strength in terms of the
equivalent amount of piperazine hydrate.
Assay. Weigh accurately about 0.2 g, dissolve in 10 ml of
anhydrous glacial acetic acid with gentle heating and dilute
to 70 ml with the same solvent. Titrate with 0.1 M perchloric
acid, using 0.25 ml of 1-naphtholbenzein solution as indicator Piperazine Citrate
and titrating until the colour of the solution changes from
brownish yellow to green. Carry out a blank titration.
H
1 ml of 0.1 M perchloric acid is equivalent to 0.01161 g of N COOH
HO
C4H10N2, C6H10O4. ,
HOOC COOH
Storage. Store protected from moisture. N
H
3 2

(C4H10N2)3,2C6H8O7 Mol. Wt. 642.7 (anhydrous)


Piperazine Adipate Tablets Piperazine Citrate is a salt of piperazine with citric acid
containing a variable amount of water of crystallisation.
Piperazine Adipate Tablets contain not less than 92.5 per cent
and not more than 107.5 per cent of the stated amount of Piperazine Citrate contains not less than 98.0 per cent and not
piperazine hydrate, C4H10N2, 6H2O. more than 101.0 per cent of (C4H10N2)3, 2C6H8O7, calculated on
the anhydrous basis.
Identification Description. A white, granular powder; almost odourless.
A. Extract a quantity of the powdered tablets containing 1 g of Identification
Piperazine Hydrate with 20 ml of water and filter. Dilute 1 ml of
the filtrate to 5 ml with water, add 0.5 g of sodium bicarbonate, Test A may be omitted if tests B and C are carried out. Test B
0.5 ml of a freshly prepared 5.0 per cent w/v solution of may be omitted if tests A and C are carried out.
potassium ferricyanide and 0.1 ml of mercury, shake A. Determine by infrared absorption spectrophotometry (2.4.6).
vigorously for 1 minute and allow to stand for 20 minutes; a Compare the spectrum with that obtained with piperazine
reddish colour slowly develops. citrate RS or with the reference spectrum of piperazine citrate.
B. To 10 ml of the filtrate obtained in test A add 5 ml of B. In the test for Related substances, examine the plate after
hydrochloric acid and extract with three quantities, each of spraying with both the ninhydrin solutions. The principal
10 ml, of ether, evaporate the combined ether extracts to spot in the chromatogram obtained with test solution (b)
dryness; the residue, after washing with a small volume of corresponds to that in the chromatogram obtained with
water and drying at 105º, melts at about 152º (2.4.21). reference solution (a).

948
IP 2007 PIPERAZINE CITRATE SYRUP

C. A 10 per cent w/v solution gives the reactions of citrates to 70 ml with the same solvent. Titrate with 0.1 M perchloric
(2.3.1). acid, using 0.25 ml of 1-naphtholbenzein solution as indicator
and titrating until the colour of the solution changes from
Tests brownish yellow to green. Carry out a blank titration.
Appearance of solution. A 5.0 per cent w/v solution is clear 1 ml of 0.1 M perchloric acid is equivalent to 0.01071 g of
(2.4.1), and not more intensely coloured than reference solution (C4H10N2)3, 2C6H8O7.
BS8 (2.4.1).
Storage. Store protected from light and moisture.
pH (2.4.24). 5.0 to 6.0, determined in a 5.0 per cent w/v solution.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
Mobile phase. A freshly prepared mixture of 80 volumes of Piperazine Citrate Syrup
acetone and 20 volumes of strong ammonia solution. Piperazine Citrate Oral Solution; Piperazine Citrate Elixir
Test solution (a). Dissolve 1 g of the substance under Piperazine Citrate Syrup is a solution of Piperazine Citrate in a
examination in 10 ml of a mixture of 3 volumes of strong suitable flavoured Vehicle.
ammonia solution and 2 volumes of ethanol.
Piperazine Citrate Syrup contains the equivalent of not less
Test solution (b). Dissolve 1 g of the substance under
than 92.5 per cent and not more than 107.5 per cent of the
examination in 100 ml of the same solvent mixture.
stated amount of piperazine hydrate, C4H10N2, 6H2O.
Reference solution (a). A 1 per cent w/v solution of piperazine
citrate RS in the same solvent mixture. Identification
Reference solution (b). A 0.025 per cent w/v solution of A. To 1 ml add 5 ml of 2 M hydrochloric acid and, with stirring,
ethylenediamine in the same solvent mixture. 1 ml of a freshly prepared 50 per cent w/v solution of sodium
Reference solution (c). A 0.025 per cent w/v solution of nitrite, cool in ice for 15 minutes, induce crystallisation, wash
triethylenediamine in the same solvent mixture. the crystalline precipitate with water and dry at 105º. The
crystals melt at about 159º (2.4.21).
Reference solution (d). A solution containing 0.025 per cent
w/v of triethylenediamine and 1 per cent w/v of the substance B. Warm 10 ml with activated charcoal and filter. Boil a portion
under examination in the same solvent mixture. of the filtrate with an excess of mercuric sulphate solution,
Apply to the plate 5 µl of each solution. After development, filter, boil the filtrate and add 0.25 ml of dilute potassium
dry the plate at 105º, spray with a 0.3 per cent w/v solution of permanganate solution; the permanganate solution is
ninhydrin in a mixture of 3 volumes of anhydrous acetic acid decolorised and a white precipitate is produced.
and 100 volumes of 1-butanol and then with a 0.15 per cent C. Acidify a portion of the filtrate obtained in test B with 1 M
w/v solution of ninhydrin in ethanol and dry at 105º for sulphuric acid, add 0.25 ml of dilute potassium permanganate
10 minutes. Any secondary spot in the chromatogram obtained solution, warm until the colour is discharged and add an excess
with test solution (a) is not more intense than the spot in the of bromine water; a white precipitate is produced either
chromatogram obtained with reference solution (b). Spray the immediately or on cooling.
plate with 0.05 M iodine and allow to stand for about
10 minutes. Any spot corresponding to triethylenediamine in Tests
the chromatogram obtained with test solution (a) is not more
Other tests. Complies with the tests stated under Oral Liquids.
intense than the spot in the chromatogram obtained with
reference solution (c). The test is not valid unless the Assay. Weigh accurately a quantity containing about 0.2 g of
chromatogram obtained with reference solution (d) shows two Piperazine Hydrate, add 3.5 ml of 0.5 M sulphuric acid and
separated spots. Ignore any spot remaining on the line of 10 ml of water, add 100 ml of picric acid solution, heat on a
application. water-bath for 15 minutes, allow to stand for 1 hour and filter
through a sintered-glass crucible (porosity No. 4). Wash the
Heavy metals (2.3.13). 1.0 g complies with the limit test for
residue with successive quantities, each of 10 ml, of a mixture
heavy metals, Method A (20 ppm).
of equal volumes of a saturated solution of picric acid and
Sulphated ash (2.3.18). Not more than 0.1 per cent. water until the washings are free from sulphate. Wash the
Water (2.4.19). 10.0 to 14.0 per cent, determined on 0.3 g. residue with five quantities, each of 10 ml, of ethanol and dry
to constant weight at 105º.
Assay. Weigh accurately about 0.2 g, dissolve in 10 ml of
anhydrous glacial acetic acid with gentle heating and dilute 1 g of the residue is equivalent to 0.3567 g of C4H10N2, 6H2O.

949
PIPERAZINE HYDRATE IP 2007

Determine the weight per ml of the syrup (2.4.29), and calculate Test solution (a). Dissolve 1 g of the substance under
the content of C4H10N2, 6H2O, weight in volume. examination in 10 ml of a mixture of 3 volumes of strong
Storage. Store protected from light. ammonia solution and 2 volumes of ethanol.
Test solution (b). Dissolve 1 g of the substance under
Labelling. The label states the strength in terms of the
examination in 100 ml of the same solvent mixture.
equivalent amount of piperazine hydrate.
Reference solution (a). A 1 per cent w/v solution of piperazine
hydrate RS in the same solvent mixture.
Reference solution (b). A 0.025 per cent w/v solution of
Piperazine Hydrate ethylenediamine in the same solvent mixture.
Reference solution (c). A 0.025 per cent w/v solution of
H triethylenediamine in the same solvent mixture.
N
Reference solution (d). A solution containing 0.025 per cent
, 6H2O
w/v of triethylenediamine and 1 per cent w/v of the substance
N under examination in the same solvent mixture.
H
Apply to the plate 5 µl of each solution. After development,
C4H10N2, 6H2O Mol. Wt. 194.2 dry the plate at 105º, spray with a 0.3 per cent w/v solution of
ninhydrin in a mixture of 3 volumes of anhydrous acetic acid
Piperazine Hydrate contains not less than 98.0 per cent and
and 100 volumes of 1-butanol and then with a 0.15 per cent
not more than 101.0 per cent of C4H10N2, 6H2O.
w/v solution of ninhydrin in ethanol and dry at 105º for
Description. Colourless, glassy deliquescent crystals. 10 minutes. Any secondary spot in the chromatogram obtained
with test solution (a) is not more intense than the spot in the
Identification chromatogram obtained with reference solution (b). Spray the
Test A may be omitted if tests B, C and D are carried out. Tests plate with 0.05 M iodine and allow to stand for about
B and C may be omitted if tests A and D are carried out. 10 minutes. Any spot corresponding to triethylenediamine in
the chromatogram obtained with test solution (a) is not more
A. Determine by infrared absorption spectrophotometry (2.4.6). intense than the spot in the chromatogram obtained with
Compare the spectrum with that obtained with piperazine reference solution (c). The test is not valid unless the
hydrate RS. chromatogram obtained with reference solution (d) shows two
B. In the test for Related substances, examine the plate after separated spots. Ignore any spot remaining on the line of
spraying with both the ninhydrin solutions. The principal spot application.
in the chromatogram obtained with test solution (b) Heavy metals (2.3.13).1.0 g complies with the limit test for
corresponds to that in the chromatogram obtained with heavy metals, Method A (20 ppm).
reference solution (a). Sulphated ash (2.3.18). Not more than 0.1 per cent.
C. Dissolve 0.2 g in 5 ml of dilute hydrochloric acid, add 0.5 g Assay. Weigh accurately about 0.2 g, dissolve in 10 ml of
of sodium nitrite and heat to boiling. Cool in ice for 15 minutes, anhydrous glacial acetic acid with gentle heating and dilute
scratching the walls of the container with a glass rod and to 70 ml with the same solvent. Titrate with 0.1 M perchloric
filter. The crystals, after washing with 10 ml of ice-cold water acid, using 0.25 ml of 1-naphtholbenzein solution as indicator
and drying at 105º, melt at about 159º (2.4.21). and titrating until the colour of the solution changes from
brownish yellow to green. Carry out a blank titration.
Tests
1 ml of 0.1 M perchloric acid is equivalent to 0.009705 g of
Appearance of solution. A 5.0 per cent w/v solution in carbon C4H10N2, 6H2O.
dioxide-free water is clear (2.4.1), and not more intensely Storage. Store protected from light and moisture.
coloured than reference solution BS8 (2.4.1).
pH (2.4.24). 10.5 to 12.0, determined in a 5.0 per cent w/v
solution in carbon dioxide-free water. Piperazine Phosphate
Related substances. Determine by thin-layer chromatography C4H10N2, H3PO4, H2O Mol. Wt. 202.2
(2.4.17), coating the plate with silica gel G.
Piperazine Phosphate contains not less than 98.5 per cent
Mobile phase. A freshly prepared mixture of 80 volumes of and not more than 100.5 per cent of C4H10N2, H3PO4,
acetone and 20 volumes of strong ammonia solution. calculated on the anhydrous basis.

950
IP 2007 PIROXICAM

Description. A white, crystalline powder; odourless or almost A. To 4 ml of the filtrate, add 1 ml of hydrochloric acid, 0.5 g
odourless. of sodium nitrite and heat to boiling. Cool in ice for 15 minutes,
scratching the walls of the container with a glass rod and
Identification filter. The crystals, after washing with 10 ml of ice-cold water
and drying at 105º, melt at about 159º (2.4.21).
A. Dissolve 0.2 g in 5 ml of dilute hydrochloric acid, add
0.5 g of sodium nitrite and heat to boiling. Cool in ice for B. Dilute 1 ml of the filtrate to 5 ml with water, add 0.5 g of
15 minutes, scratching the walls of the container with a glass sodium bicarbonate, 0.5 ml of a freshly prepared 5.0 per cent
rod and filter. The crystals, after washing with 10 ml of ice- w/v solution of potassium ferricyanide and 0.1 ml of mercury,
cold water and drying at 105º, melt at about 159º (2.4.21). shake vigorously for 1 minute and allow to stand for
20 minutes; a reddish colour slowly develops.
B. Dissolve 0.1 g in 5 ml of water, add 0.5 g of sodium
bicarbonate, 0.5 ml of a freshly prepared 5.0 per cent w/v C. Gives the reactions of phosphates (2.3.1).
solution of potassium ferricyanide and 0.1 ml of mercury,
shake vigorously for 1 minute and allow to stand for Tests
20 minutes; a reddish colour slowly develops. Disintegration. The test does not apply to Piperazine
C. Gives the reactions of phosphates (2.3.1). Phosphate Tablets intended to be chewed before swallowing.

Tests Other tests. Comply with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Weigh accurately a
pH (2.4.24). 6.0 to 6.5, determined in a 1.0 per cent w/v solution. quantity of the powder containing about 0.15 g of Piperazine
Heavy metals (2.3.13). Dissolve 1.0 g in 20 ml of 2 M acetic Phosphate, shake with 10 ml of water for 1 hour, filter and
acid and add sufficient water to produce 25 ml. The solution wash the residue with two quantities, each of 10 ml, of water.
complies with the limit test for heavy metals, Method A To the combined extract and washings add 5 ml of 1 M
(20 ppm). sulphuric acid and 50 ml of picric acid solution, boil, allow
the mixture to stand for several hours and filter through a
Water (2.3.43). 8.0 to 9.5 per cent, determined on 0.25 g.
sintered glass crucible (porosity No. 4). Wash the residue
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of with successive quantities, each of 10 ml, of a mixture of equal
3.5 ml of 0.5 M sulphuric acid and 10 ml of water. Add 100 ml volumes of a saturated solution of picric acid and water until
of picric acid solution, heat on a water-bath for 15 minutes the washings are free from sulphate. Wash the residue with
and allow to stand for 1 hour. Filter through a sintered-glass five quantities, each of 10 ml, of ethanol and dry to constant
crucible (porosity No. 4) and wash the residue with successive weight at 105º.
quantities, each of 10 ml, of a mixture of equal volumes of a
1 g of the residue is equivalent to 0.3714 g of C4H10N2, H3PO4,
saturated solution of picric acid and water until the washings
H2O.
are free from sulphate. Wash the residue with five quantities,
each of 10 ml, of ethanol and dry to constant weight at 105º. Storage. Store protected from moisture.
1 g of the residue is equivalent to 0.3382 g of C4H10N2, H3PO4. Labelling. The label states, where applicable, that the tablets
are to be chewed before swallowing.
Storage. Store protected from moisture.

Piperazine Phosphate Tablets Piroxicam


If the tablets are intended to be chewed before swallowing
they may contain suitable flavouring agents. O O
S CH3
Piperazine Phosphate Tablets contain not less than 92.5 per N H
cent and not more than 107.5 per cent of the stated amount of N
piperazine phosphate, C4H10N2, H3PO4, H2O.
OH O N
Identification
Extract a quantity of the powdered tablets containing 1 g of C15H13N3O4S Mol. Wt. 331.4
Piperazine Phosphate with 20 ml of water and filter. The filtrate Piroxicam is 4-hydroxy-2-methyl-N-(2-pyridyl)-2H-1,2-
complies with the following tests. benzothiazine-3-carboxamide-1,1-dioxide.

951
PIROXICAM CAPSULES IP 2007

Piroxicam contains not less than 97.0 per cent and not more water and 5.35 g of sodium phosphate in 100 ml of water
than 103.0 per cent of C15H 13N 3O 4S, calculated on the to 1000 ml with water,
anhydrous basis. – flow rate. 1.2 ml per minute,
Description. An off-white to light tan or light yellow powder; – spectrophotometer set at 254 nm,
odourless. – a 20 µl loop injector.
Inject the reference solution. The test is not valid unless the
Identification relative standard deviation for replicate injections is not more
A. Determine by infrared absorption spectrophotometry (2.4.6). than 2.0 per cent.
Compare the spectrum with that obtained with piroxicam RS Inject alternately the test solution and the reference solution.
or with the reference spectrum of piroxicam.
Calculate the content of C15H13N3O4S.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Storage. Store protected from light and moisture.
0.001 per cent w/v solution in 0.01 M methanolic hydrochloric
acid shows absorption maxima at about 242 nm and 334 nm
and a minimum at about 270 nm; absorbance at about 334 nm,
about 0.87.
Piroxicam Capsules
C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254. Piroxicam Capsules contain not less than 92.5 per cent and
not more than 107.5 per cent of the stated amount of piroxicam,
Mobile phase. A mixture of 95 volumes of toluene and
C15H13N3O4S.
5 volumes of acetic acid.
Test solution. Dissolve 0.1 g of the substance under Identification
examination in 100 ml of a mixture of equal volumes of
Determine by thin-layer chromatography (2.4.17), coating the
chloroform and methanol.
plate with silica gel GF254.
Reference solution. A 0.1 per cent w/v solution of piroxicam
RS in a mixture of equal volumes of chloroform and methanol. Mobile phase. A mixture of 95 volumes of toluene and
5 volumes of acetic acid.
Apply to the plate 20 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. Test solution. Dissolve a portion of the contents of the
The principal spot in the chromatogram obtained with the test capsules in sufficient of a mixture of equal volumes of
solution corresponds to that in the chromatogram obtained chloroform and methanol to obtain a solution containing
with the reference solution. about 0.1 per cent w/v of Piroxicam. Shake for 10 minutes, filter
and use the filtrate.
Tests Reference solution. A 0.1 per cent w/v solution of piroxicam
Heavy metals (2.3.13). 0.4 g complies with the limit test for RS in a mixture of equal volumes of chloroform and methanol.
heavy metals, Method B (50 ppm). Apply to the plate 20 µl of each solution. After development,
Sulphated ash (2.3.18). Not more than 0.3 per cent. dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with the test
Water (2.3.43). Not more than 0.5 per cent , determined on solution corresponds to that in the chromatogram obtained
2.0 g. with the reference solution.
Assay. Determine by liquid chromatography (2.4.14).
Tests
Test solution. A 0,005 per cent w/v solution of the substance
under examination in 0.1 M methanolic hydrochloric acid. Dissolution (2.5.2).
Reference solution. A 0.005 per cent w/v solution of piroxicam Apparatus No. 2
RS in 0.1 M methanolic hydrochloric acid. Medium. 900 ml of 0.1 M hydrochloric acid
Speed and time. 100 rpm and 45 minutes.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Withdraw 10 ml of the medium and filter. Measure the
octadecylsilane bonded to porous silica (5 µm), absorbance of the filtrate (2.4.7), suitably diluted if necessary,
– mobile phase: a mixture of 45 volumes of methanol and at the maximum at about 242 nm. Calculate the content of
55 volumes of a buffer solution prepared by diluting a C15H13N3O4S, in the medium taking 352 as the specific
mixture of 7.72 g of anhydrous citric acid in 400 ml of absorbance at 242 nm.

952
IP 2007 POLYETHYLENE GLYCOL 1500

D. Not less than 75 per cent of the stated amount of is substantially converted into the hemihydrate, CaSO4,½H2O,
C15H13N3O4S. with minimum production of the anhydrous phases of calcium
Uniformity of content. (For capsules containing 10 mg or sulphate. It may contain suitable accelerators or decelerators.
less) — Comply with the test stated under Capsules. Description. A white or almost white powder; odourless or
almost odourless; hygroscopic.
Test solution. Dissolve the contents of a capsule in 100.0 ml
of 0.1 M methanolic hydrochloric acid and filter. Dilute further Identification
if necessary.
Gives the reactions of calcium salts and of sulphates (2.3.1).
Determine by liquid chromatography (2.4.14) using the
chromatographic system and the reference solution described Tests
under Assay.
pH (2.4.24). 6.5 to 9.0, determined in a 20.0 per cent w/v slurry
Calculate the content of C15H13N3O4S in the capsule. in water.
Water (2.3.43). Not more than 8.0 per cent, determined on Acid insoluble matter. Dissolve 0.5 g in 30 ml of a mixture of
0.25 g. 1 volume of hydrochloric acid and 2 volumes of water and
Other tests. Comply with the tests stated under Capsules. evaporate to dryness in a dish on a water-bath. Heat for
Assay. Determine by liquid chromatography (2.4.14). 2 hours at 120º and again add 20 ml of the acid mixture. Warm
for a few minutes and filter. Wash the residue with warm water
Test solution. Dissolve an accurately weighed quantity of the until free from chlorides, dry, ignite and weigh. The residue
mixed contents of the capsules containing about 50 mg of weighs not more than 5 mg (1.0 per cent).
Piroxicam in 100.0 ml of 0.1 M methanolic hydrochloric acid.
Further dilute 1.0 ml of the solution to 10.0 ml with the same Setting properties. 20 g mixed with 10 ml of water at 15º to 20º
solvent. in a cylindrical mould about 2.4 cm in diameter sets in not less
than 4 minutes and not more than 6 minutes. The mass thus
Reference solution. A 0.005 per cent w/v solution of piroxicam formed, after standing for 3 hours, possesses sufficient
RS in 0.1 M methanolic hydrochloric acid. hardness to resist pressure of the fingers at the edges, which
Chromatographic system retain their sharpness of outline and do not crumble under
– a stainless steel column 25 cm x 4.6 mm, packed with pressure.
octadecylsilane bonded to porous silica (5 µm), Loss on ignition (2.4.20). 4.5 to 8.0 per cent, determined by
– mobile phase: a mixture of 45 volumes of methanol and igniting to constant weight at red heat.
55 volumes of a buffer solution prepared by diluting a
mixture of 7.72 g of anhydrous citric acid in 400 ml of Storage. Store protected from moisture
water and 5.35 g of sodium phosphate in 100 ml of water
to 1000 ml with water,
– flow rate. 1.2 ml per minute, Polyethylene Glycol 1500
– spectrophotometer set at 254 nm, Macrogol 1500
– a 20 µl loop injector.
Polyethylene Glycol 1500 is a mixture of the polycondensation
Inject the reference solution. The test is not valid unless the
products of ethylene oxide and water obtained under
relative standard deviation for replicate injections is not more
controlled conditions. It is represented by the formula
than 2.0 per cent.
HOCH2[CH2OCH2]nCH2OH, where n is between 28 and 36.
Inject alternately the test solution and the reference solution. Description. A white or creamy white, soft, wax-like solid;
Calculate the content of C15H13N3O4S in the capsules. odour, faint and characteristic.
Storage. Store protected from light and moisture. Tests
Appearance of solution (2.4.1). A 25.0 per cent w/v solution is
not more intensely coloured than reference solution BYS6.
Plaster Of Paris pH (2.4.24). 4.0 to 7.0, determined in a 5.0 per cent w/v solution.
Dried Calcium Sulphate; Exsiccated Calcium Sulphate Freezing point (2.4.11). 42º to 46º.
CaSO4, ½ H2O Mol. Wt. 145.2 Hydroxyl value (2.3.27). 70 to 86, determined on 5.0 g.
Plaster of Paris is prepared by heating powdered gypsum, Viscosity (2.4.28). 25 mm2s–1 to 32 mm2s–1, determined at 100º
CaSO4,½H2O, at about 150º in a controlled manner such that it by Method A using a U-tube viscometer (size D).

953
POLYETHYLENE GLYCOL 4000 IP 2007

Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium Polyethylene Glycol 6000
carbonate, add 10 ml of bromine solution and mix thoroughly.
Evaporate to dryness on a water-bath, gently ignite and Macrogol 6000
dissolve the cooled residue in 16 ml of brominated Polyethylene Glycol 6000 is a mixture of the polycondensation
hydrochloric acid and 45 ml of water. Remove the excess of products of ethylene oxide and water obtained under
bromine with 2 ml of stannous chloride solution AsT. The controlled conditions. It is represented by the formula
resulting solution complies with the limit test for arsenic HOCH2[CH2OCH2]nCH2OH, where n is between 112 and 158.
(3 ppm).
Description. A white to creamy white, wax-like solid or flakes;
Heavy metals (2.3.13). Dissolve 4.0 g in 5 ml of a 1.0 per cent odour, faint and characteristic.
w/v solution of hydrochloric acid and sufficient water to
produce 25 ml. The solution complies with the limit test for Tests
heavy metals, Method A (5 ppm).
Appearance of solution (2.4.1). A 15.0 per cent w/v solution is
Sulphated ash (2.3.18). Not more than 0.1 per cent.
not more intensely coloured than reference solution BYS6.
Storage. Store protected from moisture.
pH (2.4.24). 4.5 to 7.5, determined in a 5.0 per cent w/v solution.
Freezing point (2.4.11). 56º to 60º.
Polyethylene Glycol 4000 Viscosity (2.4.28). 250 mm2s–1 to 390 mm2s–1, determined at
100º by Method A using a U-tube viscometer (size F).
Macrogol 4000
Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium
Polyethylene Glycol 4000 is a mixture of the polycondensation carbonate, add 10 ml of bromine solution and mix thoroughly.
products of ethylene oxide and water obtained under Evaporate to dryness on a water-bath, gently ignite and
controlled conditions. It is represented by the formula dissolve the cooled residue in 16 ml of brominated
HOCH2[CH2OCH2]nCH2OH, where n is between 69 and 84. hydrochloric acid and 45 ml of water. Remove the excess of
Description. A creamy white, hard, wax-like solid, powder or bromine with 2 ml of stannous chloride solution AsT. The
flakes; odour, faint and characteristic. resulting solution complies with the limit test for arsenic
(3 ppm).
Tests
Heavy metals (2.3.13). Dissolve 4.0 g in 5 ml of a 1.0 per cent
Appearance of solution (2.4.1). A 20.0 per cent w/v solution is w/v solution of hydrochloric acid and sufficient water to
not more intensely coloured than reference solution BYS6. produce 25 ml. The resulting solution complies with the limit
pH (2.4.24). 4.5 to 7.5, determined in a 5.0 per cent w/v solution. test for heavy metals, Method A (5 ppm).

Freezing point (2.4.11). 53º to 56º. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Hydroxyl value (2.3.27). 30 to 36, determined on 20.0 g. Storage. Store protected from moisture.
Viscosity (2.4.28). 76 mm2s–1 to 110 mm2s–1, determined at 100º
by Method A using a U-tube viscometer (size E).
Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium Polysorbate 20
carbonate, add 10 ml of bromine solution and mix thoroughly.
Evaporate to dryness on a water-bath, gently ignite and Polyoxyethylene 20 Sorbitan Monolaurate
dissolve the cooled residue in 16 ml of brominated
hydrochloric acid and 45 ml of water. Remove the excess of HO(H2CH2CO)w (OCH2CH2)xOH
bromine with 2 ml of stannous chloride solution AsT. The
(OCH2 CH2)yOH
resulting solution complies with the limit test for arsenic O
O
(3 ppm).
(OCH2CH2)z O C11H23
Heavy metals (2.3.13). Dissolve 4.0 g in 5 ml of a 1.0 per cent
w/v solution of hydrochloric acid and sufficient water to w + x + y + z = 20
produce 25 ml. The solution complies with the limit test for
heavy metals, Method A (5 ppm). Polyoxyethylene 20 Sorbitan Monolaurate
Sulphated ash (2.3.18). Not more than 0.1 per cent. Polysorbate 20 is a mixture of partial lauric esters of D-glucitol
Storage. Store protected from moisture. and its anhydrides copolymerised with approximately 20 moles

954
IP 2007 POLYSORBATE 80

of ethylene oxide for each mole of D-glucitol and its anhydrides. Polysorbate 80
The lauric acid used for the esterification may contain other
fatty acids. Polyoxyethylene 80 Sorbitan Monooleate
Description. A clear or slightly opalescent, oily, yellowish or
brownish yellow liquid; odour, faint and characteristic. HO(H2CH2CO)w (OCH2CH2)xOH

Identification (OCH2CH2)yOH
O O
A. Dissolve 0.5 g in water at about 50º and dilute to 10 ml with
the same solvent; the solution produces a copious foam on (OCH2CH2)z O C17H33
shaking. Add 0.5 g of sodium chloride and heat to boiling; the
resulting cloudiness disappears during cooling to about 50º. w+x+y+z = 20

B. Dissolve 0.1 g in 5 ml of chloroform, add 0.1 g of potassium Polysorbate 80 is a mixture of partial oleic esters of D-glucitol
thiocyanate and 0.1 g of cobalt nitrate and stir with a glass and its anhydrides copolymerised with approximately 20 moles
rod; a blue colour is produced. of ethylene oxide for each mole of D-glucitol and its anhydrides.

Tests Description. A clear or almost clear, oily, yellowish or brownish


yellow liquid; odour, faint and characteristic.
pH (2.4.24). 5.0 to 7.0, determined in a 5.0 per cent w/v solution
in carbon dioxide-free water. Identification
Acid value (2.3.23). Not more than 2.0, determined on 5.0 g. A. Dissolve 0.5 g in water at about 50º and dilute to 10 ml with
Hydroxyl value (2.3.27). 96 to 108, determined on 2.0 g. the same solvent; the solution produces a copious foam on
shaking. Add 0.5 g of sodium chloride and heat to boiling; the
Saponification value (2.3.37). 40 to 50, using 15 ml of 0.5 M resulting cloudiness disappears during cooling to about 50º.
ethanolic potassium hydroxide and diluting with 50 ml of
water before carrying out the titration. B. Dissolve 0.1 g in 5 ml of chloroform, add 0.1 g of potassium
thiocyanate and 0.1 g of cobalt nitrate and stir with a glass
Iodine value (2.3.28). Not more than 5.0, determined by the rod; a blue colour is produced.
iodine bromide method.
C. To 2 ml of a 5 per cent w/v solution add 0.5 ml of bromine
Heavy metals (2.3.13). 2.0 g complies with the limit test for water; the bromine is decolourised.
heavy metals, Method B (10 ppm).
Reducing impurities. Dissolve 2.0 g in 25 ml of hot water, add Tests
25 ml of 1 M sulphuric acid and 0.1 ml of ferroin solution and pH (2.4.24). 6.0 to 8.0, determined in a 5.0 per cent w/v solution
titrate with 0.01 M ceric ammonium sulphate shaking in carbon dioxide-free water.
continuously until the colour changes from red to greenish
blue persists for 30 seconds. Carry out a blank titration. Not Acid value (2.3.23). Not more than 2.0, determined on 5 g.
more than 2.0 ml of 0.01 M ceric ammonium sulphate is Hydroxyl value (2.3.27). 65 to 80, determined on 2.0 g.
required. Saponification value (2.3.37). 45 to 55, using 15 ml of 0.5 M
Sulphated ash. Not more than 0.2 per cent, determined by the ethanolic potassium hydroxide and diluting with 50 ml of
following method. Weigh accurately about 2.0 g in a silica or water before carrying out the titration.
platinum crucible, add 0.5 ml of sulphuric acid and heat on a Iodine value (2.3.28). 18 to 24, determined by the iodine bromide
water-bath for 2 hours. Carefully ignite at a low temperature method.
until thoroughly charred. Add to the carbonised mass 2 ml of
nitric acid and 0.25 ml of sulphuric acid, cautiously heat until Reducing impurities. Dissolve 2.0 g in 25 ml of hot water, add
white fumes are evolved and then ignite at 600º until the carbon 25 ml of 1 M sulphuric acid and 0.1 ml of ferroin solution and
is completely burnt off. Allow to cool, weigh and repeat the titrate with 0.01 M ceric ammonium sulphate shaking
operation for periods of 15 minutes until two successive continuously until the colour changes from red to greenish
weighings do not differ by more than 0.5 mg. blue persists for 30 seconds. Carry out a blank titration. Not
more than 5.0 ml of 0.01 M ceric ammonium sulphate is
Water (2.3.43). Not more than 3.0 per cent , determined on required.
1.0 g.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
Storage. Store protected from light and moisture. heavy metals, Method B (10 ppm).

955
POTASSIUM CHLORIDE IP 2007

Sulphated ash. Not more than 0.2 per cent, determined by the Iron (2.3.14). 20 ml of solution A complies with the limit test for
following method. Weigh accurately about 2.0 g in a silica or iron (20 ppm).
platinum crucible, add 0.5 ml of sulphuric acid and heat on a
Bromides. Dilute 1.0 ml of solution A to 50.0 ml with water. To
water-bath for 2 hours. Carefully ignite at a low temperature
5.0 ml of the solution add 2.0 ml of phenol red reagent and
until thoroughly charred. Add to the carbonised mass 2 ml of
1.0 ml of a 0.01 per cent solution of chloramine T and mix
nitric acid and 0.25 ml of sulphuric acid, cautiously heat until
immediately. After exactly 2 minutes, add 0.15 ml of 0.1 M
white fumes are evolved and then ignite at 600º until the carbon
sodium thiosulphate, mix and dilute to 10.0 ml with water. The
is completely burnt off. Allow to cool, weigh and repeat the
absorbance of the solution measured at about 590 nm (2.4.7),
operation for periods of 15 minutes until two successive
using water as the blank, is not more than that of a standard
weighings do not differ by more than 0.5 mg.
prepared at the same time and in the same manner, using 5.0 ml
Water (2.3.43). Not more than 3.0 per cent, determined on 1.0 g. of a 0.0003 per cent w/v solution of potassium bromide
Storage. Store protected from light and moisture. (0.1 per cent).
Iodides. Moisten 5 g by adding dropwise, a solution freshly
prepared by mixing 25 ml of iodide-free starch solution, 2 ml
Potassium Chloride of 0.5 M sulphuric acid, 0.15 ml of sodium nitrite solution
and 25 ml of water and examine the mixture in daylight; the
KCl Mol. Wt. 74.6
substance shows no blue colour after 5 minutes.
Potassium Chloride contains not less than 99.0 per cent and
Sulphates (2.3.17). 0.5 g complies with the limit test for
not more than 100.5 per cent of KCl, calculated on the dried
sulphates (300 ppm).
basis.
Description. Colourless crystals or a white, crystalline powder; Loss on drying (2.4.19). Not more than 1.0 per cent, determined
odourless. on 1.0 g by drying in an oven at 105º.
Assay. Weigh accurately about 0.15 g, dissolve in 50 ml of
Identification water and titrate with 0.1 M silver nitrate using potassium
Dissolve 10 g in 100 ml of carbon dioxide-free water prepared chromate solution as indicator.
from distilled water (solution A). The solution gives the 1 ml of 0.1 M silver nitrate is equivalent to 0.007455 g of KCl.
reactions of potassium salts and of chlorides (2.3.1).
Potassium Chloride intended for use in the manufacture of
Tests dialysis solutions complies with the following additional
requirement.
Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1). Aluminium. Dissolve 4.0 g in 100 ml of water and add 10 ml of
acetate buffer pH 6.0. Extract with successive quantities of
Acidity or alkalinity. 5.0 g dissolved in 50 ml of carbon dioxide-
20, 20 and 10 ml of a 0.5 per cent w/v solution of
free water requires not more than 0.5 ml of 0.01 M sodium
8-hydroxyquinoline in chloroform and dilute the combined
hydroxide or 0.01 M hydrochloric acid for neutralisation to
extracts to 50 ml with chloroform. Use as the standard solution
bromothymol blue solution.
a mixture of 2 ml of aluminium standard solution (2 ppm Al),
Arsenic (2.3.10). Dissolve 10.0 g in 50 ml of water and add 10 ml of acetate buffer pH 6.0 and 98 ml of water treated in the
10 ml of stannated hydrochloric acid AsT. The resulting same manner and as the blank solution a mixture of 10 ml of
solution complies with the limit test for arsenic (1 ppm). acetate buffer pH 6.0 and 100 ml of water treated in the same
Barium. To 5 ml of solution A add 5 ml of water and 1 ml of manner. Measure the fluorescence of the test and standard
1 M sulphuric acid; the solution, after not less than 15 minutes, solutions (2.4.5), using an excitation wavelength of about
is not more opalescent than a mixture of 5 ml of solution A and 392 nm and emission wavelength of about 518 nm and setting
6 ml of water. the instrument to zero with the blank solution in each case.
The fluorescence of the test is not greater than that of the
Heavy metals (2.3.13). Dissolve 2.0 g in 10 ml of water to standard solution (1 ppm).
which are added 2 ml of dilute acetic acid and 13 ml of water.
The solution complies with the limit test for heavy metals, Potassium chloride intended for use in the manufacture of
Method A (10 ppm). Parenteral Preparations or for the preparation of haemodialysis
solution complies with the following additional requirement.
Calcium and magnesium. Dissolve 1 g in 20 ml of water, add
1 ml of dilute ammonia solution and 1 ml of sodium phosphate Sodium. Not more than 0.1 per cent, determined by atomic
solution; the solution remains clear. absorption spectrophotometry (2.4.2), using a 1.0 per cent

956
IP 2007 POTASSIUM CHLORIDE, SODIUM CHLORIDE AND DEXTROSE INJECTION

w/v solution and measuring at 589 nm. Use sodium solution 0.1 M silver nitrate using potassium chromate solution as
AAS, suitably diluted with water, for the standard solutions. indicator.
Storage. Store protected from moisture. 1 ml of 0.1 M silver nitrate is equivalent to 0.007455 g of KCl.
Labelling. The label states whether or not the material is For dextrose — To an accurately measured volume
suitable for use in the manufacture of Parenteral Preparations containing 2 g to 5 g of Dextrose, add 0.2 ml of 05 M ammonia
or for the preparation of haemodialysis or dialysis solutions. and sufficient water to produce 100.0 ml. Mix well, allow to
stand for 30 minutes and measure the optical rotation in a
2-dm tube (2.4.22). The observed rotation in degrees multiplied
by 0.9477 represents the weight, in g, of dextrose, C6H12O6, in
Potassium Chloride And Dextrose the volume taken for assay.
Injection Storage. Store in single dose containers.
Potassium Chloride in Dextrose Injection; Potassium Chloride Labelling. The label states (1) the strength as the percentages
and Dextrose Intravenous Infusion; Potassium Chloride and w/v of Potassium Chloride and Dextrose; (2) the total osmolar
Glucose Intravenous Infusion concentration in mOsmol per litre; (3) where the contents are
Potassium Chloride and Dextrose Injection is a sterile solution less than 100 ml, or where the label states that the Injection is
of Potassium Chloride and Dextrose in Water for Injections. not for direct use but is to be diluted before use, the label
alternatively may state the total osmolar concentration in
Potassium Chloride and Dextrose Injection contains not less mOsmol per ml; (4) the content of potassium in millimoles; (5)
than 95.0 per cent and not more than 110.0 per cent of the that the injection containing visible particles in the solution
stated amount of potassium chloride, KCl, and not less than should not be used.
95.0 per cent and not more than 105.0 per cent of the stated
amount of dextrose, C6H12O6. It contains no antimicrobial agent.
Description. A clear, colourless or faintly straw-coloured Potassium Chloride, Sodium Chloride
solution.
And Dextrose Injection
Identification Potassium Chloride in Dextrose and Sodium Chloride
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution; Injection; Potassium Chloride, Sodium Chloride and
the solution remains blue and clear. Heat to boiling, a copious Dextrose Intravenous Infusion; Potassium Chloride,
red precipitate is formed. Sodium Chloride and Glucose Intravenous Infusion
B. Gives reaction B of potassium salts and reaction A of Potassium Chloride, Sodium Chloride and Dextrose Injection
chlorides (2.3.1). is a sterile solution of Potassium Chloride, Sodium Chloride
and Dextrose in Water for Injections.
Tests
Potassium Chloride, Sodium Chloride and Dextrose Injection
pH (2.4.24). 3.5 to 6.5. contains not less than 95.0 per cent and not more than
5-Hydroxymethylfurfural and Related substances. Dilute a 110.0 per cent of the stated amounts of sodium, Na, potassium,
volume containing 1.0 g of Dextrose to 500.0 ml with water K, and chloride, Cl, and not less than 95.0 per cent and not
and measure the absorbance of the resulting solution at the more than 105.0 per cent of the stated amount of dextrose,
maximum at about 284 nm, absorbance at about 284 nm (2.4.7); C6H12O6. It contains no antimicrobial agent.
not more than 0.25.
Identification
Heavy metals (2.3.13). Evaporate a volume containing 4.0 g of
Dextrose to 10 ml and add 2 ml of dilute acetic acid and A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution;
sufficient water to produce 25 ml. The solution complies with the solution remains blue and clear. Heat to boiling, a copious
the limit test for heavy metals, Method A (5 ppm). red precipitate is formed.

Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit B. Gives reaction A of potassium salts, reaction B of sodium
per ml. salts and reaction A of chlorides (2.3.1)

Other tests. Complies with the tests stated under Parenteral Tests
Preparations (Injections). pH (2.4.24). 3.5 to 6.5, determined on a portion diluted with
Assay. For potassium chloride — Titrate an accurately water, if necessary, to a concentration of not more than 5.0 per
measured volume containing 0.1 g of Potassium Chloride with cent of dextrose.

957
POTASSIUM CITRATE IP 2007

5-Hydroxymethylfurfural and Related substances. Dilute a sodium and chloride in millimoles; (5) that the injection
volume containing 1.0 g of Dextrose to 500.0 ml with water containing visible particles in the solution should not be used.
and measure the absorbance of the resulting solution at the
maximum at about 284 nm, absorbance at about 284 nm (2.4.7);
not more than 0.25.
Potassium Citrate
Heavy metals (2.3.13). Evaporate a volume containing 4.0 g of
Dextrose to 10 ml and add 2 ml of dilute acetic acid and
sufficient water to produce 25 ml. The solution complies with HO COOK
,H2O
the limit test for heavy metals, Method A (5 ppm). KOOC COOK
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
C6H5K3O7,H2O Mol. Wt. 324.4
per ml.
Potassium citrate is tripotassium 2-hydroxypropane-1,2,3-
Other tests. Complies with the tests stated under Parenteral
tricarboxylate monohydrate.
Preparations (Injections).
Potassium Citrate contains not less than 99.0 per cent and not
Assay. For sodium — Dilute appropriately with water and
more than 101.0 per cent of C6H5K3O7, calculated on the
determine by Method A for flame photometry (2.4.4), or by
anhydrous basis.
Method A for atomic absorption spectrophotometry (2.4.2),
measuring at 589 nm and using sodium solution FP or sodium Description. White, granular crystals or a crystalline powder;
solution AAS respectively, suitably diluted with water for the odourless; hygroscopic.
standard solutions.
Identification
For potassium — Dilute appropriately with water and
determine by Method A for flame photometry (2.4.4), or by A. Dissolve 10 g in 100 ml of carbon dioxide-free water
Method A for atomic absorption spectrophotometry (2.4.2), prepared from distilled water (solution A). 0.5 ml of solution
measuring at 767 nm and using potassium solution FP or A gives the reactions of potassium salts (2.3.1).
potassium solution AAS respectively, suitably diluted with B. To 1 ml of solution A add 4 ml of water; the solution gives
water for the standard solutions. the reactions of citrates (2.3.1).
For total chlorides — To 20.0 ml add 30 ml of water, 50.0 ml of
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the Tests
precipitate with water slightly acidified with nitric acid and Appearance of solution. Solution A is clear (2.4.1), and
titrate the excess of silver nitrate with 0.1 M ammonium colourless (2.4.1).
thiocyanate using ferric ammonium sulphate solution as
indicator until a reddish yellow colour is produced. Carry out Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of
a blank titration. dilute phenolphthalein solution; not more than 0.2 ml of
0.1 M hydrochloric acid or 0.1 M sodium hydroxide is
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total required to change the colour of the solution.
chloride, calculated as Cl.
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 15 ml
For dextrose — To an accurately measured volume containing
of stannated hydrochloric acid. The resulting solution
2 g to 5 g of Dextrose, add 0.2 ml of 5 M ammonia and sufficient
complies with the limit test for arsenic (2 ppm).
water to produce 100.0 ml. Mix well, allow to stand for
30 minutes and measure the optical rotation in a 2-dm tube Heavy metals (2.3.13). 2.0 g complies with the limit test for
(2.4.22). The observed rotation in degrees multiplied by 0.9477 heavy metals, Method A (10 ppm).
represents the weight, in g, of dextrose, C6H12O6, in the volume Sodium. Not more than 0.3 per cent, determined by atomic
taken for assay. absorption spectrophotometry (2.4.2), Method I I, measuring
Storage. Store in single dose containers. at 589 nm. Prepare the test solution by addition of 1 ml of 2 M
hydrochloric acid to 10.0 ml of solution A and diluting to
Labelling. The label states (1) the strength as the percentages
100.0 ml with distilled water. Use sodium solution AAS,
w/v of Potassium Chloride, Sodium Chloride and Dextrose; (2)
suitably diluted with distilled water, for the standard
the total osmolar concentration in mOsmol per litre; (3) where
solutions.
the contents are less than 100 ml, or where the label states that
the Injection is not for direct use but is to be diluted before Chlorides (2.3.12). Dilute 25.0 ml of solution A to 35 ml with
use, the label alternatively may state the total osmolar water. The resulting solution complies with the limit test for
concentration in mOsmol per ml; (4) the content of potassium, chlorides (100 ppm).

958
IP 2007 POTASSIUM CLAVULANATE

Oxalate. Dissolve 0.5 g in 4 ml of water, add 3 ml of hydrochloric Identification


acid and 1 g of granulated zinc and heat on a water-bath for
1 minute. Allow to stand for 2 minutes, decant into 0.25 ml of a A. In the Assay, the principal peak in the chromatogram
1 per cent w/v solution of phenylhydrazine hydrochloride, obtained with the test solution corresponds to the peak in the
heat to boiling, cool rapidly, add an equal volume of chromatogram obtained with reference solution (a).
hydrochloric acid and 0.25 ml of potassium ferricyanide B. Gives reaction A of potassium salts (2.3.1).
solution, shake and allow to stand for 30 minutes. Any pink
colour produced is not more intense than that obtained by Tests
treating 4.0 ml of a 0.005 per cent w/v solution of oxalic acid
pH (2.4.24). 5.5 to 8.0, determined in 1.0 per cent w/v solution.
at the same time and in the same manner (300 ppm, calculated
as anhydrous oxalic acid). Related substances. Determine by liquid chromatography
(2.4.14).
Sulphates (2.3.17). To 10.0 ml of solution A add 2 ml of 7 M
hydrochloric acid and dilute to 15 ml with distilled water; the NOTE – Prepare the solutions immediately before use.
solution complies with the limit test for sulphates (150 ppm). Test solution Dissolve 0.25 g of the substance under
Readily carbonisable substances. Heat 0.20 g, in powder, with examination in mobile phase A and dilute to 25 ml with mobile
10 ml of sulphuric acid (96 per cent w/w) in a water-bath at phase A.
90º ± 1º for 60 minutes and cool rapidly. The solution is not Reference solution (a). Dilute 1 ml of the test solution to 100
more intensely coloured than reference solution YS2 or GYS2 ml with mobile phase A.
(2.4.1).
Reference solution (b). A solution containing 0.01 per cent w/
Water (2.3.43). 4.0 to 7.0 per cent, determined on 0.5 g, titration v each of lithium clavulanate RS and amoxycillin trihydrate
being done after stirring for 15 minutes subsequent to addition RS in mobile phase A.
of the substance under examination.
Chromatographic system
Assay. Weigh accurately about 0.2 g, dissolve in 20 ml of
anhydrous glacial acetic acid, heat to about 50º, allow to – a stainless steel column 10 cm x 4.6 mm, packed with
cool. Titrate with 0.1 M perchloric acid, using 0.25 ml of octadecylsilyl silica gel (5 µm),
1-naphtholbenzein solution as indicator and titrating until a – column temperature 40º,
green colour is obtained. Carry out a blank titration. – mobile phase: A. a 0.78 per cent w/v solution of sodium
dihydrogen phosphate adjusted to pH 4.0 with
1 ml of 0.1 M perchloric acid is equivalent to 0.01021 g of phosphoric acid and filtered through a 0.5 µm filter,
C6H5K3O7. B. a mixture of equal volumes of mobile
Storage. Store protected from moisture. phase A and methanol,
– a linear gradient programme using the conditions given
below,
Potassium Clavulanate – flow rate. 1 ml per minute,
– spectrophotometer set at 230 nm,
O – a 20 µl loop injector.
OK
O H Time Mobile phase A Mobile phase B
N (min) (per cent v/v) (per cent v/v)
O 0–4 100 0
OH
H 4 – 15 100 → 50 0 50
C8H8KNO5 Mol. Wt. 237.3 15 – 18 50 50
18 – 24 50 →100 50 ◊ 0
Potassium Clavulanate is potassium (Z)-(2R,5R)-3-(2-
hydroxyethylidene)-7-oxo-4-oxa-1-azabicyclo[3.2.0]heptane- 24 – 39 100 0
2-carboxylate. Inject reference solution (b). The resolution between
Potassium Clavulanate contains not less than 96.5 per cent clavulanate (first peak) and amoxycillin (second peak ) is not
and not more than 102.0 per cent of potassium clavulanate, less than 13.
C8H8KNO5, calculated on the anhydrous basis. Inject the test solution and reference solution (b). The area of
Description. A white to off white, crystalline hygroscopic any individual impurity peak obtained is not more than the
powder. area of the principal peak in the chromatogram obtained with

959
POTASSIUM CLAVULANATE DILUTED IP 2007

reference solution (a) (1.0 per cent). The sum of all impurity Sterility (2.2.11). Complies with the test for sterility.
peaks obtained is not more than twice the area of the principal Storage. Store protected from moisture.
peak in the chromatogram obtained with reference solution
(a) (2.0 per cent). Ignore any peaks with an area 0.05 times the Labelling. The label states whether it is intended for use in
area of the principal peak in the chromatogram obtained with the manufacture of parenteral preparations.
reference solution (a) (0.05 per cent).
Water (2.3.43). Not more than 1.5 per cent, determined on 0.5 g.
Assay. Determine by liquid chromatography (2.4.14). Potassium Clavulanate Diluted
NOTE – Prepare the solutions immediately before use.
C8H8KNO5 Mol. Wt. 237.3
Test solution. Dissolve 50.0 mg of the substance under
Potassium Clavulanate Diluted is a dry mixture of Potassium
examination in a 0.41 per cent w/v solution of sodium acetate
Clavulanate and Microcrystalline Cellulose, or Silica, colloidal
previously adjusted to pH 6.0 with glacial acetic acid, and
anhydrous or Silica, colloidal hydrated.
dilute to 50.0 ml with the same solution.
Potassium Clavulanate Diluted contain not less than 91.2 per
Reference solution (a). A 0.1 per cent w/v solution of lithium
cent and not more than 107.1 per cent of the stated amount of
clavulanate RS in a 0.41 per cent w/v solution of sodium
potassium clavulanate, C8H8KNO5.
acetate previously adjusted to pH 6.0 with glacial acetic
acid. Description. A white or almost white powder, hygroscopic.
Reference solution (b). A solution containing 0.1 per cent w/ Identification
v each of lithium clavulanate RS and amoxycillin trihydrate
RS in a 0.41 per cent w/v solution of sodium acetate previously A. In the Assay, the principal peak in the chromatogram
adjusted to pH 6.0 with glacial acetic acid. obtained with the test solution corresponds to the peak in the
Chromatographic system chromatogram obtained with reference solution (a).
– a stainless steel column 30 cm x 4.6 mm, packed with B. Gives reaction A of potassium salts (2.3.1).
octadecylsilyl silica gel (5 µm),
C. Depending on the diluent used, carry out one of the relevant
– mobile phase: a mixture of 5 volumes of methanol and
identification tests given below.
95 volumes of a 1.5 per cent w/v solution of sodium
dihydrogen phosphate previously adjusted to pH 4.0 (i) On a watch-glass place a quantity of the substance under
with dilute phosphoric acid, examination corresponding to 20 mg of cellulose and disperse
– flow rate. 1 ml per minute, in 4 ml of iodinated zinc chloride solution. The powder
– spectrophotometer set at 230 nm, becomes violet-blue in colour.
– a 10 µl loop injector. (ii) It gives the reaction of silicates (2.3.1).
Inject reference solution (b). The resolution between
clavulanate (first peak) and amoxycillin (second peak) is not Tests
less than 3.5. pH (2.4.24) 4.8 to 8.0, determined on a quantity of the substance
Inject alternately the test solution and reference solution (a). under examination
Calculate the content of C8H8KNO5. containing 0.2 g of potassium clavulanate dissolved in 20 ml
1 mg of clavulanate (C8H9NO5) is equivalent to 1.191 mg of of carbon dioxide-free water.
C8H8KNO5. Light absorption. When examined in the range 230 nm to 360
Potassium Clavulanate intended for use in the manufacture nm (2.4.7), a 0.1 per cent w/v solution in 0.1 M phosphate
of Parenteral Preparations without a further procedure for buffer solution pH 7.0 shows an absorption maximum only at
the removal of bacterial endotoxins complies with the about 278 nm; absorbance at about 278 nm, not more than
following additional requirement. 0.40.
Bacterial endotoxins (2.2.3). Not more than 0.03 Endotoxin Related substances. Determine by liquid chromatography
Unit per mg of potassium clavulanate. (2.4.14).
Potassium Clavulanate intended for use in the manufacture Test solution. Dissolve a quantity of the substance under
of Parenteral Preparations without a further sterilisation examination containing 0.25 g of potassium clavulanate in 5
procedure complies with the following additional ml of mobile phase A, dilute to 25.0 ml with mobile phase A and
requirement. filter.

960
IP 2007 POTASSIUM IODIDE

Reference solution (a). Dilute 1.0 ml of the test solution to Reference solution (b). Dissolve 10 mg of amoxycillin
100.0 ml with mobile phase A. trihydrate RS in 10 ml of reference solution (a).
Reference solution (b). Dissolve 10 mg of amoxycillin Chromatographic system
trihydrate RS in 1 ml of the test solution and dilute to 100 ml – a stainless steel column 03 cm × 4.6 mm, packed with
with mobile phase A. octadecylsilane bonded to porous silica (10 µm),
– mobile phase: a mixture of 5 volumes of methanol and
Chromatographic system
95 volumes of a 1.5
– a stainless steel column 25 cm x 4.6 mm, packed with
per cent w/v solution of sodium dihydrogen phosphate
octadecylsilyl silica gel (5 µm),
with the pH previously
– mobile phase: A. a 0.78 per cent w/v solution of sodium
adjusted to 4.0 with dilute phosphoric acid,
dihydrogen phosphate with the pH adjusted to 4.0 with
– flow rate. 1 ml per minute,
dilute phosphoric acid,
– spectrophotometer set at 230 nm,
B. a mixture of equal volumes of mobile
– a 20 µl loop injector.
phase A and methanol,
– flow rate. 1 ml per minute, Inject reference solution (b). The resolution between the
– a linear gradient programme using the conditions given clavulanate peak and amoxycillin peak is not less than 3.5.
below, Inject alternately the test solution and reference solution (a).
– spectrophotometer set at 230 nm,
– a 20 µl loop injector. Calculate the content of C8H8KNO5.
Time Mobile Mobile Comment 1 mg of clavulanate (C8H9NO5) is equivalent to 1.191 mg of
phase A phase B C8H8KNO5.
(min) (per cent v/v) (per cent v/v) Storage. Store protected from moisture.
0 100 0 Isocratic
Labelling. The label states the percentage contents of
4 100 0 begin linear potassium clavulanate and the diluent used to prepare the
gradient mixture.
15 50 50
18 50 50 end chromatogram,
return to 100A
Potassium Iodide
24 100 0 end equilibration,
begin next KI Mol. Wt. 166.0
chromatogram Potassium Iodide contains not less than 99.0 per cent and
Inject reference solution (b). The resolution between the not more than 100.5 per cent of KI, calculated on the dried
clavulanate (first peak) and amoxycillin (second peak) is not basis.
less than 13. Description. Colourless crystals or a white powder; odourless.
Inject alternatively the test solution and reference solution
(a). Any individual impurity is not more than 1.0 per cent and Identification
the sum of all impurities found is not more than 2.0 per cent.
Dissolve 10 g in 100 ml of carbon dioxide-free water (solution
Water (2.3.43). Not more than 2.5 per cent, determined on A). The solution gives the reactions of potassium salts, and
1.0 g. of iodides (2.3.1).
Assay. Determine by liquid chromatography (2.4.14).
Tests
Test solution. Dissolve a quantity of the substance under
examination containing about 50.0 mg of potassium Appearance of solution. Solution A is clear (2.4.1), and
clavulanate in a 0.4 per cent w/v solution of sodium acetate colourless (2.4.1).
with the pH previously adjusted to 6.0 with glacial acetic Alkalinity. To 10 ml of solution A add 0.2 ml of 0.01 M sulphuric
acid, dilute to 50.0 ml with the same solution and filter. acid; no colour is produced on addition of a drop of
Reference solution (a). Dissolve 50.0 mg of lithium phenolphthalein solution.
clavulanate RS in a 0.4 per cent w/v solution of sodium acetate Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and 12 ml of
with the pH previously adjusted to 6.0 with glacial acetic stannated hydrochloric acid AsT. The resulting solution
acid and dilute to 50.0 ml with the same solution. complies with the limit test for arsenic (2 ppm).

961
POTASSIUM PERMANGANATE IP 2007

Heavy metals (2.3.13). 2.0 g complies with the limit test for B. Heated to redness, it decrepitates, evolves oxygen and
heavy metals, Method A (10 ppm). leaves a black residue which with water forms potassium
Iron (2.3.14). Solution A complies with the limit test for iron hydroxide solution; the resulting solution when neutralised
(20 ppm). with dilute hydrochloric acid gives the reactions of potassium
salts (2.3.1).
Barium. Dissolve 0.5 g in 10 ml of water and add 1 ml of dilute
sulphuric acid; no turbidity develops within one minute. Tests
Cyanide. Warm 5 ml of Solution A, add one drop of ferrous Appearance of solution. Dissolve 1.5 g in 50 ml of distilled
sulphate solution and 0.5 ml of sodium hydroxide solution water, heat on a water-bath and add gradually 6 ml of ethanol
and acidify with hydrochloric acid; no blue colour is (95 per cent), cool, dilute to 60 ml with distilled water and
produced. filter. The filtrate (solution A) is colourless (2.4.1).
Iodate. Dissolve 0.5 g in 10 ml of carbon dioxide-free water Chlorides (2.3.12). 40 ml of solution A complies with the limit
and add 0.15 ml of dilute sulphuric acid and a drop of iodide- test for chlorides (250 ppm).
free starch solution; no blue colour is produced within
2 minutes. Sulphates (2.3.17). 10 ml of solution A complies with the limit
test for sulphates (600 ppm).
Sulphates (2.3.17). 1.0 g dissolved in 15 ml of water complies
with the limit test for sulphates (150 ppm). Water-insoluble matter. Dissolve 0.5 g in 50 ml of water, heat
to boiling, filter through a tared, sintered-glass filter (porosity
Thiosulphate. Dissolve 1 g in 10 ml of water, add 0.1 ml of No. 4) and wash with water until the filtrate is colourless. The
starch solution and 0.1 ml of 0.005 M iodine; a blue colour is weight of the residue, dried at 105º to constant weight, is not
produced. more than 5 mg (1.0 per cent).
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Assay. Weigh accurately about 0.3 g, dissolve in sufficient
on 1.0 g of the powdered substance by drying in an oven at
water to produce 100.0 ml. To 20.0 ml add 20 ml of water, 1 g of
105º for 3 hours.
potassium iodide and 10 ml of 2 M hydrochloric acid and
Assay. Weigh accurately about 0.35 g, dissolve in about 10 ml titrate the liberated iodine with 0.1 M sodium thiosulphate
of water, add 35 ml of hydrochloric acid and 5 ml of chloroform. using starch solution, added towards the end of the titration,
Titrate with 0.05 M potassium iodate until the purple colour as indicator.
of iodine disappears from the chloroform. Add the last portion
1 ml of 0.1 M sodium thiosulphate is equivalent to 0.003160 g
of the iodate solution dropwise and agitate vigorously and
of KMnO4.
continuously. Allow to stand for 5 minutes. If any colour
develops in the chloroform layer continue the titration until Storage. Store protected from moisture.
the chloroform is decolorised. CAUTION — Great care should be taken in handling
1 ml of 0.05 M potassium iodate is equivalent to 0.0166 g of potassium permanganate as dangerous explosions are liable
KI. to occur if it is brought into contact with organic or other
Storage. Store protected from light and moisture. readily oxidisable substances, either in solution or in the
dry condition.

Potassium Permanganate
Povidone
KMnO4 Mol. Wt. 158.0
Potassium Permanganate contains not less than 99.0 per cent
Polyvinylpyrrolidone; Polyvidone
and not more than 100.5 per cent of KMnO4.
Description. A dark purple or brownish black, granular powder
or dark purple or almost black slender, prismatic crystals, having
CH CH2
a metallic lustre; odourless. It decomposes on contact with
certain organic substances. N O

Identification
n
A. A solution in water acidified with sulphuric acid and heated
to 70º is decolorised by hydrogen peroxide solution (20 vol). (C6H9NO)n Mol. Wt. (111.2)n

962
IP 2007 POTASSIUM PERMANGANATE

Povidone is poly(2-oxopyrrolidin-1-ylethylene) and consists not more intense than that obtained by treating in the same
of linear polymers of 1-vinylpyrrolidin-2-one. The different manner a mixture of 2 ml of the test solution obtained above
types of Povidone are characterised by their viscosity in and 10 ml of the 20 ml of solution obtained by repeating the
solution, expressed as K-value, in the range 10 to 95. procedure using 2 ml of lead standard solution (10 ppm Pb)
Povidone with a nominal K-value of 15 or less has a K-value in place of the substance under examination, adding 0.5 g of
of not less than 85.0 per cent and not more than 115.0 per cent magnesium oxide in a silica crucible and continuing as
of the declared value. The K-value of povidone with a nominal described above beginning at the words “Ignite to dull red
K-value of more than 15, or a nominal K-value range with an heat....” (10 ppm).
average of more than 15, is not less than 90.0 per cent and not Aldehydes. Boil 20.0 g in 180 ml of a 25 per cent v/v solution of
more than 107.0 per cent of the declared value or of the average sulphuric acid in a ground-glass-stoppered flask under a reflux
of the declared range. It contains not less than 11.5 per cent condenser for 45 minutes and allow to cool. Fit a distillation
and not more than 12.8 per cent of nitrogen, N, calculated on head, distil and collect 60 ml of the distillate in 20 ml of a 7.0 per
the anhydrous basis. cent w/v solution of hydroxylamine hydrochloride, previously
adjusted to pH 3.1 using 1 M sodium hydroxide, and cooled
Description. A white or yellowish white powder or flakes;
in ice. Titrate with 0.1 M sodium hydroxide to pH 3.1. Carry
odourless or almost odourless; hygroscopic.
out a blank titration. Not more than 9.1 ml of 0.1 M sodium
hydroxide is required (0.2 per cent, calculated as acetaldehyde,
Identification
C2H4O).
Test A may be omitted if tests B, C and D are carried out. Tests Vinylpyrrolidone. Dissolve 10.0 g in 80 ml of water and add
B and C may be omitted if tests A and D are carried out. 1 g of sodium acetate. Titrate with 0.05 M iodine until a
A. Determine by infrared absorption spectrophotometry (2.4.6). persistent colour is obtained and add a further 3.0 ml of the
Compare the spectrum with that obtained with povidone RS. iodine solution. Allow to stand for 10 minutes and titrate the
excess of iodine with 0.1 M sodium thiosulphate using 3 ml of
B. Add 2.5 g in small portions to a suitable volume of carbon
starch solution, added towards the end of the titration, as
dioxide-free water, stirring with a magnetic stirrer, and dilute
indicator. Repeat the operation without the substance under
to 25 ml with the same solvent (solution A). To 0.4 ml of solution
examination using the same total volume of 0.05M iodine. The
A add 10 ml of water, 5 ml of 2 M hydrochloric acid and 2 ml
difference between the titrations represents the amount of
of potassium dichromate solution; an orange-yellow
iodine consumed by the vinylpyrrolidone monomer that may
precipitate is produced.
be present. Not more than 3.6 ml of 0.05 M iodine is required
C. To 1 ml of solution A add 0.2 ml of dimethylamino- (0.2 per cent).
benzaldehyde reagent and 0.1 ml of sulphuric acid; a pink
Sulphated ash (2.3.18). Not more than 0.1 per cent.
colour is produced.
Water (2.3.43). Not more than 5.0 per cent determined on 0.5 g.
D. To 0.1 ml of solution A add 5 ml of water and 0.2 ml of
0.05 M iodine; a red colour is produced. K-value. For Povidone with a stated K-value of 18 or less,
prepare a 5.0 per cent w/v solution. For Povidone with a
Tests declared K-value of more than 18, prepare a 1.0 per cent w/v
solution. Allow the solution to stand for 1 hour and carry out
Appearance of solution. Solution A is clear (2.4.1), and not Method B for the determination of viscosity (2.4.28), at 25º ±
more intensely coloured than reference solution BS6 or BYS6 0.2º using a size no. 1 viscometer with a minimum flow time of
(2.4.1). 100 seconds. Calculate the K-value (Ko) from the expression
Heavy metals. Mix 2.0 g with 0.5 g of magnesium oxide in a
silica crucible. Ignite to dull red heat until a homogeneous 1.5 log z − 1 300 c log z + (c + 1.5 c log z) 2
white or greyish white mass is produced. Heat at 800º for ΚΟ = +
0.15 + 0.003 c 0.15 c + 0.003 c 2
about 1 hour, dissolve the residue using two quantities, each
of 5 ml, of 5 M hydrochloric acid, add 0.1 ml of where c is the percentage concentration w/v of the substance
phenolphthalein solution and strong ammonia solution until under examination, calculated on the anhydrous basis, and z
a pink colour is produced. Cool, add glacial acetic acid until is the viscosity of the solution relative to that of water.
the solution is decolorised and add a further 0.5 ml. Filter if Nitrogen (2.3.30). Follow Method C, using 0.3 g, accurately
necessary and dilute the solution to 20 ml with water. To 12 ml weighed and 11 ml of nitrogen-free sulphuric acid. For
of the resulting solution add 2 ml of acetate buffer pH 3.5, mix, complete destruction of organic matter repeat the addition of
add 1.2 ml of thioacetamide reagent, mix immediately and hydrogen peroxide (10 vol) (usually 3 to 6 times) until a clear,
allow to stand for 2 minutes. Any brown colour produced is light-green solution is obtained, then heat for a further 4 hours.

963
POVIDONE-IODINE IP 2007

Storage. Store protected from moisture. on the dried basis, subtract the percentage of available iodine
Labelling. The label states the viscosity in terms of a K-value determined in the Assay and obtain the percentage of iodide.
or K-range. Sulphated ash (2.3.18). Not more than 0.2 per cent.
Loss on drying (2.4.19). Not more than 8.0 per cent, determined
on 0.5 g by drying in an oven at 105º.
Povidone-Iodine Assay. Weigh accurately about 3 g, transfer to a beaker and
add 200 ml of water. Cover the beaker and stir with a mechanical
stirrer at room temperature for not more than 1 hour to dissolve
as completely as possible. Titrate immediately thereafter with
CH CH2 0.1 M sodium thiosulphate using 3 ml of starch solution,
,xI added towards the end of the titration, as indicator.
N O
1 ml of 0.1 M sodium thiosulphate is equivalent to 0.01269 g
of I.
n
Storage. Store protected from light.
Povidone-Iodine is a complex produced by interaction
between iodine and poly(2-oxopyrrolidin-1-ylethylene).
Povidone-Iodine contains not less than 9.0 per cent and not Povidone-Iodine Solution
more than 12.0 per cent of available iodine, I, calculated on the
Povidone-Iodine Solution is a solution of Povidone-Iodine in
dried basis.
Purified Water. It may contain a small amount of Ethanol.
Description. A yellowish brown amorphous powder; odour,
Povidone-Iodine Solution contains not less than 85.0 per cent
slight and characteristic of iodine.
and not more than 120.0 per cent of the stated amount of
Identification iodine, I.
Description. A deep brown liquid; odour, characteristic of
A. Add 0.05 ml of a 10 per cent w/v solution to a mixture of 1 ml
iodine.
of starch solution and 9 ml of water; a deep blue colour is
produced. Identification
B. Spread 1 ml of a 10 per cent w/v solution over an area of A. Dilute 1 ml to 20 ml with water and add 1 ml of the resulting
about 20 cm x 20 cm on a glass plate and allow it to dry in air at solution to a mixture of 1 ml of starch solution and 9 ml of
room temperature and in an atmosphere of low humidity water; a deep blue colour is produced.
overnight; a brown, dry, non-smearing film is formed which
dissolves readily in water. B. Transfer 10 ml to a small flask and cover the mouth of the
flask with a filter paper soaked in starch solution; no blue
Tests colour is produced on the paper within 60 seconds.
Heavy metals (2.3.13). 1.0 g complies with the limit test for C. Dilute 20 ml to 100 ml with water. To 10 ml add dropwise
heavy metals, Method B (20 ppm). 0.1 M sodium thiosulphate until the colour is just discharged.
To 5 ml of the resulting solution add 5 ml of 2 M hydrochloric
Nitrogen (2.3.30). 9.5 to 11.5 per cent, calculated on the dried acid and 2 ml of potassium dichromate solution; a red
basis, determined on about 0.3 g, by Method A. precipitate is produced.
Iodide. Not more than 6.6 per cent, calculated on the dried
basis, determined by the following method. Weigh accurately Tests
about 0.5 g and dissolve in 100 ml of water. Add sufficient pH (2.4.24). 3.0 to 6.5.
sodium bisulphite solution until the colour of iodine is
discharged. Add 25.0 ml of 0.1 M silver nitrate and 10 ml of Ethanol (if present) (2.3.45). 90.0 to 110.0 per cent of the stated
nitric acid and mix. Titrate with 0.1 M ammonium thiocyanate, amount of C2H5OH, determined by Method A after decolorising
using ferric ammonium sulphate solution as indicator. Repeat the solution with just sufficient quantity of a 10 per cent w/v
the procedure without the substance under examination. The solution of sodium thiosulphate and treating with a few drops
difference between the titrations represents the amount of of dilute sodium hydroxide solution.
silver nitrate required. 1 ml of 0.1 M silver nitrate is equivalent Assay. To an accurately measured volume containing about
to 0.01269 g of I. From the percentage of total iodine, calculated 50 mg of iodine add sufficient water to produce not less than

964
IP 2007 PRALIDOXIME CHLORIDE INJECTION

30 ml and titrate immediately with 0.02 M sodium thiosulphate, nonaethoxyethanol and titrate with 0.1 M silver nitrate,
using 3 ml of starch solution, added towards the end of the determining the end-point potentiometrically (2.4.25).
titration, as indicator. Carry out a blank titration. 1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of Cl.
1 ml of 0.02 M sodium thiosulphate is equivalent to 0.002538
Sulphated ash (2.3.18). Not more than 0.5 per cent.
g of I.
Loss on drying (2.4.19). Not more than 2.0 per cent, determined
Storage. Store protected from light.
on 1.0 g by drying in an oven at 105º for 3 hours.
Labelling. The label states the quantities of iodine and ethanol Assay. Weigh accurately about 0.5 g, dissolve in sufficient
(if present) as percentages w/v. water to produce 250.0 ml and mix. Dilute 5.0 ml of this solution
to 100.0 ml with water. Transfer 5.0 ml to a 50-ml volumetric
flask, dilute to about 40 ml with water, add 5.0 ml of 1 M
Pralidoxime Chloride sodium hydroxide and dilute to volume with water. Within 10
minutes of the addition of the alkali, measure the absorbance
CH3 of the solution at the maximum at about 332 nm (2.4.7), using a
solution of 5.0 ml of 1 M sodium hydroxide diluted with water
N
NOH to 50.0 ml as the blank.
Cl
Calculate the content of C7H9ClN2O from the absorbance
obtained by carrying out the assay simultaneously using about
C7H9ClN2O Mol. Wt. 172.6 0.5 g, accurately weighed, of pralidoxime chloride RS.
Pralidoxime Chloride is 2-hydroxyiminomethyl-1- Storage. Store protected from moisture.
methylpyridinium chloride.
Pralidoxime Chloride contains not less than 97.0 per cent and
not more than 103.0 per cent of C7H9ClN2O, calculated on the Pralidoxime Chloride Injection
dried basis.
Pralidoxime Chloride Injection is a sterile material consisting
Description. A white to pale yellow, crystalline powder; of Pralidoxime Chloride with or without buffering agents and
odourless. other excipients. It is filled in a sealed container.

Identification The injection is constituted by dissolving the contents of the


sealed container in the requisite amount of sterile Water for
A. Determine by infrared absorption spectrophotometry (2.4.6). Injections, immediately before use.
Compare the spectrum with that obtained with pralidoxime
The constituted solution complies with the requirements for
chloride RS or with the reference spectrum of pralidoxime
Clarity of solution and Particulate matter stated under
chloride.
Parenteral Preparations (Injections).
B. When examined in the range 230 nm to 360 nm (2.4.7), a Storage. The constituted solution should be used immediately
0.0005 per cent w/v solution in 0.1 M hydrochloric acid shows after preparation but, in any case, within the period
absorption maxima at about 242 nm and 292 nm and in recommended by the manufacturer.
0.1 M sodium hydroxide, a maximum at about 332 nm.
Pralidoxime Chloride Injection contains not less than 90.0 per
C. To 0.1 ml of a 20 per cent w/v solution add 1 ml of a 0.6 per cent and not more than 110.0 per cent of the stated amount of
cent w/v solution of ferric chloride; an amber-brown colour pralidoxime chloride, C7H9ClN2O.
is produced.
Description. A white to pale yellow powder.
D. Gives the reactions of chlorides (2.3.1).
The contents of the sealed container comply with the
Tests requirements stated under Parenteral Preparations (Powders
for Injection) and with the following requirements.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method A (20 ppm). Identification
Chloride content. 20.2 to 20.8 per cent, calculated on the dried A. Determine by infrared absorption spectrophotometry (2.4.6).
basis, determined by the following method. Weigh accurately Compare the spectrum with that obtained with pralidoxime
about 0.3 g, dissolve in about 150 ml of water, add 20 ml of chloride RS or with the reference spectrum of pralidoxime
glacial acetic acid and 10 drops of (4-tert-octylphenoxy) chloride.

965
PRAZOSIN HYDROCHLORIDE IP 2007

B. When examined in the range 230 nm to 360 nm (2.4.7), a Description. A white or almost white powder; odourless or
0.0005 per cent w/v solution in 0.1 M hydrochloric acid shows almost odourless.
absorption maxima at about 242 nm and 292 nm and in
0.1 M sodium hydroxide, a maximum at about 332 nm. Identification
C. To 0.1 ml of a 20 per cent w/v solution add 1 ml of a 0.6 per Test A may be omitted if tests B, C and D are carried out. Tests
cent w/v solution of ferric chloride; an amber-brown colour B and C may be omitted if tests A and D are carried out.
is produced. A. Determine by infrared absorption spectrophotometry (2.4.6).
D. Gives the reactions of chlorides (2.3.1). Compare the spectrum with that obtained with prazosin
hydrochloride RS.
Tests B. Prepare a 0.05 per cent w/v solution of the substance under
pH (2.4.24). 3.5 to 4.5, determined in a 5.0 per cent w/v solution. examination in a 0.1 per cent v/v solution of hydrochloric
acid in methanol. Dilute separately 1 ml and 5 ml of this solution
Heavy metals (2.3.13). 1.0 g complies with the limit test for to 100 ml with the same acid solution (solutions A and B
heavy metals, Method A (20 ppm). respectively). When examined in the range 220 nm to 280 nm
Bacterial endotoxins (2.2.3). Not more than 0.1 Endotoxin Unit (2.4.7), solution A shows an absorption maximum at about
per mg of pralidoxime chloride. 247 nm; absorbance at the maximum, 0.66 to 0.70. When
examined in the range 280 nm to 400 nm, solution B exhibits
Assay. Weigh accurately about 0.5 g, dissolve in sufficient
two maxima at about 330 nm and 343 nm; absorbances at the
water to produce 250.0 ml and mix. Dilute 5.0 ml of this solution
two maxima, 0.65 to 0.70 and 0.60 to 0.66 respectively.
to 100.0 ml with water. Transfer 5.0 ml to a 50-ml volumetric
flask, dilute to about 40 ml with water, add 5.0 ml of C. In the test for Related substances, the principal spot in the
1 M sodium hydroxide and dilute to volume with water. Within chromatogram obtained with test solution (a) corresponds to
10 minutes of the addition of the alkali, measure the absorbance that in the chromatogram obtained with reference solution (b).
of the solution at the maximum at about 332 nm (2.4.7), using a D. A 0.1 per cent w/v solution gives the reactions of chlorides
solution of 5.0 ml of 1 M sodium hydroxide diluted with water (2.3.1).
to 50.0 ml as the blank.
Tests
Calculate the content of C7H9ClN2O from the absorbance
obtained by carrying out the assay simultaneously using about Iron. To 1.0 g add dropwise about 1.5 ml of nitric acid, heat
0.5 g, accurately weighed, of pralidoxime chloride RS. cautiously on a water-bath until fumes are no longer evolved.
Ignite by slowly raising the temperature from 150º to 1000º,
Labelling. The label states the period within which the
maintaining the final temperature for 1 hour. Cool, dissolve
constituted injection should be used.
the residue in 20 ml of 2 M hydrochloric acid, evaporate to
about 5 ml, dilute to 25 ml with 2 M hydrochloric acid and
examine the resulting solution by atomic absorption
Prazosin Hydrochloride spectrophotometry (2.4.2), measuring at 248 nm using an iron
hollow-cathode light as a source of radiation, an air-acetylene
flame and iron standard solution (8 ppm Fe), diluted as
O necessary with water to prepare the standard solutions
O (100 ppm).
N
H3CO N N Reserve about 10 ml of the solution for the Nickel test.
, HCl
Nickel. Examine the final solution prepared in the test for Iron
N by atomic absorption spectrophotometry (2.4.2), measuring
H3CO
at 232 nm using a nickel hollow-cathode light as a source of
NH2 radiation, an air-acetylene flame and using nickel standard
solution (10 ppm Ni), diluted as necessary with water to
C19H21N5O4, HCl Mol. Wt. 419.9 prepare the standard solutions (50 ppm).
Prazosin Hydrochloride is 2-[4-(2-furoyl)piperazin-1-yl]-6,7- Related substances. Determine by liquid chromatography
dimethoxyquinazolin-4-ylamine hydrochloride. (2.4.14).
Prazosin Hydrochloride contains not less than 98.5 per cent Test solution. Dissolve 50.0 mg of the substance under
and not more than 101.0 per cent of C19H21N5O4, HCl, calculated examination in the mobile phase and dilute to 50 ml with the
on the anhydrous basis. mobile phase.

966
IP 2007 PRAZOSIN TABLETS

Reference solution (a). Dilute 1 ml of the test solution to Prazosin Tablets


100 ml with the mobile phase. Further dilute 1 ml of the solution
to 10 ml with the mobile phase. Prazosin Hydrochloride Tablets
Reference solution (b). Dissolve 8 mg of metoclopramide Prazosin Tablets contain not less than 90.0 per cent and not
hydrochloride RS in 1 ml of the test solution and dilute to more than 110.0 per cent of the stated amount of prazosin,
25 ml with the mobile phase. Further dilute 1 ml of the solution C19H21N5O4. The tablets may contain permitted colours.
to 10 ml with the mobile phase.
Identification
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Shake a quantity of the powdered tablets containing about
octadecylsilyl silica gel (5 µm), 10 mg of prazosin with a mixture of 10 ml of dichloromethane
– mobile phase: a mixture of 50 volumes of methanol and and 10 ml of 0.05 M potassium hydroxide, filter the organic
50 volumes of a solution containing 3.484 g per litre of layer through cotton, evaporate to dryness and dry the residue
sodium pentanesulphonate and 3.64 g per litre of at 60º at a pressure not exceeding 2 kPa at 60º for 2 hours.
tetramethylammonium hydroxide adjusted to pH 5.0 On the residue determine by infrared absorption
with glacial acetic acid, spectrophotometry (2.4.6). Compare the spectrum with that
– flow rate.1 ml per minute, obtained with prazosin RS or with the reference spectrum of
– spectrophotometer set at 254 nm, prazosin.
– a 20 µl loop injector.
Tests
Inject reference solution (b). The retention times are: prazocine
about 9 minutes and metoclopramide about 5 minutes.The Related substances. Determine by thin-layer chromatography
test is not valid unless the resolution between the peaks (2.4.17), coating the plate with silica gel HF254.
corresponding to prazocine and metoclopramide is at least 8. Mobile phase. A mixture of 95 volumes of ethyl acetate and
Inject the test solution and reference solution (a). Continue 5 volumes of diethylamine.
the chromatography of the test solution for 6 times the Test solution. Shake a quantity of the powdered tablets
retention time of the principal peak. Any secondary peak containing 5 mg of prazosin with 2 ml of a mixture of 10 volumes
obtained with the test solution is not more than twice the area of dichloromethane, 10 volumes of methanol and 1 volume of
of the principal peak in the chromatogram obtained with diethylamine, centrifuge and pass the supernatant liquid
reference solution (a) (0.2 per cent).The sum of all the impurities through a 0.5 µm PTFE (Polytetrafluoroethylene) filter.
is not more than five times the area of the principal peak in the
chromatogram obtained with reference solution (a) (0.5 per Reference solution (a). Dilute 1 volume of test solution to
cent). Ignore any peak with an area 0.5 times the area of the 200 volumes with the same solvent mixture
principal peak in the chromatogram obtained with reference Reference solution (b). Dilute 2 volumes of reference solution
solution (a) (0.05 per cent). (a) to 5 volumes with the same solvent mixture.
Heavy metals (2.3.13). Determine on the residue obtained in Apply to the plate 20 µl of each solution. Any secondary spot
the test for Sulphated ash. Dissolve in sufficient 2 M nitric in the chromatogram obtained with the test solution is not
acid to produce 50 ml. The resulting solution complies with more intense than the spot in the chromatogram obtained
the limit test for heavy metals, Method B (50 ppm). with reference solution (a) and not more than two such spots
are more intense than the spot in the chromatogram obtained
Sulphated ash (2.3.18). Not more than 0.2 per cent.
with reference solution (b).
Water (2.3.43). Not more than 0.5 per cent w/w, using 2.0 g Uniformity of content. Comply with the tests stated under
dissolved in a mixture of equal volumes of dichloromethane Tablets.
and methanol.
Determine by liquid chromatography (2.4.14).
Assay. Weigh accurately about 0.35 g, dissolve in 20 ml of
anhydrous formic acid and 30 ml of acetic anhydride. Titrate Test solution. Shake one tablet for 1 hour in a suitable volume
with 0.1 M perchloric acid, determining the end point of a mixture of 96 volumes of methanol, 2 volumes of glacial
potentiometrically (2.4.25). Carry out a blank titration. acetic acid and 2 volumes of water to produce a solution
containing 0.002 per cent w/v solution of prazosin, centrifuge
1 ml of 0.1 M perchloric acid is equivalent to 0.04199 g of and use the supernatant liquid.
C19H21N504, HCl.
Reference solution. A 0.0022 per cent w/v solution of prazosin
Storage. Store protected from light. hydrochloride RS in the same solvent mixture.

967
PREDNISOLONE IP 2007

Chromatographic system Identification


– a stainless steel column 20 cm x 4 mm, packed with porous
silica particles, (5 µm), Test A may be omitted if tests B and C are carried out. Test C
– mobile phase: 0.01 per cent w/v solution of diethylamine may be omitted if tests A and B are carried out.
in a mixture of 96 volumes of methanol, 2 volumes of A. Determine by infrared absorption spectrophotometry (2.4.6).
glacial acetic acid and 2 volumes of water, Compare the spectrum with that obtained with prednisolone
– flow rate. 1 ml per minute, RS or with the reference spectrum of prednisolone.
– spectrophotometer set at 254 nm,
B. Determine by thin-layer chromatography (2.4.17), coating
– a 20 µl loop injector.
the plate with silica gel G.
Calculate the content of C19H21N5O4 in the tablet.
Solvent mixture. A mixture of 90 volumes of acetone and
Other tests. Comply with the tests stated under Tablets. 10 volumes of formamide.
Assay. Determine by liquid chromatography (2.4.14). Mobile phase. Chloroform.
Test solution. Weigh and powder 20 tablets. Shake a quantity Test solution. Dissolve 25 mg of the substance under
of the powdered tablets containing about 2 mg of prazosin examination in 10 ml of the solvent mixture.
with 100 ml in a mixture of 96 volumes of methanol, 2 volumes
Reference solution (a). Dissolve 25 mg of prednisolone RS in
of glacial acetic acid and 2 volumes of water for 30 minutes,
10 ml of the solvent mixture.
centrifuge and use the supernatant liquid.
Reference solution (b). Mix equal volumes of the test solution
Reference solution. A 0.0022 per cent w/v solution of prazosin
and reference solution (a).
hydrochloride RS in a mixture of 96 volumes of methanol,
2 volumes of glacial acetic acid and 2 volumes of water. Place the dry plate in a tank containing a shallow layer of the
solvent mixture, allow the solvent mixture to ascend to the
The chromatographic conditions as described under
top, remove the plate from the tank and allow the solvent to
Uniformity of content may be used.
evaporate. Use within 2 hours, with the flow of the mobile
Calculate the content of C19H21N5O4 in the tablets. phase in the direction in which the aforementioned treatment
Storage. Store protected from light. was done.

Labelling. The label states the strength in terms of the Apply to the plate 2 µl of each solution. Allow the mobile
equivalent amount of prazosin. phase to rise 12 cm. Dry the plate in a current of warm air, allow
the solvent to evaporate, heat at 120º for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120º for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
Prednisolone spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with
O reference solution (a). The principal spot in the chromatogram
H3C OH obtained with reference solution (b) appears as a single,
HO OH compact spot. A.
H3C H
C. Dissolve 2 mg in 2 ml of sulphuric acid by shaking and
H H allow to stand for 5 minutes; an intense red colour is produced
with a reddish brown fluorescence when examined in ultraviolet
O
light at 365 nm. Pour the solution into 10 ml of water and mix;
the colour fades and there is a yellow fluorescence under
C21H28O5 Mol. Wt. 360.5 ultra-violet light (365 nm).
Prednisolone is 11β,17α,21-trihydroxypregna-1,4-diene-3,20-
dione. Tests
Prednisolone contains not less than 96.0 per cent and not Specific optical rotation (2.4.22). +96.0º to +102º, determined
more than 104.0 per cent of C21H28O5, calculated on the dried in a 1.0 per cent w/v solution in dioxan.
basis. Light absorption (2.4.7). Absorbance of a 0.001 per cent w/v
Description. A white or almost white, crystalline powder; solution in ethanol (95 per cent) at the maximum at about
hygroscopic. 240 nm, 0.40 to 0.43.

968
IP 2007 PREDNISOLONE TABLETS

Related substances. Determine by liquid chromatography Sulphated ash (2.3.18). Not more than 0.1 per cent.
(2.4.14). Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Test solution. Dissolve 25 mg of the substance under on 1.0 g by drying in an oven at 105º for 3 hours.
examination in 2 ml of tetrahydrofuran and dilute to 10ml with
Assay. Weigh accurately about 0.1 g and dissolve in sufficient
water.
ethanol to produce 100.0 ml. Dilute 2.0 ml of this solution to
Reference solution (a). Dissolve 2 mg of prednisolone RS 100.0 ml with ethanol. Measure the absorbance of the resulting
and 2 mg of hydrocortisone RS in the mobile phase and dilute solution at the maximum at about 243.5 nm. Calculate the
to 100 ml with the mobile phase. content of C21H28O5 taking 415 as the specific absorbance at
Reference solution (b). Dilute 1 ml of the test solution to 243.5 nm.
100 ml with the mobile phase. Storage. Store protected from light and moisture.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with
base deactivated end-capped octadecylsilyl silica gel
(5 µm), Prednisolone Tablets
– column temperature. 45º,
Prednisolone Tablets contain not less than 90.0 per cent and
– mobile phase: a mixture of 220 ml of tetrahydrofuran
not more than 110.0 per cent of the stated amount of
and 700 ml of water, allowed to equilibrate, diluted to
prednisolone, C21H28O5.
1000 ml with water and mixed again,
– flow rate. 1 ml per minute, Identification
– spectrophotometer set at 254 nm,
– a 20 µl loop injector. Extract a quantity of the powdered tablets containing 30 mg of
Prednisolone with 10 ml of chloroform, filter and evaporate
Equilibrate the column with the mobile phase for about 30
the filtrate to dryness. The residue complies with the following
minutes.
tests.
Adjust the sensitivity of the system so that the height of the
A. Determine by infrared absorption spectrophotometry (2.4.6).
principal peak in the chromatogram obtained with reference
Compare the spectrum with that obtained with prednisolone
solution (b) is not less than 50 per cent of the full scale of the
RS or with the reference spectrum of prednisolone.
recorder.
B. Determine by thin-layer chromatography (2.4.17), coating
Inject reference solution (a). The retention times are:
the plate with silica gel G.
prednisolone, about 14 minutes and hydrocortisone about
15.5 minutes. The test is not valid unless the resolution between Solvent mixture. A mixture of 90 volumes of acetone and
the peaks corresponding to prednisolone and hydrocortisone 10 volumes of formamide.
is at least 2.2. If necessary, adjust the concentration of Mobile phase. Chloroform.
tetrahydrofuran in the mobile phase.
Test solution. Dissolve 25 mg of the residue in 10 ml of the
Inject separately the solvent mixture of the test solution as a solvent mixture.
blank, the test solution and reference solution (b). Continue
the chromatography of the test solution for 4.5 times the Reference solution (a). Dissolve 25 mg of prednisolone RS in
retention time of the principal peak. In the chromatogram 10 ml of the solvent mixture.
obtained with the test solution, the area of any peak other Reference solution (b). Mix equal volumes of the test solution
than the principal peak, is not greater than half the area of the and reference solution (a).
principal peak in the chromatogram obtained with reference
Place the dry plate in a tank containing a shallow layer of the
solution (b) (1 per cent) and not more than one such peak has
solvent mixture, allow the solvent mixture to ascend to the
an area greater than half the area of the principal peak in the
top, remove the plate from the tank and allow the solvent to
chromatogram obtained with reference solution (b) (0.5 per
evaporate. Use within 2 hours, with the flow of the mobile
cent); the sum of the areas of all the peaks other than the
phase in the direction in which the aforementioned treatment
principal peak is not greater than 2.0 times the area of the
was done.
principal peak in the chromatogram obtained with reference
solution (b) (2 per cent). Ignore any peak obtained with the Apply to the plate 2 µl of each solution. Allow the mobile
blank run and any peak with an area less than 0.05 times that phase to rise 12 cm. Dry the plate in a current of warm air, allow
of the principal peak in the chromatogram obtained with the solvent to evaporate, heat at 120º for 15 minutes and spray
reference solution (b). the hot plate with ethanolic sulphuric acid (20 per cent v/v).

969
PREDNISOLONE TABLETS IP 2007

Heat at 120º for a further 10 minutes, allow to cool and examine retention time of the principal peak. In the chromatogram
in daylight and in ultraviolet light at 365 nm. The principal obtained with the test solution, the area of any peak other
spot in the chromatogram obtained with the test solution than the principal peak, is not greater than half the area of the
corresponds to that in the chromatogram obtained with principal peak in the chromatogram obtained with reference
reference solution (a). The principal spot in the chromatogram solution (b) (1 per cent) and the sum of the areas of all the
obtained with reference solution (b) appears as a single, peaks other than the principal peak is not greater than three
compact spot. times the area of the principal peak in the chromatogram
obtained with reference solution (b) (3 per cent). Ignore any
Tests peak obtained with the blank run and any peak with an area
less than 0.05 times that of the principal peak in the
Related substances. Determine by liquid chromatography chromatogram obtained with reference solution (b) (0.05 per
(2.4.14). cent) and any peak with a retention time of 3 minutes or less.
Test solution. Shake a quantity of the powdered tablets Uniformity of content. (For tablets containing 10 mg or less)
containing 10 mg of Prednisolone with 25 ml of methanol for — Comply with the test stated under Tablets.
10 minutes and mix with the aid of ultrasound for 2 minutes;
filter the extract (Whatman GF/F is suitable), wash the filter Powder one tablet, add 50 ml of ethanol (95 per cent), shake
with two 10-ml quantities of methanol, combine the filtrate for 30 minutes, add sufficient ethanol (95 per cent) to produce
and washings and evaporate to dryness using a rotary 100.0 ml. Centrifuge and pipette a suitable volume of the
evaporator and a warm water-bath, dissolve the residue in supernatant liquid containing 0.5 mg of Prednisolone and
10 ml of tetrahydrofuran and dilute to 20 ml with water. dilute to 50.0 ml with ethanol (95 per cent). Measure the
absorbance of the resulting solution at the maximum at about
Reference solution (a). Dissolve 2 mg of prednisolone RS 240 nm (2.4.7). Calculate the content of C21H28O5 taking
and 2 mg of hydrocortisone RS in the mobile phase and dilute 415 specific absorbance at 240 nm.
to 100 ml with the mobile phase.
Dissolution (2.5.2).
Reference solution (b). Dilute 1ml of the test solution to
100 ml with a 50 per cent v/v solution of tetrahydrofuran. Apparatus. No 1
Medium. 900 ml of water
Chromatographic system
Speed and time. 50 rpm and 30 minutes.
– a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilyl silica gel (5 µm), Withdraw a suitable volume of the medium, filter and dilute a
– column temperature. 45º, suitable volume of the filtrate with the same solvent. Measure
– mobile phase: a mixture of 220 ml of tetrahydrofuran the absorbance of the filtrate at the maximum at about 240 nm
and 700 ml of water, allowed to equilibrate, diluted to (2.4.7). Calculate the content of C21H28O5 in the medium from
1000 ml with water and mixed again, the absorbance obtained from a solution of known
– flow rate. 1 ml per minute, concentration of prednisolone RS.
– spectrophotometer set at 254 nm,
D. Not less than 70 per cent of the stated amount of C21H28O5.
– a 20 µl loop injector.
Other tests. Comply with the tests stated under Tablets.
Equilibrate the column with the mobile phase for about
30 minutes. Assay. Determine by liquid chromatography (2.4.14).
Adjust the sensitivity of the system so that the height of the Test solution. Weigh and powder 20 tablets. Weigh accurately
principal peak in the chromatogram obtained with reference a quantity of the powder containing about 5 mg of
solution (b) is not less than 50 per cent of the full scale of the Prednisolone, add 58 ml of methanol, shake for 10 minutes
recorder. and add sufficient water to produce 100.0 ml. Mix well and
filter.
Inject reference solution (a). The retention times are:
prednisolone, about 14 minutes and hydrocortisone about Reference solution (a). A solution containing 0.005 per cent
15.5 minutes. The test is not valid unless the resolution between w/v of prednisolone RS and 0.0075 per cent w/v of
the peaks corresponding to prednisolone and hydrocortisone dexamethasone (internal standard) in a mixture of 58 volumes
is at least 2.2. If necessary, adjust the concentration of of methanol and 42 volumes of water.
tetrahydrofuran in the mobile phase.
Reference solution (b). Prepare in the same manner as the test
Inject separately the solvent mixture of the test solution as a solution but adding 10 ml of a 0.075 per cent w/v solution of
blank, the test solution and reference solution (b). Continue dexamethasone in methanol and 48 ml of methanol in place of
the chromatography of the test solution for 4.5 times the 58 ml of methanol.

970
IP 2007 PREDNISOLONE SODIUM PHOSPHATE

Chromatographic system paper with dilute ammonia solution. The solution complies
– a stainless steel column 20 cm x 4.6 mm, packed with with reaction A of sodium salts and reaction B of phosphates
octadecylsilyl silica gel (5 µm), (2.3.1).
– mobile phase: a mixture of 42 volumes of water and
58 volumes of methanol, Tests
– flow rate. 1 ml per minute,
– spectrophotometer set at 254 nm, pH (2.4.24). 7.5 to 10.5 determined in 1.0 per cent w/v solution.
– a 20 µl loop injector. Specific optical rotation (2.4.22). +95.0º to +102.0º, determined
The assay is not valid unless the resolution factor between in a 1.0 per cent w/v solution in a mixture of 9 volumes of
the peaks due to prednisolone and dexamethasone is greater phosphate buffer pH 7.0 and 1 volume of carbon dioxide-free
than 2.5 and the column efficiency, determined using the peak water.
due to prednisolone in the chromatogram obtained with the
Related substances. Determine by liquid chromatography
test solution is greater than 15,000 theoretical plates per metre.
(2.4.14).
Calculate the content of C21H28O5 in the tablets.
Test solution. Dissolve 62.5 mg of the substance under
Storage. Store protected from light. examination in the mobile phase and dilute to 25.0 ml with the
mobile phase.
Reference solution (a). Dissolve 25 mg of prednisolone
Prednisolone Sodium Phosphate sodium phosphate RS and 25 mg of prednisolone RS in the
mobile phase and dilute to 25.0 ml with the same solvent.
Dilute 1.0 ml of the solution to 25.0 ml with the mobile phase.
O O
O P ONa Reference solution (b). Dilute 1.0 ml of the test solution to
H3C
HO 50.0 ml with the mobile phase.
OH ONa
H3C H Chromatographic system
– a stainless steel column 15 cm x 4.6 mm, packed with
H H octadecylsilane bonded to porous silica or ceramic
O microparticles (5 µm),
– mobile phase: weigh 1.360 g of potassium dihydrogen
C21H27Na2O8P Mol. Wt. 484.4 phosphate and 0.6 g of hexylamine, mix and allow to
Predisolone Sodium Phosphate is 11β,17α, 21- stand for 10 minutes and then dissolve in 185 ml of
trihydroxypregna-1,4-diene-3,20-dione-21- water, add 65 ml of acetonitrile, mix and filter through a
(dihydrogenphosphate)disodium salt. 0.45 µm filter,
– flow rate. 1 ml per minute,
– spectrophotometer set at 254 nm,
Prednisolone Sodium Phosphate contains not less than 96.0 – a 20 µl loop injector.
per cent and not more than 102.0 per cent of C21H27Na2O8P, Inject reference solution (b). Adjust the sensitivity of the
calculated on the dried basis. system so that the height of the principal peak in the
chromatogram obtained with reference solution (b) is 70 per
Identification
cent to 90 per cent of the full scale of the recorder.
A. Determine by infrared absorption spectrophotometry (2.4.6). Equilibrate the column with the mobile phase for about 30
Compare the spectrum with that obtained with prednisolone minutes.
sodium phosphate RS. If the spectra obtained in the solid
state show differences, dissolve the substance under Inject reference solution (a). The retention times are:
examination and the reference substance separately in the prednisolone sodium phosphate, about 6.5 minutes and
minimum volume of ethanol (95 per cent) evaporate to dryness prednisolone, about 8.5 minutes. The test is not valid unless
on a water-bath and record the spectra again using the residues. the resolution between the peaks due to prednisolone sodium
phosphate and prednisolone is at least 4.5; if this resolution is
B. To about 40 mg add 2 ml of sulphuric acid and heat gently
not achieved, increase the concentration of acetonitrile or
until white fumes are evolved. Add nitric acid dropwise,
increase the concentration of water in the mobile phase.
continue the heating until the solution is almost colourless
and cool. Add 2 ml of water, heat until white fumes are again Inject separately the test solution and reference solution (b).
evolved, cool, add 10 ml of water and neutralise to red litmus Continue the chromatography for three times the retention

971
PREDNISOLONE SODIUM PHOSPHATE INJECTION IP 2007

time of the principal peak. In the chromatogram obtained with Tests


the test solution, the area of any peak other than that from the
principal peak is not greater than the area of the principal peak pH (2.4.24). 7.0 to 8.0.
in the chromatogram obtained with reference solution (b) (2 Bacterial endotoxins (2.2.3). Not more than 5.0 Endotoxin Units
per cent) and not more than one such peak has an area greater per mg of prednisolone phosphate.
than half the area of the principal peak in the chromatogram Other tests. Complies with the tests stated under Parenteral
obtained with reference solution (b) (1 per cent); the sum of Preparations (Injections).
the areas of all the peaks other than the principal peak is not
greater than 1.5 times the area of the principal peak in the Assay. Determine by liquid chromatography (2.4.14).
chromatogram obtained with reference solution (b) (3 per Test solution. Dilute an accurately measured volume of the
cent). Ignore any peak due to the solvent and any peak with injection to obtain a solution containing 0.001 per cent w/v of
an area less than 0.025 times the area of the principal peak in prednisolone phosphate.
the chromatogram obtained with reference solution (b). Reference solution (a). Weigh accurately about 10 mg of
Inorganic phosphate. Not more than 1.0 per cent of phosphate, prednisolone sodium phosphate RS, dissolve in sufficient
PO4. water to produce 100.0 ml (solution A) and dilute 10.0 ml of
the solution to 100.0 ml with water.
Dissolve 50 mg in sufficient water to produce 100 ml. To 10 ml
of the resulting solution add 5 ml of molybdovanadic reagent, Reference solution (b). Add 10 ml of a 0.01 per cent w/v
mix and allow to stand for 5 minutes. Any yellow colour in the solution of betamethasone sodium phosphate RS in water to
solution is not more intense than that produced in a standard 10 ml of solution A and dilute to 100 ml with water.
prepared at the same time in the same manner using 10 ml of Chromatographic system
phosphate standard solution (5 ppm PO4). – a stainless steel column 20 cm x 4.6 mm, packed with
Water (2.3.43). Not more than 6.5 per cent, determined on octadecylsilane bonded to porous silica or ceramic
1.0 g. microparticles (10 µm) (such as Spherisorb ODS 1),
– mobile phase: a mixture of 45 volumes of methanol and
Assay. Weigh accurately about 0.1 g, dissolve in sufficient
55 volumes of citro-phosphate buffer pH 5.0,
water to produce 100.0 ml and mix. Dilute 5.0 ml of the resulting
– flow rate. 2.0 ml per minute,
solution to 250.0 ml with water. Measure the absorbance at
– spectrophotometer set at 247 nm,
the maximum at about 247 nm (2.4.7). Calculate the content of
– a 10 µl loop injector.
C21H27Na2O8P taking 312 as the specific absorbance at 247 nm.
Inject reference solution (b). The test is not valid unless the
Storage. Store protected from light.
resolution between the peaks due to betamethasone sodium
phosphate and prednisolone sodium phosphate is at least 2.5.
Inject alternatively the test solution and reference solution (a).
Prednisolone Sodium Phosphate Calculate the content of C21H29O8P in the injection.
Injection Storage. Store protected from light, in a single-dose or in multi-
Prednisolone Sodium Phosphate Injection is a sterile solution dose containers.
of Prednisolone Sodium Phosphate in Water for Injections.
Prednisolone Sodium Phosphate Injection contains not less
than 90.0 per cent and not more than 110.0 per cent of the Prednisone
stated amount of prednisolone phosphate, C21H29O8P.
Description. A clear, colourless liquid. O
H3C OH
Identification O OH
H3C H
A. In the Assay, the chromatogram obtained with the test
solution corresponds to the peak due to prednisolone sodium H H
phosphate in the chromatogram obtained with reference
O
solution (a).
B. To a volume containing 0.2 mg of Prednisolone Sodium C21H26O5 Mol. Wt. 358.4
Phosphate slowly add 1 ml of sulphuric acid and allow to Prednisone is 17α,21-dihydroxypregna-1,4-diene-3,11,20-
stand for 2 minutes. A deep red colour is produced. trione.

972
IP 2007 PREDNISOLONE SODIUM PHOSPHATE INJECTION

Prednisone contains not less than 96.0 per cent and not more Tests
than 104.0 per cent of C21H26O5, calculated on the dried basis.
Specific optical rotation (2.4.22). +167º to +175º, determined
Description. A white or almost white, crystalline powder;
in a 1.0 per cent w/v solution in dioxan.
odourless.
Light absorption (2.4.7). Absorbance of a 0.001 per cent w/v
Identification solution in methanol at the maximum at about 240 nm, 0.40 to
Tests A and B may be omitted if tests C and D are carried out. 0.43; the ratio of the absorbance at the maximum at about
Tests C and D may be omitted if tests A and B are carried out. 240 nm to that at about 263 nm, 1.85 to 2.05.
A. Determine by infrared absorption spectrophotometry (2.4.6). Related substances. Determine by liquid chromatography
Compare the spectrum with that obtained with prednisone RS (2.4.14).
or with the reference spectrum of prednisone.
Test solution. Dissolve 25 mg of the substance under
B. Determine by thin-layer chromatography (2.4.17), coating examination in methanol and dilute to 10 ml with the same
the plate with silica gel G. solvent.
Solvent mixture. A mixture of 90 volumes of acetone and
Reference solution (a). Dissolve 2.0 mg of prednisolone RS
10 volumes of formamide.
and 2.0 mg of prednisone RS in methanol and dilute to 100 ml
Mobile phase. Chloroform. with the same solvent.
Test solution. Dissolve 25 mg of the substance under
Reference solution (b). Dilute 1 ml of the test solution to
examination in 10 ml of the solvent mixture.
100 ml with methanol.
Reference solution (a). Dissolve 25 mg of prednisone RS in
10 ml of the solvent mixture. Chromatographic system
Reference solution (b). Mix equal volumes of the test solution – a stainless steel column 25 cm x 4.6 mm, packed with
and reference solution (a). octadecylsilyl silica gel (5 µm),
– column temperature. 45º,
Place the dry plate in a tank containing a shallow layer of the
– mobile phase: A. a mixture of 100 ml of acetonitrile,
solvent mixture, allow the solvent mixture to ascend to the
200 ml of methanol and 650 ml of water, allowed to
top, remove the plate from the tank and allow the solvent to
equilibrate, diluted to 1000 ml with water and mixed again,
evaporate. Use within 2 hours, with the flow of the mobile
B. acetonitrile,
phase in the direction in which the aforementioned treatment
–flow rate. 2.5 ml per minute,
was done.
– a linear gradient programme using the conditions given
Apply to the plate 2 µl of each solution. Allow the mobile below,
phase to rise 12 cm. Dry the plate in a current of warm air, allow – spectrophotometer set at 254 nm,
the solvent to evaporate, heat at 120º for 15 minutes and spray – a 20 µl loop injector.
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120º for a further 10 minutes, allow to cool and examine Time Mobile Mobile Comment
in daylight and in ultraviolet light at 365 nm. The principal Phase A Phase B
spot in the chromatogram obtained with the test solution (min) (per cent v/v) (per cent v/v)
corresponds to that in the chromatogram obtained with 0 100 0 Isocratic
reference solution (a). The principal spot in the chromatogram 25 100 0 begin linear
obtained with reference solution (b) appears as a single, gradient
compact spot. 40 40 60 end chromatogram,
C. Dissolve 2 mg in 2 ml of sulphuric acid and allow to stand change to 100B
for 5 minutes; an orange colour is produced within 5 minutes, 41 0 100 being treatment
which exhibits a blue fluorescence in ultraviolet light at 365 with B
nm. Pour the solution into 10 ml of water; the colour changes
first to yellow and then fades gradually but the blue 46 0 100 end treatment,
fluorescence in ultraviolet light remains. return to 100A
D. Dissolve 1 mg in 1 ml of ethanol (95 per cent), evaporate to 47 100 0 begin equilibration
dryness at a pressure not exceeding 0.7 kPa, add 5 ml of 1 M with A
sodium hydroxide and heat at 70º for 30 minutes; not more 52=0 100 0 end equilibration,
than a slight yellow colour is produced (distinction from begin next
cortisone acetate). chromatogram

973
PREDNISONE TABLETS IP 2007

Equilibrate the column with the mobile phase B for at least A. Determine by infrared absorption spectrophotometry (2.4.6).
30 minutes and then with mobile phase A for 5 minutes. For Compare the spectrum with that obtained with prednisone RS
subsequent chromatograms, use the conditions described from or with the reference spectrum of prednisone.
40.0 to 52.0 minutes. B. Determine by thin-layer chromatography (2.4.17), coating
Adjust the sensitivity of the system so that the height of the the plate with silica gel G.
principal peak in the chromatogram obtained with reference Solvent mixture. A mixture of 90 volumes of acetone and
solution (b) is not less than 50 per cent of the full scale of the 10 volumes of formamide.
recorder.
Mobile phase. Chloroform.
Inject reference solution (a). The retention times are:
prednisone, about 19 minutes and prednisolone about Test solution. Dissolve 25 mg of the residue in 10 ml of the
23 minutes. The test is not valid unless the resolution between solvent mixture.
the peaks corresponding to prednisone and prednisolone is Reference solution (a). Dissolve 25 mg of prednisone RS in
at least 2.7. If necessary, adjust the concentration of acetonitrile 10 ml of the solvent mixture.
in mobile phase A.
Reference solution (b). Mix equal volumes of the test solution
Inject separately methanol as a blank, the test solution and and reference solution (a).
reference solution (b). In the chromatogram obtained with the
Place the dry plate in a tank containing a shallow layer of the
test solution: the area of any peak other than the principal
solvent mixture, allow the solvent mixture to ascend to the
peak is not greater than 0.25 times the area of the principal
top, remove the plate from the tank and allow the solvent to
peak in the chromatogram obtained with reference solution
evaporate. Use within 2 hours, with the flow of the mobile
(b) (0.25 per cent); the sum of the areas of all the peaks, apart
phase in the direction in which the aforementioned treatment
from the principal peak, is not greater than 0.75 times the area
was done.
of the principal peak in the chromatogram obtained with
reference solution (b) (0.75 per cent). Ignore any peak due to Apply to the plate 2 µl of each solution. Allow the mobile
the blank run and any peak with an area less than 0.05 times phase to rise 12 cm. Dry the plate in a current of warm air, allow
the area of the principal peak in the chromatogram obtained the solvent to evaporate, heat at 120º for 15 minutes and spray
with reference solution (b). the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120º for a further 10 minutes, allow to cool and examine
Sulphated ash (2.3.18). Not more than 0.1 per cent.
in daylight and in ultraviolet light at 365 nm. The principal
Loss on drying (2.4.19). Not more than 1.0 per cent, determined spot in the chromatogram obtained with the test solution
on 1.0 g by drying in an oven at 105º for 3 hours. corresponds to that in the chromatogram obtained with
reference solution (a). The principal spot in the chromatogram
Assay. Weigh accurately about 0.1 g and dissolve in sufficient
obtained with reference solution (b) appears as a single,
ethanol to produce 100.0 ml. Dilute 2.0 ml of this solution to
compact spot.
100.0 ml with ethanol. Measure the absorbance of the resulting
solution at the maximum at about 238 nm. Calculate the content
Tests
of C21H26O5 taking 425 as the specific absorbance at 238 nm.
Storage. Store protected from light. Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Shake a quantity of the powdered tablets
containing 25 mg of Prednisone with 10 ml of methanol for
Prednisone Tablets 10 minutes, mix with the aid of ultrasound for 2 minutes and
filter the extract (Whatman GF/F is suitable).
Prednisone Tablets contain not less than 90.0 per cent and
Reference solution (a). Dissolve 2.0 mg of prednisolone RS
not more than 110.0 per cent of the stated amount of
and 2.0 mg of prednisone RS in methanol and dilute to 100 ml
prednisone, C21H26O5.
with the same solvent.
Identification Reference solution (b). Dilute 1 ml of the test solution to
100 ml with methanol.
Shake a quantity of the powdered tablets containing 30 mg of
Prednisone with 10 ml of chloroform, filter and evaporate the Chromatographic system
filtrate to dryness. The residue complies with the following – a stainless steel column 25 cm x 4.6 mm, packed with
tests. octadecylsilyl silica gel (5 µm),

974
IP 2007 PREDNISONE TABLETS

– column temperature. 45º, the blank run and any peak with an area less than 0.05 times
– mobile phase: A. a mixture of 100 ml of acetonitrile, the area of the principal peak in the chromatogram obtained
200 ml of methanol and 650 ml of water, allowed to with reference solution (b).
equilibrate, diluted to 1000 ml with water and mixed again, Uniformity of content. Comply with the test stated under
B. acetonitrile, Tablets.
– flow rate. 2.5 ml per minute,
– a linear gradient programme using the conditions given Powder one tablet, add 50 ml of ethanol (95 per cent), shake
below, for 30 minutes, add sufficient ethanol (95 per cent) to produce
– spectrophotometer set at 254 nm, 100.0 ml. Centrifuge and pipette a suitable volume of the
– a 20 µl loop injector. supernatant liquid equivalent to 0.5 mg of Prednisone and
Time Mobile Mobile Comment dilute to 50.0 ml with ethanol (95 per cent). Measure the
Phase A Phase B absorbance of the resulting solution at the maximum at about
(min) (per cent v/v) (per cent v/v) 240 nm (2.4.7). Calculate the content of C21H26O5 taking 415 as
specific absorbance at 240 nm.
0 100 0 Isocratic
25 100 0 begin linear Dissolution (2.5.2).
gradient Apparatus. No 1
40 40 60 end chromatogram, Medium. 900 ml of water
change to 100B Speed and time. 50 rpm and 30 minutes.
41 0 100 being treatment Withdraw a suitable volume of the medium and filter. Measure
with B the absorbance of the filtrate at the maximum at about 240 nm
46 0 100 end treatment, (2.4.7). Calculate the content of C21H26O5 in the medium from
return to 100A the absorbance obtained from a solution of known
47 100 0 begin equilibration concentration of prednisone RS.
with A D. Not less than 70 per cent of the stated amount of C21H26O5.
52=0 100 0 end equilibration, Other tests. Comply with the tests stated under Tablets.
begin next
chromatogram Assay. Determine by liquid chromatography (2.4.14).
Equilibrate the column with the mobile phase B for at least Internal standard solution. Dissolve an accurately weighed
30 minutes and then with mobile phase A for 5 minutes. For quantity of acetanilide in a 50 per cent v/v solution of methanol
subsequent chromatograms use the conditions described from to obtain a solution having a known concentration of 0.11 mg
40.0 to 52.0 minutes. per ml.
Adjust the sensitivity of the system so that the height of the Test solution. Weigh and powder 20 tablets. To an accurately
principal peak in the chromatogram obtained with reference weighed quantity of the powdered tablets containing about
solution (b) is not less than 50 per cent of the full scale of the 20 mg of Prednisone add 5 ml of water, mix with the aid of
recorder. ultrasound for 1 minute, add 50 ml of methanol and mix with
Inject reference solution (a). The retention times are: the aid of ultrasound for I minute. Dilute with water to 100.0 ml
prednisone, about 19 minutes and prednisolone about and mix. To 5.0 ml of this solution add 5.0 ml of internal standard
23 minutes. The test is not valid unless the resolution between solution and dilute to 50.0 ml with a 50 per cent v/v solution of
the peaks corresponding to prednisone and prednisolone is methanol and mix. Filter through a 5 µm filter and discard the
at least 2.7. If necessary, adjust the concentration of acetonitrile first 20 ml of the filtrate.
in mobile phase A. Reference solution. Weigh accurately a suitable quantity of
prednisone RS and dissolve in a 50 per cent v/v solution of
Inject separately methanol as a blank, the test solution and
methanol to obtain a solution having a concentration of about
reference solution (b). In the chromatogram obtained with the
0.2 mg per ml. To 5.0 ml of this solution add 5.0 ml of internal
test solution: the area of any peak other than the principal
standard solution and dilute to 50.0 ml with a 50 per cent v/v
peak is not greater than 0.25 times the area of the principal
solution of methanol.
peak in the chromatogram obtained with reference solution
(b) (0.25 per cent); the sum of the areas of all the peaks, apart Chromatographic system
from the principal peak, is not greater than 0.75 times the area – a stainless steel column 25 cm x 4 mm, packed with
of the principal peak in the chromatogram obtained with octadecylsilane chemically bonded to porous silica
reference solution (b) (0.75 per cent). Ignore any peak due to particles (3 to 10 µm),

975
PRIMAQUINE PHOSPHATE IP 2007

– mobile phase: a suitable filtered mixture of 688 volumes 310 nm, the resulting solution shows absorption maxima at
of water, 250 volumes of peroxide-free tetrahydrofuran about 225 nm, 265 nm and 282 nm; absorbances at the maxima
and 62 volumes of methanol such that at a flow rate of are 0.74 to 0.77, 0.50 to 0.53 and 0.50 to 0.52, respectively.
1 ml per minute the retention times of prednisone and
C. Determine by thin-layer chromatography (2.4.17), coating
acetanilide are about 8 and 6 minutes respectively,
the plate with silica gel F254.
– spectrophotometer set at 254 nm,
– a 10 µl loop injector. Mobile phase. A mixture of 60 volumes of chloroform,
40 volumes of methanol and 1 volume of strong ammonia
Inject the reference solution. Adjust the operating parameters
solution.
such that the peak obtained is about 50 per cent of the full
scale. The relative standard deviation for replicate injections Test solution. Dissolve 0.2 g in 5 ml of water, dilute to 10 ml
is not more than 2.0 per cent and the resolution between with methanol and dilute 1 volume of the resulting solution to
prednisone and the internal standard is not less than 3.0. 10 volumes with methanol (50 per cent).
Inject alternately the test solution and the reference solution. Reference solution. Dissolve 20 mg of primaquine phosphate
Calculate the content of C21H26O5 in the tablets. RS in 5 ml of water, dilute to 10 ml with methanol.
Storage. Store protected from light. Apply to the plate 5 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
Primaquine Phosphate with the reference solution.

CH3 D. Dissolve 50 mg in 5 ml of water, add 2 ml of 2 M sodium


hydroxide and shake with two quantities, each of 5 ml, of
NH2
HN chloroform. The aqueous layer, acidified by the addition of
N nitric acid, gives reaction B of phosphates (2.3.1).
, 2H3PO4
Tests
H3CO
pH (2.4.24). 2.5 to 3.5, determined in a 1.0 per cent w/v solution.
C15H21N3O,2H3PO4 Mol. Wt. 455.3 Related substances. Determine by liquid chromatography
Primaquine Phosphate is (RS)-8-(4-amino-1- (2.4.14).
methylbutylamino)-6-methoxyquinoline diphosphate. Test solution. Add 0.2 ml of strong ammonia solution to 1 ml
Primaquine Phosphate contains not less than 98.5 per cent of a 1.0 per cent w/v solution of the substance under
and not more than 101.5 per cent of C15H21N3O,2H3PO4, examination, shake with 10 ml of the mobile phase and use the
calculated on the dried basis. clear, lower layer.
Description. An orange, crystalline powder; odourless or Reference solution (a). Dilute 3 ml of the test solution to
almost odourless. 100 ml with the mobile phase.
Reference solution (b). Dilute 1 ml of the test solution to 10 ml
Identification
with the mobile phase and further dilute 1 ml of the resulting
Test A may be omitted if tests B, C and D are carried out. Tests solution to 50 ml with the same solvent.
B and C may be omitted if tests A and D are carried out. Reference solution (c). Add 0.2 ml of strong ammonia solution
A. Determine by infrared absorption spectrophotometry (2.4.6). to 1 ml of a 1.0 per cent w/v solution of the primaquine
Compare the spectrum with that obtained with primaquine phosphate RS, shake with 10 ml of the mobile phase and use
phosphate RS or with the reference spectrum of primaquine the clear, lower layer.
phosphate. Chromatographic system
B. When examined in the range 300 nm to 450 nm (2.4.7), a – a stainless steel column 20 cm x 4.6 mm, packed with
0.015 per cent w/v solution in 0.01 M hydrochloric acid shows porous silica particles (10 µm),
absorption maxima at about 332 nm and 415 nm; absorbance – mobile phase: a mixture of 45 volumes of chloroform,
at about 332 nm, about 0.68 to 0.78 and at about 415 nm, 0.41 to 45 volumes of hexane, 10 volumes of methanol and
0.53. Dilute 5 ml of the solution to 50 ml with 0.01 M 0.1 volume of strong ammonia solution,
hydrochloric acid. When examined in the range 215 nm to – flow rate. 3 ml per minute,

976
IP 2007 PROBENECID

– spectrophotometer set at 261 nm, Tests


– a 20 µl loop injector.
Uniformity of content. Comply with the tests stated under
The test is not valid unless in the chromatogram obtained Tablets.
with reference solution (c) there is a peak just before the
principal peak with an area of about 6 per cent of that of the Transfer one tablet into a suitable container, add 5 ml of
principal peak and the resolution between these peaks is not hydrochloric acid, disperse the tablet in about 25 g of crushed
less than 2.0. ice and add sufficient water to produce 50.0 ml. Carry out the
nitrite titration (2.3.31), using 0.01 M sodium nitrite as titrant.
Inject each solution and record the chromatograms for at least Carry out a blank titration.
twice the retention time of primaquine. The sum of the areas of
any secondary peaks in the chromatogram obtained with the 1 ml of 0.01 M sodium nitrite is equivalent to 0.002594 g of
test solution is not greater than the area of the principal peak C15H21N3O.
in the chromatogram obtained with reference solution (a). Other tests. Comply with the tests stated under Tablets.
Ignore any peak the area of which is less than that of the
Assay. Weigh and powder 20 tablets. Weigh accurately a
principal peak in the chromatogram obtained with reference
quantity of the powder containing about 0.15 g of primaquine,
solution (b).
dissolve in 20 ml of water, add 5 ml of 2 M sodium hydroxide
Loss on drying (2.4.19). Not more than 0.5 per cent, determined and extract with four quantities, each of 25 ml, of chloroform.
on 1.0 g by drying in an oven at 105º. Combine the chloroform extracts and evaporate to a volume
Assay. Weigh accurately about 0.2 g, dissolve in 40 ml of of about 10 ml. Add 40 ml of anhydrous glacial acetic acid,
anhydrous glacial acetic acid with gentle heating. Titrate Titrate with 0.1 M perchloric acid, determining the end point
with 0.1 M perchloric acid, determining the end point potentiometrically (2.4.25). Carry out a blank titration.
potentiometrically (2.4.25). Carry out a blank titration. 1 ml of 0.1 M perchloric acid is equivalent to 0.01297 g of
1 ml of 0.1 M perchloric acid is equivalent to 0.02277 g of C15H21N3O.
C15H21N3O,2H3PO4. Storage. Store protected from moisture.
Storage. Store protected from light and moisture. Labelling. The label states the strength in terms of the
equivalent amount of primaquine.

Primaquine Tablets
Primaquine Phosphate Tablets Probenecid
Primaquine Tablets contain not less than 90.0 per cent and not
more than 110.0 per cent of the stated amount of primaquine, H3C
H 3C COOH
C15H21N3O. The tablets are coated.
N
Identification S
O
A. Dissolve a quantity of the powdered tablets containing O
60 mg of primaquine in a mixture of 10 ml of water and 2 ml of C13H19NO4S Mol. Wt. 285.5
2 M sodium hydroxide and extract with two quantities, each
of 20 ml, of chloroform. Wash the chloroform extracts with Probenecid is 4-(dipropylsulphamoyl)benzoic acid.
water, dry over anhydrous sodium sulphate, evaporate to Probenecid contains not less than 99.0 per cent and not more
dryness and dissolve the residue in 2 ml of chloroform. The than 101.0 per cent of C13H19NO4S, calculated on the dried
residue complies with the following test. basis.
Determine by infrared absorption spectrophotometry (2.4.6). Description. A white or almost white, crystalline powder.
Compare the spectrum with that obtained with primaquine
phosphate RS or with the reference spectrum of primaquine. Identification

B. Extract a quantity of the powdered tablets containing 25 mg Test A may be omitted if tests B and C are carried out. Tests B
of primaquine with 10 ml of water and filter. To 2 ml of the and C may be omitted if test A is carried out.
filtrate add 3 ml of water and 1 ml of a 5 per cent w/v solution A. Determine by infrared absorption spectrophotometry (2.4.6).
of ceric ammonium sulphate in 2 M nitric acid; a deep violet Compare the spectrum with that obtained with probenecid RS
colour is produced immediately. or with the reference spectrum of probenecid.

977
PROBENECID TABLETS IP 2007

B. When examined in the range 210 nm to 360 nm (2.4.7), a Probenecid Tablets


0.001 per cent w/v solution in a mixture of 9 volumes of ethanol
(95 per cent) and 1 volume of 0.1 M hydrochloric acid shows Probenecid Tablets contain not less than 95.0 per cent and
absorption maxima at about 223 nm and 248 nm; absorbance not more than 105.0 per cent of the stated amount of
at about 248 nm is about 0.33. probenecid, C13H19NO4S.
C. Dissolve 0.2 g in about 0.6 ml of 2 M ammonia and add 3 ml Identification
of 1.7 per cent w/v solution of silver nitrate; a white precipitate
is produced which is soluble in an excess of dilute ammonia A.Triturate a quantity of the powdered tablets containing 0.5
solution. g of Probenecid with ethanol (95 per cent), filter and
concentrate the filtrate by evaporation on a water-bath. Cool,
Tests filter and recrystallise the residue from ethanol (50 per cent).
The residue complies with the following test.
Appearance of solution. A 10.0 per cent w/v solution in 2 M
sodium hydroxide is clear (2.4.1), and not more intensely Determine by infrared absorption spectrophotometry (2.4.6).
coloured than reference solution YS6 (2.4.1). Compare the spectrum with that obtained with probenecid RS
or with the reference spectrum of probenecid.
Acidity. Add 2.0 g to 100 ml of water, heat on a water-bath for
30 minutes, cool, filter and dilute with water to 100.0 ml. Titrate B. When examined in the range 210 nm to 360 nm (2.4.7), the
50.0 ml of the solution with 0.1 M sodium hydroxide using final solution obtained in the Assay shows absorption maxima
phenolphthalein solution as indicator. Not more than 0.5 ml at about 225 nm and 248 nm.
of 0.1 M sodium hydroxide is required to change the colour of C. The residue obtained in test A melts at about 199º (2.4.21).
the solution.
Tests
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 55 volumes of toluene, 20 volumes
of di-isopropyl ether, 15 volumes of chloroform and Mobile phase. A mixture of 55 volumes of toluene, 20 volumes
10 volumes of glacial acetic acid. of di-isopropyl ether, 15 volumes of chloroform and
10 volumes of glacial acetic acid.
Test solution. Dissolve 0.1 g of the substance under
examination in 10 ml of acetone. Test solution. Extract a quantity of the powdered tablets
containing 0.2 g of Probenecid with 20 ml of acetone, filter and
Reference solution. A 0.005 per cent w/v solution of the
use the filtrate.
substance under examination in acetone.
Reference solution. Dilute 1 ml of the test solution to 200 ml
Apply to the plate 20 µl of each solution. After development, with acetone.
dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the Apply to the plate 20 µl of each solution. After development,
test solution is not more intense than the spot in the dry the plate in air and examine in ultraviolet light at 254 nm.
chromatogram obtained with the reference solution. Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the
Heavy metals (2.3.13).1.0 g complies with the limit test for chromatogram obtained with the reference solution.
heavy metals, Method B (20 ppm).
Dissolution (2.5.2).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Apparatus. No 1
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Medium. 900 ml of phosphate buffer pH 7.6
on 1.0 g by drying in an oven at 105º.
Speed and time. 50 rpm and 30 minutes.
Assay. Weigh accurately about 0.5 g and dissolve in 100 ml of
Withdraw a suitable volume of the medium and filter, Dilute
ethanol (95 per cent), shaking well and heating gently if
4.0 ml of the filtrate to 100.0 ml with 0.1 M sodium hydroxide.
necessary. Cool and titrate with 0.1 M sodium hydroxide,
Measure the absorbance of the resulting solution at the
using bromothymol blue solution as indicator. Carry out a
maximum at about 244 nm (2.4.7). Similarly measure the
blank titration.
absorbance of a solution of a known concentration of
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02854 g of probenecid RS. Calculate the content of C13H19O4S.
C13H19NO4S.
D. Not less than 80 per cent of the stated amount of
Storage. Store protected from moisture. C13H19NO3S.

978
IP 2007 PROCAINAMIDE INJECTION

Other tests. Comply with the tests stated under Tablets. Tests
Assay. Weigh and powder 20 tablets. Weigh accurately a Appearance of solution. A 10.0 per cent w/v solution in carbon
quantity of the powder containing about 0.2 g of Probencid, dioxide-free water is clear (2.4.1), and not more intensely
add 200 ml of ethanol (95 per cent) and 5 ml of 1 M coloured than reference solution BS6 (2.4.1).
hydrochloric acid, heat on a water-bath at 70º for 30 minutes,
shaking occasionally. Cool, add sufficient ethanol (95 per pH (2.4.24). 5.6 to 6.3, determined in a 10.0 per cent w/v solution.
cent) to produce 250.0 ml and filter. To 5.0 ml of the filtrate add Related substances. Determine by thin-layer chromatography
5 ml of 0.1 M hydrochloric acid, dilute to 250.0 ml with ethanol (2.4.17), coating the plate with silica gel GF254.
(95 per cent) and measure the absorbance of the resulting Mobile phase. A mixture of 60 volumes of 1-butanol,
solution at the maximum at about 248 nm (2.4.7). Calculate the 30 volumes of water and 15 volumes of glacial acetic acid.
content of C13H19NO4S taking 332 as specific absorbance at
248 nm. Test solution. Dissolve 0.1 g of the substance under
examination in 10 ml of ethanol (95 per cent).
Storage. Store protected from moisture.
Reference solution. A 0.005 per cent w/v solution of the
substance under examination in ethanol (95 per cent).
Apply to the plate 5 µl of each solution. After development,
Procainamide Hydrochloride dry the plate in air and examine in ultraviolet light at 254 nm.
CH3 Any secondary spot in the chromatogram obtained with the
O test solution is not more intense than the spot in the
N CH3 chromatogram obtained with the reference solution.
N
H . HCl Heavy metals (2.3.13). 1.0 g complies with the limit test for
H2N heavy metals, Method B (20 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
C13H21N3O,HCl Mol. Wt. 271.8
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Procainamide Hydrochloride is 4-amino-N-[2-(diethylamino) on 1.0 g by drying in an oven at 105º.
ethyl)benzamide hydrochloride.
Assay. Weigh accurately about 0.25 g, dissolve in 75 ml of
Procainamide Hydrochloride contains not less than 98.0 per water and 10 ml of hydrochloric acid and carry out the nitrite
cent and not more than 101.0 per cent of C13H21N3O,HCl, titration (2.3.31).
calculated on the dried basis.
1 ml of 0.1 M sodium nitrite is equivalent to 0.02718 g of
Description. A white or very slightly yellow, crystalline C13H21N3O, HCl.
powder; hygroscopic.
Storage. Store protected from light.
Identification
Test A may be omitted if tests B, C and D are carried out. Tests
B, C and D may be omitted if test A is carried out.
Procainamide Injection
A. Determine by infrared absorption spectrophotometry (2.4.6). Procainamide Hydrochloride Injection
Compare the spectrum with that obtained with procainamide Procainamide Injection is a sterile solution of Procainamide
hydrochloride RS or with the reference spectrum of Hydrochloride in Water for Injections. It may contain Sodium
procainamide hydrochloride. Metabisulphite as a stabilising agent.
B. When examined in the range 230 nm to 360 nm (2.4.7), a Procainamide Injection contains not less than 95.0 per cent
0.001 per cent w/v solution in 0.1 M sodium hydroxide shows and not more than 105.0 per cent of the stated amount of
an absorption maximum at about 273 nm; absorbance at about procainamide hydrochloride, C13H21N3O, HCl.
273 nm, 0.58 to 0.61.
C. Dilute 1 ml of a solution, prepared by dissolving 2.5 g in Identification
sufficient carbon dioxide-free water to produce 25 ml, to 2 ml A. Dilute a volume containing 0.25 g of Procainamide
with water. 1 ml of this solution gives the reaction of primary Hydrochloride to 25 ml with water, make alkaline with 5 M
aromatic amines (2.3.1). sodium hydroxide and extract with two quantities, each of
D. A 2 per cent w/v solution gives reaction A of chlorides 5 ml, of chloroform. Filter the combined extracts through
(2.3.1). anhydrous sodium sulphate, evaporate the filtrate to dryness

979
PROCAINAMIDE TABLETS IP 2007

using a rotatory evaporator and dissolve the residue in 5 ml of quantities, each of 5 ml, of chloroform. Filter the combined
chloroform. extracts through anhydrous sodium sulphate, evaporate the
Determine by infrared absorption spectrophotometry (2.4.6). filtrate to dryness using a rotatory evaporator and dissolve
Compare the spectrum with that obtained with procainamide the residue in 5 ml of chloroform.
hydrochloride RS or with the reference spectrum of Determine by infrared absorption spectrophotometry (2.4.6).
procainamide. Compare the spectrum with that obtained with procainamide
B. Dilute a suitable volume of the injection with water to hydrochloride RS or with the reference spectrum of
produce a solution containing 0.0005 per cent w/v of procainamide.
Procainamide Hydrochloride. Absorbance of the resulting B. Triturate a quantity of the powdered tablets containing 1 g
solution at about 280 nm, about 0.30 (2.4.7). of Procainamide Hydrochloride with 10 ml of water and filter.
C. Gives the reactions of chlorides (2.3.1). The filtrate gives the reactions of chlorides (2.3.1).

Tests Tests

pH (2.4.24). 4.0 to 5.5. Related substances. Determine by thin-layer chromatography


(2.4.17), coating the plate with silica gel GF254.
Related substances. Determine by thin-layer chromatography
Mobile phase. A mixture of 60 volumes of 1-butanol,
(2.4.17), coating the plate with silica gel GF254.
30 volumes of water and 15 volumes of glacial acetic acid.
Mobile phase. A mixture of 60 volumes of 1-butanol,
Test solution. Shake a quantity of the powdered tablets
30 volumes of water and 15 volumes of glacial acetic acid.
containing 0.4 g of Procainamide Hydrochloride with 20 ml of
Test solution. Dilute a volume of the injection containing 0.1 g methanol (90 per cent) for 15 minutes and filter.
of Procainamide Hydrochloride to 5 ml with methanol.
Reference solution. Dilute 1 volume of the test solution to
Reference solution. Dilute 1 volume of the test solution to 100 volumes with methanol.
100 volumes with methanol.
Apply to the plate 5 µl of each solution. After development,
Apply to the plate 5 µl of each solution. After development, dry the plate in air and examine in ultraviolet light at 254 nm.
dry the plate in air and examine in ultraviolet light at 254 nm. Any secondary spot in the chromatogram obtained with the
Any secondary spot in the chromatogram obtained with the test solution is not more intense than the spot in the
test solution is not more intense than the spot in the chromatogram obtained with the reference solution.
chromatogram obtained with the reference solution.
Other tests. Comply with the tests stated under Tablets.
Other tests. Complies with the tests stated under Parenteral
Assay. Weigh and powder 20 tablets. Weigh accurately a
Preparations (Injections).
quantity of the powder containing about 0.25 g of Procainamide
Assay. To a volume containing about 0.25 g of Procainamide Hydrochloride, add 100 ml of 6 M hydrochloric acid, shake
Hydrochloride add 45 ml of 6 M hydrochloric acid and boil for 15 minutes and boil for 1 minute. Carry out the nitrite
for 1 minute. Carry out the nitrite titration (2.3.31). titration (2.3.31).
1 ml of 0.1 M sodium nitrite is equivalent to 0.02718 g of 1 ml of 0.1 M sodium nitrite is equivalent to 0.02718 g of
C13H21N3O, HCl. C13H21N3O, HCl.
Storage. Store protected from light. Storage. Store protected from light and moisture.

Procainamide Tablets Procaine Hydrochloride


Procainamide Hydrochloride Tablets
CH3
Procainamide Tablets contain not less than 95.0 per cent and O
not more than 105.0 per cent of the stated amount of N CH3 , HCl
O
procainamide hydrochloride, C13H21N3O,HCl.

Identification H2N

A. Shake a quantity of the powdered tablets containing 0.25 g C13H20N2O2,HCl Mol. Wt. 272.8
of Procainamide Hydrochloride with 25 ml of water, make Procaine Hydrochloride is 2-(diethylamino)ethyl 4-
alkaline with 5 M sodium hydroxide and extract with two aminobenzoate hydrochloride.

980
IP 2007 PROCAINE AND ADRENALINE INJECTION

Procaine Hydrochloride contains not less than 99.0 per cent Sulphated ash (2.3.18). Not more than 0.1 per cent.
and not more than 101.0 per cent of C13H20N2O2,HCl, calculated Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on the dried basis. on 1.0 g by drying in an oven at 105º.
Description. Colourless crystals or a white, crystalline powder;
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of 2 M
odourless.
hydrochloric acid, add 3 g of potassium bromide, cool in ice
Identification and titrate slowly with 0.1 M sodium nitrite, stirring constantly.
Determine the end point potentiometrically (2.4.25).
Test A may be omitted if tests B, C, D and E are carried out. 1 ml of 0.1 M sodium nitrite is equivalent to 0.02728 g of
Tests B, C and D may be omitted if tests A and E are carried C13H20N2O2, HCl.
out.
Storage. Store protected from light.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with procaine
hydrochloride RS or with the reference spectrum of procaine
hydrochloride. Procaine and Adrenaline Injection
B. To 0.2 ml of a 5 per cent w/v solution add 2 ml of water and Procaine Hydrochloride and Adrenaline Bitartrate
0.5 ml of 1 M sulphuric acid, shake and add 1 ml of a 0.1 per
Injection; Procaine Hydrochloride and Epinephrine
cent w/v solution of potassium permanganate; the colour is
immediately discharged.
Bitartrate Injection
C. To about 5 mg add 0.5 ml of fuming nitric acid, evaporate to Procaine and Adrenaline Injection is a sterile solution of
dryness on a water-bath, cool, dissolve the residue in 5 ml of Procaine Hydrochloride and Adrenaline Bitartrate in Water
acetone and add 1 ml of 0.1 M ethanolic potassium for Injections.
hydroxide; only a brownish red colour develops. Procaine and Adrenaline Injection contains not less than
D. Dilute 1 ml of a 5 per cent w/v solution to 100 ml with water; 1.9 per cent and not more than 2.1 per cent w/v of procaine
2 ml of the solution gives the reaction of primary aromatic hydrochloride, C13H20N2O2, HCl and the equivalent of not less
amines (2.3.1). than 0.00175 per cent and not more than 0.00225 per cent w/v
of adrenaline, C9H13NO3.
E. Gives reaction A of chlorides (2.3.1).
Description. A clear, colourless solution.
Tests
Identification
Appearance of solution. A 5.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and colourless (2.4.1). A. To 5 ml add 5 ml of water and 10 ml of picric acid solution,
shake gently and set aside for 1 hour; the crystalline precipitate,
pH (2.4.24). 5.0 to 6.5, determined in a 2.0 per cent w/v solution.
after washing with water and drying at 100º, melts at about
Related substances. Determine by thin-layer chromatography 134º (2.4.21).
(2.4.17), coating the plate with silica gel GF254.
B. To 5 ml add 1 ml of hydrochloric acid, cool to 0º, add 5 ml of
Mobile phase. A mixture of 80 volumes of dibutyl ether, sodium nitrite solution and pour the mixture into 2 ml of
16 volumes of n-hexane and 4 volumes of glacial acetic acid. 2-naphthol solution containing 1 g of sodium acetate; an
Test solution. A 10 per cent w/v solution of the substance orange-red colour is produced.
under examination in water. C. To 10 ml add 4 ml of disodium hydrogen phosphate solution
Reference solution. A 0.005 per cent w/v solution of and sufficient 0.1 M iodine to produce a distinct brown colour.
4-aminobenzoic acid in water. Add 0.1 M sodium thiosulphate to remove the excess of
iodine; a pink colour is produced.
Apply to the plate 5 µl of each solution. After development,
dry the plate at 105º for 10 minutes and examine in ultraviolet Tests
light at 254 nm. Any secondary spot in the chromatogram
obtained with the test solution is not more intense than the pH (2.4.24). 3.0 to 5.5.
spot in the chromatogram obtained with the reference solution. Other tests. Complies with the tests stated under Parenteral
The principal spot remains on the line of application. Preparations (Injections).
Heavy metals (2.3.13). 2.0 g complies with the limit test for Assay. For procaine hydrochloride — To 10.0 ml of the
heavy metals, Method A (10 ppm). injection add 0.5 g of sodium carbonate and extract with three

981
PROCAINE PENICILLIN IP 2007

quantities, each of 20 ml, of a mixture of 1 volume of 2-propanol penicillin RS or with the reference spectrum of procaine
and 3 volumes of chloroform until complete extraction of penicillin.
procaine is effected. Shake the combined extracts with 5 ml of B. In the Assay, the principal peak in the chromatogram
water, wash the water with the solvent mixture and add the obtained with the test solution corresponds to the peak in the
washing to the combined extracts. Shake the combined extracts chromatogram obtained with the reference solution (a).
and washings with 10.0 ml of 0.1 M hydrochloric acid,
separate the acid layer, wash the combined extracts and C. A turbid solution of 0.1 g in 2 ml of 2 M hydrochloric acid
washings with 5 ml of water, add the aqueous extract to the gives the reaction of primary aromatic amines (2.3.1).
separated acid layer and titrate with 0.1 M sodium hydroxide, Tests
using methyl red-methylene blue solution as indicator.
pH (2.4.24). 5.0 to 7.5, determined in a solution prepared by
1 ml of 0.1 M hydrochloric acid is equivalent to 0.02728 g of
shaking 50 mg in sufficient carbon dioxide-free water to
C13H20N2O2,HCl.
produce 15 ml until dissolution is complete.
For adrenaline — To 10.0 ml of the injection add 20 mg of
Specific optical rotation (2.4.22). +165º to +180º, determined
sodium metabisulphite, 0.1 ml of ferrous sulphate-citrate
in a solution prepared by dissolving 0.25 g in sufficient of a
solution, 1 ml of glycine buffer solution and mix. Allow to
mixture of 3 volumes of acetone and 2 volumes of water to
stand for 10 minutes, extract with 10 ml of ether, allow to
produce 25 ml.
separate, reject the ether and measure the absorbance of a
4-cm layer of the solution at about 540 nm (2.4.7). Calculate Water (2.3.43). 2.8 to 4.2 per cent, determined on 0.5 g.
the content of adrenaline, C9H13NO3, from a reference curve Assay. Determine the contents of benzyl penicillin and
prepared by treating suitable aliquots of a solution of procaine by liquid chromatography, (2.4.14).
adrenaline bitartrate RS in the same manner.
Test solution. Weigh accurately 70 mg of the substance under
1 mg of adrenaline bitartrate is equivalent to 0.0005497 g of examination and dissolve in 30 ml of the mobile phase with the
C9H13NO3. aid of ultrasound for about 2 minutes. Dilute to 50.0 ml with
Storage. Store protected from light. the mobile phase.
Reference solution (a). Weigh accurately about 56 mg of
Labelling. The label states the strength as “Procaine
benzyl penicillin potassium RS and 40 mg of procaine
Hydrochloride, 2 per cent w/v; Adrenaline, 0.002 per cent
hydrochloride RS and dissolve in 25 ml of the mobile phase
w/v”.
with the aid of ultrasound for about 2 minutes. Dilute to
50.0 ml with the mobile phase.
Reference solution (b). Mix 1 volume of a 0.24 per cent w/v
Procaine Penicillin solution of phenoxymethylpenicillin potassium RS in the
H COOH
mobile phase with 3 volumes of reference solution (a).
CH3 O
O N CH3 Chromatographic system
H
N CH3 N
O S CH3
, H2O – a stainless steel column 30 cm x 4 mm, packed with
, H H
H2N
O octadecylsilyl silica gel (10 µm),
– mobile phase: a mixture of 50 volumes of a solution
C13H20N2O2,C16H18N2O4S,H2O Mol. Wt. 588.7 prepared by mixing 14 g of potassium dihydrogen
Procaine Penicillin is 2-diethylaminoethyl 4-aminobenzoate phosphate in 6.5 g of tetrabutylammonium hydroxide
(6R)-6-(2-phenylacetamido)penicillanate monohydrate. (40 per cent) and adjusting the pH to 7.0 with 1 M
potassium hydroxide or dilute phosphoric acid,
Procaine Penicillin contains not less than 51 per cent and not 25 volumes of acetonitrile and 25 volumes of water,
more than 59.6 per cent of penicillin G, calculated as – flow rate. 1 ml per minute,
C16H18N2O4S, and not less than 37.5 per cent and not more – spectrophotometer set at 235 nm,
than 43.0 per cent of procaine, C13H20N2O2, both calculated on – a 20 µl loop injector.
the anhydrous basis.
Inject reference solution (b). The resolution between benzyl
Description. A white, crystalline powder. penicillin potassium and phenoxymethylpenicillin potassium
is not less than 2. The relative retention time of procaine with
Identification reference to benzyl penicillin is about 2.2.
A. Determine by infrared absorption spectrophotometry (2.4.6). Inject the test solution and reference solution (a). Record the
Compare the spectrum with that obtained with procaine peak responses of the main peaks of benzyl penicillin and

982
IP 2007 FORTIFIED PROCAINE PENICILLIN INJECTION

procaine. Calculate the content of benzyl penicillin, The contents of the sealed container comply with the tests
C16H18N2O4S and the content of procaine, C13H20N2O2 using stated under Parenteral Preparations (Powders for
the factor of 0.866 for converting the content of procaine Injection) and with the following requirements.
hydrochloride to that of procaine.
Procaine Penicillin intended for use in the manufacture of
Identification
Parenteral Preparations without a further procedure for the A. Dissolve 10 mg in 10 ml of water and add 0.5 ml of neutral
removal of bacterial endotoxins complies with the following red solution. Add sufficient 0.01 M sodium hydroxide to give
additional requirement. a permanent orange colour and then add 1 ml of penicillinase
Bacterial endotoxins (2.2.3). Not more than 0.10 Endotoxin solution; a red colour is produced rapidly.
Unit per mg. B. In the Assay, the principal peak in the chromatogram
Procaine Penicillin intended for use in the manufacture of obtained with the test solution corresponds to the peak in the
Parenteral Preparations without a further sterilisation chromatogram obtained with the reference solution.
procedure complies with the following additional C. Give the reaction of primary aromatic amines (2.3.1),
requirement. producing a bright, orange-red precipitate.
Sterility. Complies with the test for sterility (2.2.11).
Tests
Storage. Store protected from moisture. If the material is
intended for use in the manufacture of parenteral preparations, Water (2.3.43). Not more than 3.5 per cent, determined on
the container should be sterile and sealed so as to exclude 0.5 g.
micro-organisms.
Assay. Determine by liquid chromatography (2.4.14).
Labelling. The label states (1) where applicable, that the
Test solution. Weigh accurately a quantity of the mixed
contents are free from bacterial endotoxins; (2) where
contents of 10 containers equivalent to about 70 mg of
applicable, that the contents are sterile.
procaine penicillin and dissolve in 30 ml of the mobile phase
with the aid of ultrasound for about 2 minutes. Dilute to
50.0 ml with the mobile phase.
Fortified Procaine Penicillin Injection Reference solution (a). Weigh accurately about 56 mg of
Procaine Penicillin with Benzylpenicillin Injection benzylpenicillin potassium RS and 40 mg of procaine
hydrochloride RS and dissolve in 25 ml of the mobile phase
Fortified Procaine Penicillin Injection is a sterile mixture of five with the aid of ultrasound for about 2 minutes. Dilute to
parts of Procaine Penicillin and one part of Benzylpenicillin 50.0 ml with the mobile phase.
Potassium or Benzylpenicillin Sodium, together with suitable
dispersing and buffering agents. It is filled in a sealed container. Reference solution (b). Mix 1 volume of a 0.24 per cent w/v
solution of phenoxymethylpenicillin potassium RS in the
The injection is constituted by suspending the contents of mobile phase with 3 volumes of reference solution (a).
the sealed container in the requisite amount of sterile Water
for Injections, immediately before use. Chromatographic system
– a stainless steel column 30 cm x 4 mm, packed with
Storage. The constituted injection should be used within
octadecylsilyl silica gel (10 µm),
24 hours (4 days if a buffering agent is present) of preparation
– mobile phase: a mixture of 50 volumes of a solution
when stored at a temperature not exceeding 20º or within
prepared by mixing 14 g of potassium dihydrogen
7 days (14 days if a buffering agent is present) when stored at
phosphate in 6.5 g of tetrabutyl ammonium hydroxide
a temperature between 2º and 8º.
(40 per cent) and adjusting the pH to 7.0 with 1 M
Fortified Procaine Penicillin Injection contains a quantity of potassium hydroxide or dilute phosphoric acid,
total penicillins calculated as C16H17N2NaO4S and equivalent 25 volumes of acetonitrile and 25 volumes of water,
to not less than 60.0 per cent and not more than 74.0 per cent – flow rate. 1 ml per minute,
of the stated amounts of procaine penicillin and benzylpenicillin – spectrophotometer set at 235 nm,
potassium or benzylpenicillin sodium and a quantity of – a 20 µl loop injector.
procaine, C13H20N2O2, equivalent to not less than 36.0 per cent
Inject reference solution (b). The resolution between
and not more than 44.0 per cent of the stated amount of
benzylpenicillin potassium and phenoxymethylpenicillin
procaine penicillin.
potassium is not less than 2. The relative retention time of
Description. A white or almost white powder. procaine with reference to benzylpenicillin is about 2.2.

983
PROCHLORPERAZINE MALEATE IP 2007

Inject the test solution and reference solution (a). Record the C. Complies with the test for identification of phenothiazines
peak responses of the main peaks of benzyl penicillin and (2.3.3).
procaine. Calculate the total penicillin content as Test solution. A solution containing 0.1 per cent w/v of the
benzylpenicillin, C16H18N2O4S and the content of procaine, substance under examination in a mixture of equal volumes of
C13H20N2O2 using the factor of 0.866 for converting the content methanol and chloroform.
of procaine hydrochloride to that of procaine.
Reference solution. A 0.1 per cent w/v solution of
Labelling. The label states (1) the quantities of Procaine prochlorperazine maleate RS in the same solvent mixture
Penicillin and Benzylpenicillin Potassium or Benzylpenicillin
Sodium contained in it; (2) the names of any added dispersing Apply 4 µl of each solution.
and buffering agents; (3) “for intramuscular injection only”. D. Triturate 0.2 g with a mixture of 3 ml of water and 1 ml of 10
M sodium hydroxide and shake with three quantities, each of
5 ml, of ether. To 0.1 ml of the aqueous layer add a solution of
Prochlorperazine Maleate 10 mg of resorcinol in 3 ml of sulphuric acid and heat in a
water-bath for 15 minutes; no colour develops. To the
remainder of the aqueous layer add 2 ml of bromine solution,
heat in a water-bath for 15 minutes, then heat to boiling and
COOH cool. To 0.1 ml of the solution add a solution of 10 mg of
N N , resorcinol in 3 ml of sulphuric acid and heat in a water-bath
S N for 15 minutes; a blue colour develops.
CH3 COOH
2 Tests
Cl
pH (2.4.24). 3.0 to 4.0, determined in a freshly prepared saturated
C20H24ClN3S,2C4H4O4 Mol. Wt. 606.1
solution.
Prochlorperazine Maleate is 2-chloro-10-[3-(4-
Related substances. Complies with the test for related
methylpiperazin-1-yl)propyl]phenothiazine di(hydrogen
substances in phenothiazines (2.3.5), using mobile phase A.
maleate).
Prepare the test solution immediately before use.
Prochlorperazine Maleate contains not less than 98.0 per cent Sulphated ash (2.3.18). Not more than 0.1 per cent.
and not more than 101.0 per cent of C20H24ClN3S, 2C4H4O4,
calculated on the dried basis. Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105º.
Description. A white or pale yellow, crystalline powder;
practically odourless. Assay. Weigh accurately about 0.2 g, in powder, dissolve in
50 ml of anhydrous glacial acetic acid, heating gently on a
Identification water-bath, allow to cool. Titrate with 0.1 M perchloric acid,
determining the end-point potentiometrically (2.4.25). Carry
Test A may be omitted if tests B, C and D are carried out. Tests
out a blank titration.
B, C and D may be omitted if test A is carried out.
1 ml of 0.1 M perchloric acid is equivalent to 0.03031 g of
A.To 20 mg add 5 ml of water and 1 ml of 1 M sodium hydoxide,
C20H24ClN3S, 2C4H4O4.
shake and extract with 10 ml of ether. Wash the ether extract
with 5 ml of water, dry with anhydrous sodium sulphate, Storage. Store protected from light and moisture.
evaporate to dryness and dissolve the residue in 0.2 ml of
chloroform.
Determine by infrared absorption spectrophotometry (2.4.6). Prochlorperazine Tablets
Compare the spectrum with that obtained with
Prochlorperazine Maleate Tablets
prochlorperazine maleate RS or with the reference spectrum
of prochlorperazine. Prochlorperazine Tablets contain not less than 92.5 per cent
and not more than 107.5 per cent of the stated amount of
B. When examined in the range 230 nm to 360 nm (2.4.7), a
prochlorperazine maleate, C20H24ClN3S, 2C4H4O4.
0.001 per cent w/v solution in ethanol (95 per cent) containing
0.01 per cent v/v of strong ammonia solution shows an Identification
absorption maximum at about 258 nm and a less well-defined
maximum at about 313 nm; absorbance at about 258 nm, about A.To a quantity of the powdered tablets containing 40 mg of
0.6. Prochlorperazine Maleate add 10 ml of water and 2 ml of 1 M

984
IP 2007 PROCHLORPERAZINE MESYLATE

sodium hydroxide, shake and extract with 15 ml of ether. Wash Weigh and powder 20 tablets. Weigh accurately a quantity of
the ether with 5 ml of water, dry with anhydrous sodium the powder containing about 25 mg of Prochlorperazine
sulphate, evaporate to dryness and dissolve the residue in Maleate and extract with three quantities, each of 10 ml, of
0.4 ml of chloroform. ethanol containing 1 per cent v/v of strong ammonia solution.
Determine by infrared absorption spectrophotometry (2.4.6). Filter the extracts and to the combined filtrates add sufficient
Compare the spectrum with that obtained with ethanol to produce 100.0 ml. Dilute 10.0 ml to 50.0 ml with
prochlorperazine maleate RS or with the reference spectrum ethanol, dilute 10.0 ml of this solution to 50.0 ml with ethanol
of prochlorperazine. and measure the absorbance of the resulting solution at the
maximum at about 258 nm (2.4.7). Calculate the content of
B. To a quantity of the powdered tablets containing 5 mg of C20H24ClN3S, 2C4H4O4 taking 620 as the specific absorbance at
Prochlorperazine Maleate add 5 ml of sulphuric acid and allow 258 nm.
to stand for 5 minutes; a red colour is produced.
Storage. Store protected from light and moisture.
C. Shake a quantity of the powdered tablets containing 0.2 g
of Prochlorperazine Maleate with 2 ml of water and 1 ml of 5 M
sodium hydroxide, mix and extract with three quantities, each
of 10 ml, of ether. Dry the combined extracts with anhydrous Prochlorperazine Mesylate
sodium sulphate, filter, evaporate the filtrate to dryness and
dissolve the residue in 10 ml of methanol and add a solution
of 0.15 g of picric acid in 10 ml of methanol. The precipitate,
after washing with a small quantity of methanol, melts at about N N
, 2CH3SO3H
255º, with decomposition (2.4.21). S N
CH3
Tests
Cl
Related substances. Comply with the test for related
substances in phenothiazines (2.3.5), using mobile phase A. C20H24ClN3S,2CH4SO3 Mol. Wt. 566.2
Prochlorperazine Mesylate is 2-chloro-10-[3-(4-
Prepare the following solutions freshly.
methylpiperazin-1-yl)propyl]phenothiazine
Test solution. Extract a quantity of the powdered tablets di(methanesulphonate).
containing 0.1 g of Prochlorperazine Maleate with 10 ml of
Prochlorperazine Mesylate contains not less than 98.0 per
methanol containing 0.5 per cent v/v of strong ammonia
cent and not more than 101.0 per cent of C20H24ClN3S,2CH4SO3,
solution and filter.
calculated on the dried basis.
Reference solution. Dilute 1 volume of the test solution to
Description. A white or almost white powder; odourless.
200 volumes with the same solvent.
Apply to the plate 20 µl of each solution. Identification
Uniformity of content. (For tablets containing 10 mg or less) A. Determine by infrared absorption spectrophotometry (2.4.6).
— Comply with the test stated under Tablets. Compare the spectrum with that obtained with
prochlorperazine mesylate RS or with the reference spectrum
Protect the solutions from light throughout the Assay. of prochlorperazine mesylate.
Crush one tablet and extract with three quantities, each of B. When examined in the range 230 nm to 360 nm (2.4.7), a
10 ml, of ethanol containing 1 per cent v/v of strong ammonia 0.001 per cent w/v solution in ethanol containing 0.01 per
solution. Filter the extracts and to the combined filtrates add cent v/v of strong ammonia solution shows an absorption
sufficient ethanol to produce 50.0 ml. Dilute 10.0 ml of this maximum at about 258 nm and a less well-defined maximum at
solution to 100.0 ml with ethanol. Dilute further with ethanol, about 313 nm; absorbance at about 258 nm, about 0.6.
if necessary, to give a final solution containing 10 µg of
Prochlorperazine Maleate per ml and measure the absorbance C. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand
of the resulting solution at the maximum at about 258 nm (2.4.7). for 5 minutes; a red colour is produced.
Calculate the content of C20H24ClN3S, 2C4H4O4 taking 620 as D. Mix 50 mg with 0.2 g of powdered sodium hydroxide, heat
the specific absorbance at 258 nm. to fusion and continue the heating for a few seconds longer.
Cool, add 0.5 ml of water and a slight excess of 2 M
Other tests. Comply with the tests stated under Tablets.
hydrochloric acid and warm; sulphur dioxide is evolved which
Assay. Protect the solutions from light throughout the Assay. turns moistened starch iodate paper blue.

985
PROCHLORPERAZINE INJECTION IP 2007

Tests B. To a volume containing 5 mg of Prochlorperazine Mesylate


add carefully 2 ml of sulphuric acid and allow to stand for
pH (2.4.24). 2.0 to 3.0, determined in a 2.0 per cent w/v solution. 5 minutes; a red colour is produced.
Related substances. Complies with the test for related Tests
substances in phenothiazines (2.3.5), using mobile phase A.
pH (2.4.24). 5.5 to 6.5.
Test solution. Dissolve the substance under examination in
Related substances. Carry out the test for related substances
methanol containing 0.5 per cent v/v of strong ammonia
in phenothiazines (2.3.5), using mobile phase A.
solution.
Test solution. Use the injection under examination.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Reference solution (a). Dilute 1 volume of the test solution to
Loss on drying (2.4.19). Not more than 1.0 per cent, determined 40 volumes with methanol containing 0.5 per cent v/v of strong
on 1.0 g by drying in an oven at 100º at a pressure not exceeding ammonia solution immediately before use.
0.7 kPa.
Reference solution (b). Dilute 1 volume of the test solution to
Assay. Weigh accurately about 0.5 g, dissolve in 10 ml of 200 volumes with the methanol-ammonia mixture immediately
water, add 5 ml of 1 M sodium hydroxide and extract with before use.
successive quantities of 50, 25, 25 and 25 ml of ether. Wash
the combined ether extracts with 5 ml of water, shake the Any secondary spot in the chromatogram obtained with the
washings with 5 ml of ether, add the ether to the combined test solution is not more intense than the spot in the
ether extracts and evaporate to dryness. Add 2 ml of ethanol, chromatogram obtained with reference solution (a) and not
evaporate to dryness. Titrate with 0.1 M perchloric acid, more than one such spot is more intense than the spot in the
using 1-naphtholbenzein solution as indicator. Carry out a chromatogram obtained with reference solution (b).
blank titration. Other tests. Complies with the tests stated under Parenteral
1 ml of 0.1 M perchloric acid is equivalent to 0.02831 g of Preparations (Injections).
C20H24ClN3S, 2CH4SO3. Assay. Protect the solutions from light throughout the Assay.
Storage. Store protected from light and moisture. To an accurately measured volume containing about 25 mg of
Prochlorperazine Mesylate add sufficient ethanol containing
0.01 per cent v/v of strong ammonia solution to produce
200.0 ml. Dilute 5.0 ml of this solution to 100.0 ml with the
Prochlorperazine Injection ammoniacal ethanol and measure the absorbance of the
resulting solution at the maximum at about 258 nm (2.4.7).
Prochlorperazine Mesylate Injection Calculate the content of C20H24ClN3S, 2CH4SO3 taking 635 as
Prochlorperazine Injection is a sterile solution of the specific absorbance at 258 nm.
Prochlorperazine Mesylate in Water for Injections free from Storage. Store protected from light.
dissolved air.
Prochlorperazine Injection contains not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
prochlorperazine mesylate, C20H24ClN3S,2CH4SO3. Procyclidine Hydrochloride
Identification
A. To a volume containing 0.1 g of Prochlorperazine Mesylate
add 20 ml of water and 2 ml of 10 M sodium hydroxide. Shake
and extract with 25 ml of ether. Wash the ether layer with two OH , HCl
quantities, each of 5 ml, of water, dry with anhydrous sodium N
sulphate and evaporate to dryness. Dissolve the residue in
1 ml of chloroform.
Determine by infrared absorption spectrophotometry (2.4.6).
C19H29NO,HCI Mol. wt. 323.9
Compare the spectrum with that obtained with
prochlorperazine mesylate RS treated in the same manner or Procyclidine Hydrochloride is (RS)-1-cyclohexyl-1-phenyl-3-
with the reference spectrum of prochlorperazine. pyrrolidin-1-ylpropan-1-ol hydrochloride.

986
IP 2007 PROCYCLIDINE TABLETS

Procyclidine Hydrochloride contains not less than 99.0 per ether, shake with anhydrous sodium sulphate and filter.
cent and not more than 101.0 per cent of C19H29NO,HCI, Evaporate the filtrate and dissolve the residue in 1 ml of ether.
calculated on the dried basis. Reference solution (a). Prepare in the same manner as the test
Description. A white, crystalline powder, odourless or almost solution but using 20 ml of a 0.5 per cent w/v solution of the
odourless. substance under examination and omitting the addition of the
internal standard solution.
Identification
Reference solution (b). Prepare in the same manner as the test
A. Determine by infrared absorption spectrophotometry (2.4.6). solution but using 20 ml of a 0.5 per cent w/v solution of the
Compare the spectrum with that obtained with procyclidine substance under examination.
hydrochloride RS. Chromatographic system
B. Dissolve 0.25 g in 10 ml of water, make alkaline with 5 M – a glass column 1.5 m x 4 mm, packed with acid-washed,
ammonia and extract with three quantities, each of 10 ml, of silanised diatomaceous support (such as HP
ether. Dry the combined ethereal extracts over anhydrous chromosorb W) coated with 10 per cent w/w of modified
sodium sulphate, filter, remove the ether and scratch the polyethylene glycol 20M (such as SP-1000) and 2 per
residue with a glass rod to induce solidification. The residue cent w/w of potassium hydroxide,
melts at about 85º (2.4.21). – temperature :
column. 240º,
C. Gives the reactions of chlorides (2.3.1).
inlet port and detector at 240°,
Tests – flow rate. 30 ml per minute of the carrier gas.

pH (2.4.24). 4.5 to 6.5, determined in a 1.0 per cent w/v solution. The ratio of the sum of the areas of any secondary peaks to
the area of the peak due to the internal standard in the
Related substances. A. Determine by thin-layer chromatogram obtained with reference solution (b) is not more
chromatography (2.4.17), coating the plate with silica gel than the ratio of the area of the principal peak to the area of the
GF254. internal standard peak in the chromatogram obtained with the
Mobile phase. A mixture of 100 volumes of ether and 1 volume test solution.
of strong ammonia solution. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Test solution. Dissolve 0.2 g of the substance under
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
examination in 10 ml of chloroform.
on 1.0 g by drying in an oven at 105º.
Reference solution (a). A 0.004 per cent w/v solution of
Assay. Weigh accurately about 0.7 g, dissolve in 75 ml of
1-phenyl-3-pyrrolidinopropan-1-one hydrochloride RS in
anhydrous glacial acetic acid, warm if necessary to effect
chloroform.
solution and cool. Add 10 ml of mercuric acetate solution.
Reference solution (b). A 0.01 per cent w/v solution of the Titrate with 0.1 M perchloric acid, using crystal violet
substance under examination in chloroform. solution as indicator. Carry out a blank titration.
Apply to the plate 5 µl of each solution. After development, 1 ml of 0.1 M perchloric acid is equivalent to 0.03239 g of
dry the plate at 105º for 15 minutes and examine in ultraviolet C19H29NO,HCI.
light at 254 nm. Any spot corresponding to 1-phenyl-3-
Storage. Store protected from moisture.
pyrrolidinopropan-1-hydrochloride-1-one in the chromatogram
obtained with the test solution is not more intense than the
spot in the chromatogram obtained with reference solution
(a). Spray the plate with dilute potassium iodobismuthate Procyclidine Tablets
solution. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the Procyclidine Hydrochloride Tablets
chromatogram obtained with reference solution (b). Procyclidine Tablets contain not than 90.0 per cent and not
B. Determine by gas chromatography (2.4.13). more than 110.0 per cent of the stated amount of procyclidine
hydrochloride, C19H29NO,HCI.
Test solution. Add 5 ml of 1.25 M sodium hydroxide to 20 ml
of a 0.015 per cent w/v solution of the substance under Identification
examination and mix. Extract with two quantities, each of
20 ml, of ether, add to the combined extracts 5 ml of a 0.06 per A. Dissolve a quantity of the powdered tablets containing
cent w/v solution of triphenylethylene (internal standard) in about 25 mg of Procyclidine Hydrochloride in 10 ml of water,

987
PROGUANIL HYDROCHLORIDE IP 2007

shake with 20 ml of ether and discard the ether layer. Make the 0.5 ml of water and 4.5 ml of a 0.025 per cent w/v solution of
aqueous layer alkaline with 2 M sodium hydroxide and extract bromocresol purple in the same manner beginning at the
with two quantities, each of 20 ml, of ether. Wash the combined words “extract with 20 ml of chloroform …….”.
ether extracts with two quantities, each of 10 ml, of water, dry Calculate the content of C19H29NO,HCI from the absorbance
by shaking with anhydrous sodium sulphate, filter and obtained by repeating the operation using procyclidine
evaporate the filtrate to dryness. It necessary, induce hydrochloride RS in place of the powdered tablets.
crystallization by scratching with a glass rod.
Storage. Store protected from moisture.
On the residue, determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with procyclidine hydrochloride RS.
B. The powdered tablets give the reactions of chlorides (2.3.1). Proguanil Hydrochloride
Tests Chloroguanide Hydrochloride

Related substances. Determine by thin-layer chromatography Cl


(2.4.17), coating the plate with silica gel GF254. NH NH CH3
, HCl
Mobile phase. A mixture of 100 volumes of ether and 1 volume N N N CH3
of strong ammonia solution. H H H
Test solution. Shake a quantity of the powdered tablets C11H16CIN5, HCl Mol. Wt. 290.2
containing 25 mg of Procyclidine Hydrochloride with 5 ml of
Proguanil Hydrochloride is 1-(4-chlorophenyl)-5-
chloroform and filter.
isopropylbiguanide hydrochloride.
Reference solution (a). A 0.001 per cent w/v solution of
Proguanil Hydrochloride contains not less than 99.0 per cent
1-phenyl-3-pyrrolidinopropan-1-one hydrochloride RS in
and not more than 101.0 per cent of C11H16ClN5, HCl, calculated
chloroform.
on the dried basis.
Reference solution (b). Dilute 1 volume of test solution to
Description. A white, crystalline powder; odourless.
200 volumes with chloroform.
Apply to the plate 20 µl of each solution. After development, Identification
dry the plate at 105º for 15 minutes and examine in ultraviolet
light at 254 nm. Any spot corresponding to 1-phenyl-3- Test A may be omitted if tests B, C and D are carried out. Tests
pyrrolidinopropan-1-one in the chromatogram obtained with B and C may be omitted if tests A and D are carried out.
the test solution is not more intense than the spot in the A. Determine by infrared absorption spectrophotometry (2.4.6).
chromatogram obtained with reference solution (a) (0.2 per Compare the spectrum with that obtained with proguanil
cent). Spray the plate with dilute potassium iodobismuthate hydrochloride RS or with the reference spectrum of proguanil
solution. Any secondary spot in the chromatogram obtained hydrochloride.
with the test solution is not more intense than the spot in the
B. To 10 ml of a saturated solution add 0.25 ml of potassium
chromatogram obtained with reference solution (b) (0.5 per
ferrocyanide solution; a white precipitate is produced which
cent). Ignore any spot due to excipients on the line of
dissolves on addition of a few drops of dilute nitric acid.
application.
C. Dissolve 5 mg in 5 ml of a warm 1.0 per cent w/v solution of
Other tests. Comply with the tests stated under Tablets.
cetrimide and add 1 ml of 5 M sodium hydroxide and 1 ml of
Assay. Weigh and powder 20 tablets. Weigh accurately a bromine solution; a deep red colour is produced.
quantity of the powder containing about 2.5 mg of Procyclidine
D. Gives the reactions of chlorides (2.3.1).
Hydrochloride, transfer to a 100-ml volumetric flask, add
10.0 ml of water and mix, and dilute to volume with a 0.025 per Tests
cent w/v solution of bromocresol purple in 2 per cent v/v
solution of glacial acetic acid. Allow the undissolved particles Acidity or alkalinity. To 35 ml of water maintained at 60º to
to settle. Transfer 5.0 ml of the supernatant solution to a 65º add 0.2 ml of methyl red solution, neutralise with 0.01 M
separating funnel, extract with 20.0 ml of chloroform and filter sodium hydroxide or 0.01 M hydrochloric acid, add 0.4 g of
the extract, discarding the first 5 ml of the filtrate. Measure the the substance under examination and stir until dissolved. The
absorbance of the filtrate at the maximum at about 405 nm resulting solution is not acidic and requires for neutralisation
(2.4.7), using as the blank a solution prepared by treating not more than 0.2 ml of 0.01 M hydrochloric acid.

988
IP 2007 PROMETHAZINE HYDROCHLORIDE

4-Chloroaniline. Dissolve 0.1 g in 1 ml of 2 M hydrochloric Tests


acid, add sufficient water to produce 20 ml, cool to 5º, add 1
ml of 0.05 M sodium nitrite, allow to stand at 5º for 5 minutes, 4-Chloroaniline. To a quantity of the powdered tablets
add 2 ml of a 5 per cent w/v solution of ammonium sulphamate containing about 0.1 g of Proguanil Hydrochloride add 5 ml of
and allow to stand for 10 minutes. Add 2 ml of a 0.1 per cent ethanol (95 per cent) and shake for 10 minutes. Add 2.5 ml of
w/v solution of N-(1-naphthyl)ethylenediamine 2 M hydrochloric acid and 15 ml of water, mix and filter
dihydrochloride, dilute to 50 ml with water and allow to stand through a wetted filter paper, washing the filter with 5 ml of
for 30 minutes. Any magenta colour produced is not more water. Cool to 5º, add 1 ml of 0.05 M sodium nitrite, allow to
intense than that obtained by treating in the same manner and stand at 5º for 5 minutes, add 2 ml of a 5 per cent w/v solution
at the same time 20 ml of a solution containing 1.25 µg of of ammonium sulphamate and allow to stand for 10 minutes.
4-chloroaniline. Add 2 ml of a 0.1 per cent w/v solution of
N-(1-naphthyl)ethylenediamine dihydrochloride, dilute to
Sulphated ash (2.3.18). Not more than 0.1 per cent. 50 ml with water and allow to stand for 30 minutes. Any
Loss on drying (2.4.19). Not more than 0.5 per cent, determined magenta colour produced is not more intense than that
on 1.0 g by drying in an oven at 105º. obtained by treating in the same manner and at the same time
20 ml of a solution containing 1.25 µg of 4-chloroaniline.
Assay. Weigh accurately about 0.15 g, dissolve in 25 ml of
anhydrous glacial acetic acid and 10 ml of mercuric acetate Other tests. Comply with the tests stated under Tablets.
solution. Titrate with 0.1 M perchloric acid, determining the Assay. Weigh and powder 20 tablets. Weigh accurately a
end-point potentiometrically (2.4.25). Carry out a blank titration. quantity of the powder containing about 0.1 g of Proguanil
1 ml of 0.1 M perchloric acid is equivalent to 0.01451 g of Hydrochloride, add 5 ml of water and warm on a water-bath
C11H16ClN5, HCl. with stirring until a smooth paste is obtained. Add 50 ml of
Storage. Store protected from light and moisture. water, continue warming for 10 minutes, cool, add sufficient
water to produce 100.0 ml and filter. Dilute 10.0 ml of the filtrate
to 100.0 ml with water and to 10.0 ml of the resulting solution
add 70 ml of water, 5 ml of a 20 per cent w/v solution of
Proguanil Tablets cetrimide and 1 ml of 2-propanol. Adjust the temperature of
Proguanil Hydrochloride Tablets; Chloroguanide the solution to 20º and add 2 ml of alkaline sodium
hypobromite solution and sufficient water to produce
Hydrochloride Tablets
100.0 ml. Allow to stand at 20º for 25 minutes and measure the
Proguanil Tablets contain not less than 95.0 per cent and not absorbance of the resulting solution at the maximum at about
more than 105.0 per cent of the stated amount of proguanil 480 nm (2.4.7).
hydrochloride, C11H16ClN5, HCl.
Calculate the content of C11H16ClN5,HCl from the absorbance
Identification obtained by repeating the operation using 10 ml of a 0.01 per
cent w/v solution of proguanil hydrochloride RS beginning
A. Boil a quantity of the powdered tablets containing 0.5 g of at the words “add 70 ml of water,....”.
Proguanil Hydrochloride with 5 ml of dilute hydrochloric acid,
cool and filter. To the filtrate add a slight excess of sodium Storage. Store protected from light and moisture.
hydroxide solution, extract with 30 ml of ether and evaporate
the ethereal extract; the residue, after drying at 105º, melts at
about 131º (2.4.21). Promethazine Hydrochloride
Dissolve the residue obtained in test A in the minimum quantity
of dilute hydrochloric acid; the solution diluted with water
CH3
to about 40 ml and neutralised if necessary, with cautious
addition of dilute ammonia solution, complies with tests the N
N CH3
following tests. , HCl
S CH3
B. To 10 ml add 0.25 ml of potassium ferrocyanide solution; a
white precipitate is produced which dissolves on addition of
a few drops of dilute nitric acid.
C17H20N2S,HCl Mol. Wt. 320.9
C. To 0.5 ml add 5 ml of a warm 1.0 per cent w/v solution of
cetrimide and add 1 ml of 5 M sodium hydroxide and 1 ml of Promethazine Hydrochloride is (RS)-dimethyl(2-
bromine solution; a deep red colour is produced. phenothiazin-10-ylpropyl)amine hydrochloride.

989
PROMETHAZINE INJECTION IP 2007

Promethazine Hydrochloride contains not less than 98.5 per Assay. Weigh accurately about 0.25 g, dissolve in a mixture of
cent and not more than 101.0 per cent of C17H20N2S, HCl, 5 ml of 0.01 M hydrochloric acid and 50 ml of ethanol (95 per
calculated on the dried basis. cent) and titrate with 0.1 M sodium hydroxide, determining
Description. A white or faintly yellowish, crystalline powder. the end-point potentiometrically (2.4.25). Record the volume
added between the two inflections.
Identification 1 ml of 0.1 M sodium hydroxide is equivalent to 0.03209 g of
C17H20N2S, HCl.
Test A may be omitted if tests B, C and D are carried out. Tests
B, C and D may be omitted if test A is carried out. Storage. Store protected from light and moisture.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with promethazine
hydrochloride RS or with the reference spectrum of Promethazine Injection
promethazine hydrochloride.
Promethazine Hydrochloride Injection
B. Complies with the test for identification of phenothiazines
(2.3.3). Promethazine Injection is a sterile solution of Promethazine
Hydrochloride in Water for Injections free from dissolved air.
C. Dissolve 0.1 g in 3 ml of water and add 1 ml of nitric acid
It may contain suitable stabilising agents.
dropwise; a precipitate is produced which dissolves rapidly
to give a red solution which becomes orange and then yellow. Promethazine Injection contains not less than 95.0 per cent
Heat the solution to boiling; it becomes orange and an orange- and not more than 105.0 per cent of the stated amount of
red precipitate is produced. promethazine hydrochloride, C17H20N2S, HCl.
D. Gives reaction B of chlorides (2.3.1).
Identification
Tests A. To a volume containing 0.1 g of Promethazine Hydrochloride
add 20 ml of water and 2 ml of 10 M sodium hydroxide. Shake
pH (2.4.24). 4.0 to 5.0, determined in a 10.0 per cent w/v solution
and extract the mixture with 25 ml of ether. Wash the ether
prepared immediately before use.
layer with two quantities, each of 5 ml, of water, dry with
Related substances. Carry out the test for related substances anhydrous sodium sulphate and evaporate to dryness.
in phenothiazines (2.3.5), protected from bright light using Dissolve the residue in 1 ml of chloroform.
mobile phase B.
Determine by infrared absorption spectrophotometry (2.4.6).
Prepare the following solutions immediately before use. Compare the spectrum with that obtained with promethazine
Test solution. Dissolve 0.2 g of the substance under hydrochloride RS treated in the same manner or with the
examination in 10 ml of a mixture of 95 volumes of methanol reference spectrum of promethazine.
and 5 volumes of diethylamine. B. To a volume containing 0.2 g of Promethazine Hydrochloride
Reference solution (a). A 0.01 per cent w/v solution of the add sufficient potassium carbonate to saturate the solution,
substance under examination in the same solvent mixture. extract with two quantities, each of 10 ml, of ether and
evaporate the combined extracts to dryness. Dissolve the
Reference solution (b). A 0.02 per cent w/v solution of residue in 2 ml of methanol and pour into a solution of 0.4 g of
isopromethazine hydrochloride RS in the same solvent picric acid in 10 ml of methanol, previously warmed to about
mixture.. 50º. Cool, scratch the sides of the tube to induce crystallisation,
Apply to the plate 10 µl of each solution. allow to stand for 3 to 4 hours and filter. The residue, after
washing with methanol and drying, melts at about 160º (2.4.21).
Any spot corresponding to isopromethazine in the
chromatogram obtained with the test solution is not more C. To a volume containing 5 mg of Promethazine Hydrochloride
intense than the spot in the chromatogram obtained with add carefully 2 ml of sulphuric acid and allow to stand for
reference solution (b) and any other secondary spot is not 5 minutes; a red colour is produced.
more intense than the spot in the chromatogram obtained
with reference solution (a). Tests
Sulphated ash (2.3.18). Not more than 0.1 per cent. pH (2.4.24). 5.0 to 6.0.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Related substances. Carry out the test for related substances
on 1.0 g by drying in an oven at 105º. in phenothiazines (2.3.5), using mobile phase B.

990
IP 2007 PROMETHAZINE TABLETS

Test solution. Dilute a volume of the injection with a mixture of Test solution. A solution prepared by diluting a suitable volume
95 volumes of methanol and 5 volumes of diethylamine to of the syrup with water to contain 0.08 per cent w/v of
contain 1 per cent w/v of Promethazine Hydrochloride. Promethazine Hydrochloride. Apply 5 µl to the plate.
Reference solution (a). Dilute 1 volume of the test solution to Tests
40 volumes with the same solvent mixture.
Other tests. Complies with the tests stated under Oral Liquids.
Reference solution (b). Dilute 1 volume of the test solution to
200 volumes with the same solvent mixture. Assay. Carry out the following procedure protected from
light.
Reference solution (c). A 0.01 per cent w/v solution of
isopromethazine hydrochloride RS in the same solvent Weigh accurately a quantity containing about 10 mg of
mixture. Promethazine Hydrochloride, add 25 ml of water and 5 ml of a
5 per cent w/v solution of sodium hydroxide. Extract the
Apply separately to the plate 10 µl of each solution. mixture with two quantities, each of 50 ml, of chloroform,
Any spot corresponding to isopromethazine in the shaking vigorously for 1 minute each time, evaporate the
chromatogram obtained with the test solution is not more combined extracts to dryness at about 30º at a pressure of
intense than the spot in the chromatogram obtained with 2 kPa and dissolve the residue in sufficient 0.1 M hydrochloric
reference solution (c). Any other secondary spot in the acid to produce 50.0 ml (solution A). Dilute 10 ml of solution A
chromatogram obtained with the test solution is not more to 50.0 ml with water (solution B). To a further 10 ml of solution
intense than the spot in the chromatogram obtained with A add 5 ml of peroxyacetic acid solution, allow to stand for
reference solution (a) and not more than one such spot is 10 minutes and add sufficient water to produce 50.0 ml (solution
more intense than the spot in the chromatogram obtained C). Measure the absorbance of solution C at the maximum at
with reference solution (b). about 336 nm (2.4.7), using solution B as the blank and measure
the absorbance of solution B at the same wavelength using
Other tests. Complies with the tests stated under Parenteral
water as the blank. Repeat the procedure using a 0.02 per cent
Preparations (Injections).
w/v solution of promethazine hydrochloride RS in 0.1 M
Assay. Carry out the following procedure protected from hydrochloric acid in place of solution A and beginning at the
light. words “Dilute 10 ml of......”.
To an accurately measured volume containing about 25 mg of Determine the weight per ml of the syrup (2.4.29), and calculate
Promethazine Hydrochloride add sufficient 0.01 M the content of C17H20N2S, HCl, weight in volume. The result is
hydrochloric acid to produce 100.0 ml. Dilute 10.0 ml to not valid unless the absorbance of solution B is less than
100.0 ml with 0.01 M hydrochloric acid, dilute 10.0 ml of this 0.10.
solution to 50.0 ml with 0.01 M hydrochloric acid and measure
Storage. Store protected from light and moisture.
the absorbance of the resulting solution at the maximum at
about 249 nm (2.4.7). Calculate the content of C17H20N2S, HCl
taking 910 as the specific absorbance at 249 nm.
Promethazine Tablets
Storage. Store protected from light.
Promethazine Hydrochloride Tablets
Promethazine Tablets contain not less than 92.5 per cent and
Promethazine Syrup not more than 107.5 per cent of the stated amount of
promethazine hydrochloride, C17H20N2S, HCl. The tablets are
Promethazine Hydrochloride Syrup; Promethazine coated.
Hydrochloride Oral Solution; Promethazine Oral Solution
Promethazine Syrup is a solution containing Promethazine
Identification
Hydrochloride in a suitable flavoured vehicle. A. To a quantity of the powdered tablets containing 40 mg of
Promethazine Syrup contains not less than 90.0 per cent and Promethazine Hydrochloride add 10 ml of water and 2 ml of
not more than 110.0 per cent of the stated amount of 1 M sodium hydroxide, shake and extract with 15 ml of ether.
promethazine hydrochloride, C17H20N2S, HCl. Wash the ether extract with 5 ml of water, dry with anhydrous
sodium sulphate, evaporate to dryness and dissolve the
Identification residue in 0.4 ml of chloroform.
Carry out the method for identification of phenothiazines Determine by infrared absorption spectrophotometry (2.4.6).
(2.3.3). Compare the spectrum with that obtained with promethazine

991
PROMETHAZINE THEOCLATE IP 2007

hydrochloride RS treated in the same manner or with the Weigh and powder 20 tablets. Weigh accurately a quantity of
reference spectrum of promethazine. the powder containing about 50 mg of Promethazine
B. Dissolve a quantity of the powdered tablets containing Hydrochloride with 10 ml of 2 M hydrochloric acid and add
0.2 g of Promethazine Hydrochloride in 2 ml of water, filter, 200 ml of water. Shake for 15 minutes, add sufficient water to
add sufficient potassium carbonate to saturate the solution, produce 500.0 ml and centrifuge about 50 ml of the mixture. To
extract with two quantities, each of 10 ml, of ether and 5.0 ml of the clear supernatant liquid add 10 ml of 0.1 M
evaporate the combined extracts to dryness. Dissolve the hydrochloric acid and sufficient water to produce 100.0 ml.
residue in 2 ml of methanol and pour into a solution of 0.4 g of Measure the absorbance of the resulting solution at the
picric acid in 10 ml of methanol, previously warmed to about maximum at about 249 nm (2.4.7). Calculate the content of
50º. Cool, scratch the sides of the tube to induce crystallisation, C17H20N2S, HCl taking 910 as the specific absorbance at
allow to stand for 3 to 4 hours and filter. The residue, after 249 nm.
washing with methanol and drying, melts at about 160º (2.4.21). Storage. Store protected from light and moisture.
C. To a quantity of the powdered tablets containing 5 mg of
Promethazine Hydrochloride add 5 ml of sulphuric acid and
allow to stand for 5 minutes; a red colour is produced. Promethazine Theoclate
Tests O
CH3 H
Related substances. Carry out the test for related substances H3C N
N N
in phenothiazines (2.3.5), using mobile phase B. N CH3 , Cl
Test solution. A solution freshly prepared by extracting a S CH3 O N N
quantity of the powdered tablets containing 0.1 g of CH3
Promethazine Hydrochloride with 10 ml of a mixture of
95 volumes of methanol and 5 volumes of diethylamine and
filtering. C17H20N2S,C7H7ClN4O2 Mol. Wt. 499.0
Reference solution (a). Dilute 1 volume of the test solution to Promethazine Theoclate is the (RS)-dimethyl(2-
40 volumes with the same solvent. phenothiazin-10-ylpropyl)amine salt of 8-
Reference solution (b). Dilute 1 volume of the test solution to chlorotheophylline.
200 volumes with the same solvent. Promethazine Theoclate contains not less than 98.0 per cent
Reference solution (c). A 0.01 per cent w/v solution of and not more than 101.0 per cent of C17H20N2S, C7H7ClN4O2,
isopromethazine hydrochloride RS in the same solvent. calculated on the dried basis.
Applying to the plate 10 µl of each solution. Any spot Description. A white or almost white powder; odourless or
corresponding to isopromethazine in the chromatogram almost odourless.
obtained with the test solution is not more intense than the
Identification
spot in the chromatogram obtained with reference solution
(c). Any other secondary spot in the chromatogram obtained Test A may be omitted if tests B, C, D and E are carried out.
with the test solution is not more intense than the spot in the Tests B, C and D may be omitted if tests A and E are carried
chromatogram obtained with reference solution (a) and not out.
more than one such spot is more intense than the spot in the
A. Shake 0.15 g with 2.5 ml of water, add 1 ml of 5 M ammonia
chromatogram obtained with reference solution (b).
and extract with 30 ml of ether. Wash the ether extract with
Uniformity of content. (For tablets containing 10 mg or less) 10 ml of water, dry with anhydrous sodium sulphate and
— Comply with the test stated under Tablets. evaporate to dryness. Dissolve the residue in 1 ml of
Crush one tablet, add 1 ml of dilute hydrochloric acid and 30 chloroform.
ml of water and shake for 15 minutes. Add sufficient water to Determine by infrared absorption spectrophotometry (2.4.6).
produce 50.0 ml and centrifuge. Complete the procedure Compare the spectrum with that obtained with promethazine
described in the Assay beginning at the words “To 5.0 ml of RS or with the reference spectrum of promethazine.
the clear supernatant liquid....”.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Other tests. Comply with the tests stated under Tablets. 0.0007 per cent w/v solution in ethanol containing 0.01 per
Assay. Carry out the following procedure protected from cent v/v of strong ammonia solution shows an absorption
light. maximum at about 255 nm; absorbance at about 0.53.

992
IP 2007 PROMETHAZINE THEOCLATE TABLETS

C. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand Promethazine Theoclate Tablets


for 5 minutes; a red colour is produced.
Promethazine Theoclate Tablets contain not less than 92.5 per
D. Shake 0.4 g with 10 ml of water, add 4 ml of 5 M ammonia, cent and not more than 107.5 per cent of the stated amount of
shake with two quantities, each of 30 ml, of ether and add 4 ml promethazine theoclate, C17H20N2S, C7H7ClN4O2.
of hydrochloric acid to the aqueous solution; a white
precipitate is produced. Filter, wash with water and dry at Identification
105º. Dissolve 10 mg of the residue in 1 ml of hydrochloric
acid, add 0.1 g of potassium chlorate and evaporate to A. To a quantity of the powdered tablets containing 40 mg of
dryness; a reddish residue remains which becomes purple on Promethazine Theoclate add 10 ml of water and 2 ml of 1 M
exposure to the vapour of ammonia. sodium hydroxide, shake and extract with 15 ml of ether. Wash
the ether extract with 5 ml of water, dry with anhydrous sodium
E. Fuse 50 mg of the residue obtained in test D with 0.5 g of
sulphate, evaporate to dryness and dissolve the residue in
anhydrous sodium carbonate, boil the residue with 5 ml of
0.4 ml of chloroform.
water, acidify to litmus paper with nitric acid and filter. The
filtrate gives reaction A of chlorides (2.3.1). Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with promethazine
Tests theoclate RS treated in the same manner or with the reference
spectrum of promethazine.
Chlorides (2.3.12). Shake 1.5 g with 50 ml of water for 2 minutes
and filter. 25 ml of the filtrate complies with the limit test for B. Dissolve a quantity of the powdered tablets containing
chlorides (330 ppm). 0.2 g of Promethazine Theoclate in 2 ml of water, filter, add
sufficient potassium carbonate to saturate the solution, extract
Related substances. Carry out the test for related substances
with two quantities, each of 10 ml, of ether and evaporate the
in phenothiazines (2.3.5), protected from bright light, using
combined extracts to dryness. Dissolve the residue in 2 ml of
mobile phase B.
methanol and pour into a solution of 0.4 g of picric acid in
Prepare the following solutions immediately before use. 10 ml of methanol, previously warmed to about 50º. Cool,
Test solution. Dissolve 0.2 g of the substance under scratch the sides of the tube to induce crystallisation, allow to
examination in 10 ml of a mixture of 95 volumes of methanol stand for 3 to 4 hours and filter. The residue, after washing
and 5 volumes of diethylamine. with methanol and drying, melts at about 160º (2.4.21).

Reference solution (a). A 0.01 per cent w/v solution of the C. To a quantity of the powdered tablets containing 5 mg of
substance under examination in the same solvent mixture. Promethazine Theoclate add 5 ml of sulphuric acid and allow
to stand for 5 minutes; a red colour is produced.
Reference solution (b). A 0.02 per cent w/v solution of
isopromethazine hydrochloride RS in the same solvent D. Extract a quantity of the powdered tablets containing 0.2 g
mixture. of Promethazine Theoclate with chloroform, filter and
evaporate the filtrate to dryness. Shake the residue with 10 ml
Apply to the plate 10 µl of each solution. Any spot of water, add 4 ml of 5 M ammonia and extract with two
corresponding to isopromethazine in the chromatogram quantities, each of 30 ml, of ether. Wash the combined extracts
obtained with the test solution is not more intense than the with 10 ml of water. Combine the aqueous layer and washings,
spot in the chromatogram obtained with reference solution add 4 ml of hydrochloric acid; a white precipitate is produced.
(b) and any other secondary spot is not more intense than the Filter, wash the residue with water and dry at 105º. Dissolve
spot in the chromatogram obtained with reference solution (a). 10 mg of the residue in 1 ml of hydrochloric acid, add 0.1 g of
Sulphated ash (2.3.18). Not more than 0.1 per cent. potassium chlorate and evaporate to dryness; a reddish
residue remains which becomes purple on exposure to the
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
vapour of ammonia.
on 1.0 g by drying in an oven at 105º.
Assay. Weigh accurately about 1.0 g and dissolve in 200 ml of Tests
acetone. Titrate with 0.1 M perchloric acid, using 3 ml of a
Related substances. Carry out the test for related substances
saturated solution of methyl orange in acetone as indicator.
in phenothiazines (2.3.5), using mobile phase B.
Carry out a blank titration.
Test solution. A solution freshly prepared by extracting a
1 ml of 0.1 M perchloric acid is equivalent to 0.04990 g of
quantity of the powdered tablets containing 0.1 g of
C17H20N2S,C7H7ClN4O2.
Promethazine Theoclate with 10 ml of a mixture of 95 volumes
Storage. Store protected from light and moisture. of methanol and 5 volumes of diethylamine and filtering.

993
PROPANTHELINE BROMIDE IP 2007

Reference solution (a). Dilute 1 volume of the test solution to Description. White or yellowish white crystals or powder;
40 volumes with the same solvent mixture. odourless; slightly hygroscopic.
Reference solution (b). Dilute 1 volume of the test solution to
Identification
200 volumes with the same solvent mixture.
Reference solution (c). A 0.01 per cent w/v solution of Test A may be omitted if tests B, C, D and E are carried out.
isopromethazine hydrochloride RS in the same solvent Tests B and C may be omitted if tests A, D and E are carried
mixture. out.
Apply to the plate 10 µl of each solution. Any spot A. Determine by infrared absorption spectrophotometry (2.4.6).
corresponding to isopromethazine in the chromatogram Compare the spectrum with that obtained with propantheline
obtained with the test solution is not more intense than the bromide RS or with the reference spectrum of propantheline
spot in the chromatogram obtained with reference solution bromide.
(c). Any other secondary spot in the chromatogram obtained B. When examined in the range 230 nm to 360 nm (2.4.7), a
with the test solution is not more intense than the spot in the 0.006 per cent w/v solution in methanol shows absorption
chromatogram obtained with reference solution (a) and not maxima at about 246 nm and 282 nm; absorbance at about
more than one such spot is more intense than the spot in the 246 nm, about 0.7 and at about 282 nm, about 0.37.
chromatogram obtained with reference solution (b).
C. Dissolve 0.2 g in 15 ml of water, add 1 ml of 10 M sodium
Other tests. Comply with the tests stated under Tablets. hydroxide, boil for 2 minutes, cool slightly, add 7.5 ml of 2 M
Assay. Carry out the following procedure protected from hydrochloric acid, cool and filter. Wash the residue with water,
light. recrystallise from ethanol (50 per cent) and dry at 105º for
1 hour. Dissolve about 10 mg of the crystals so obtained in
Weigh and powder 20 tablets. Weigh accurately a quantity of
5 ml of sulphuric acid (96 per cent w/w); an intense yellow
the powder containing about 50 mg of Promethazine Theoclate,
colour is produced which fluoresces strongly in ultraviolet
add 5 ml of water and 1 ml of strong ammonia solution and
light at 365 nm.
allow to stand for 5 minutes. Add 50 ml of ethanol, shake for
5 minutes and filter, washing the residue with five quantities, D. In the test for Related substances, the principal spot in the
each of 5 ml, of ethanol. Add sufficient ethanol to the filtrate chromatogram obtained with test solution (b) corresponds to
to produce 100.0 ml. Dilute 10.0 ml to 100.0 ml with ethanol that in the chromatogram obtained with reference solution
and dilute 10.0 ml of this solution to 100.0 ml with ethanol. (b).
Measure the absorbance of the resulting solution at the E. Gives the reactions of bromides (2.3.1).
maximum at about 255 nm (2.4.7). Calculate the content of
C17H20N2S, C7H7ClN4O2 taking 755 as the specific absorbance Tests
at 255 nm.
Appearance of solution. A 3.0 per cent w/v solution is clear
Storage. Store protected from light and moisture.
(2.4.1).
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Propantheline Bromide
Mobile phase. A mixture of 140 volumes of
H3C CH3 1,2-dichloroethane, 60 volumes of methanol, 2.5 volumes of
O anhydrous formic acid and 2.5 volumes of water.
N CH3 Br
O Test solution (a). Dissolve 0.1 g of the substance under
H3C examination in 10 ml of chloroform.
O CH3
Test solution (b). Dissolve 25 mg of the substance under
examination in 100 ml of chloroform.
C23H30BrNO3 Mol. Wt. 448.4 Reference solution (a). A 0.005 per cent w/v solution of the
substance under examination in chloroform.
Propantheline Bromide is N-methyl-N,N-bis(1-methylethyl)-
2-[(9H-xanthen-9-ylcarbonyl)oxy]ethanaminium bromide. Reference solution (b). A 0.025 per cent w/v solution of
propantheline bromide RS in chloroform.
Propantheline Bromide contains not less than 98.0 per cent
and not more than 102.0 per cent of C23H30BrNO3, calculated Apply to the plate 10 µl of each solution. After development,
on the dried basis. dry the plate in air and examine in ultraviolet light at 254 nm.

994
IP 2007 PROPRANOLOL HYDROCHLORIDE

Any secondary spot in the chromatogram obtained with test 25.0 ml to 100.0 ml with 0.1M sodium hydroxide. The
solution (a) is not more intense than the spot in the absorbance of the resulting solution at the maximum at about
chromatogram obtained with reference solution (a). 248 nm is not more than 0.31 (2.4.7).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Other tests. Comply with the tests stated under Tablets.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined Assay. Weigh and powder 20 tablets or more if necessary.
on 1.0 g by drying in an oven at 105º. Weigh accurately a quantity of the powder containing about
0.5 g of Propantheline Bromide, place it on a sintered-glass
Assay. Weigh accurately about 0.4 g, dissolve in a mixture of
filter, add 10 ml of peroxide-free ether, dried over sodium and
50 ml of acetic anhydride and 7 ml of mercuric acetate
distilled before use, stir and filter. Repeat the extraction with
solution. Titrate with 0.1 M perchloric acid, using crystal
four quantities, each of 10 ml, of ether and reserve the combined
violet solution as the indicator. Carry out a blank titration.
ether extracts for the test for Xanthanoic acid. Extract the
1 ml of 0.1 M perchloric acid is equivalent to 0.04484 g of residue on the filter with four quantities, each of 10 ml, of
C23H30BrNO3. chloroform, evaporate the combined chloroform extracts to
Storage. Store protected from moisture. about 10 ml, add 10 ml of mercuric acetate solution. Titrate
with 0.1 M perchloric acid, using crystal violet solution as
indicator. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.04484 g of
Propantheline Tablets C23H30BrNO3.
Propantheline Bromide Tablets Storage. Store protected from moisture.
Propantheline Tablets contain not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of
propantheline bromide, C23H30BrNO3. The tablets are coated.
Propranolol Hydrochloride
Identification
Triturate a quantity of the powdered tablets containing 75 mg
of Propantheline Bromide with 10 ml of chloroform, filter, OH H
evaporate the chloroform and stir the residue with 5 ml of O N CH3 , HCl
ether until it solidifies. The solid complies with the following
tests. CH3

A. Dissolve 60 mg in 2 ml of water, add 2 ml of 5 M sodium


hydroxide, boil for 2 minutes, cool slightly, acidify with 2 M C16H21NO2,HCl Mol. Wt. 295.8
hydrochloric acid, heat to boiling, add ethanol (95 per cent) Propranolol Hydrochloride is (2RS)-1-[(1-
dropwise until the precipitate just dissolves, cool and filter. methylethyl)amino]-3-(naphthalen-1-yloxy)propan-2-ol
The residue, after washing with water, recrystallising from hydrochloride.
ethanol (50 per cent) and drying at 105º for 1 hour, melts at
Propranolol Hydrochloride contains not less than 99.0 per
about 215º (2.4.21).
cent and not more than 101.0 per cent of C16H21NO2, HCl,
B. To 10 mg of the crystals obtained in test A add 5 ml of calculated on the dried basis.
sulphuric acid (96 per cent w/w); an intense yellow colour is
Description. A white or almost white powder.
produced which fluoresces strongly in ultraviolet light at
365 nm. Identification
C. Gives the reactions of bromides (2.3.1).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Tests Compare the spectrum with that obtained with propranolol
hydrochloride RS or with the reference spectrum of
Xanthanoic acid. Shake the combined ether extracts reserved propranolol hydrochloride.
in the Assay with two quantities, each of 30 ml, of 0.1 M
B. When examined in the range 230 nm to 360 nm (2.4.7), a
sodium hydroxide containing 1.5 per cent w/v solution of
0.002 per cent w/v solution in methanol shows absorption
sodium chloride. Remove the ether from the combined
maxima at about 290 nm, 306 nm and 319 nm.
aqueous extracts by heating on a water-bath, add sufficient
0.1 M sodium hydroxide to produce 100.0 ml and dilute C. Gives reaction A of chlorides (2.3.1).

995
PROPRANOLOL INJECTION IP 2007

Tests extracts with water until the washings are free from alkali, dry
with anhydrous sodium sulphate, filter, evaporate the filtrate
Appearance of solution. A 10.0 per cent w/v solution in to dryness and dry the residue at 50º at a pressure of 2 kPa for
methanol is clear (2.4.1), and not more intensely coloured 1 hour.
than degree 6 of the appropriate range of reference solutions
(2.4.1). On the residue determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that
pH (2.4.24). 5.0 to 6.0, determined in a 1.0 per cent w/v solution. obtained with propranolol hydrochloride RS treated in the
Specific optical rotation (2.4.22). –1.0º to +1.0º, determined in same manner or with the reference spectrum of propranolol.
a 4.0 per cent w/v solution. B. When examined in the range 230 nm to 360 nm (2.4.7), the
Related substances. Determine by thin-layer chromatography solution obtained in the Assay shows absorption maxima at
(2.4.17), coating the plate with silica gel G. about 290 nm, 306 nm and 319 nm.
Mobile phase. A mixture of 90 volumes of toluene and Tests
10 volumes of methanol.
pH (2.4.24). 3.0 to 3.5.
Test solution. Dissolve 1.0 g of the substance under
examination in 10 ml of methanol. Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Reference solution. Dissolve 20.0 mg of the substance under
examination in 100 ml of methanol. Assay. To an accurately measured volume containing about
2 mg of Propranolol Hydrochloride add sufficient methanol to
Apply to the plate 10 µl of each solution. After development,
produce 100.0 ml and measure the absorbance of the resulting
dry the plate in air, spray with anisaldehyde solution and heat
solution at the maximum at about 290 nm (2.4.7). Calculate the
at 105º for 15 minutes. Any secondary spot in the
content of C 16H 21NO 2, HCl taking 206 as the specific
chromatogram obtained with the test solution is not more
absorbance at 290 nm.
intense than the spot in the chromatogram obtained with the
reference solution. Storage. Store protected from light, in a single dose containers.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105º. Propranolol Tablets
Assay. Weigh accurately about 0.25 g, dissolve in 25 ml of Propranolol Hydrochloride Tablets
ethanol (95 per cent) and titrate with 0.1 M sodium hydroxide,
determining the end-point potentiometrically (2.4.25). Propranolol Tablets contain not less than 92.5 per cent and
not more than 107.5 per cent of the stated amount of
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02958 g of
propranolol hydrochloride, C16H21NO2, HCl.
C16H21NO2, HCl.
Storage. Store protected from moisture. Identification
A.Suspend a quantity of the powdered tablets containing
0.1 g of Propranolol Hydrochloride in 20 ml of water, filter,
Propranolol Injection make the filtrate alkaline with 1 M sodium hydroxide and extract
with three quantities, each of 10 ml, of ether. Wash the combined
Propranolol Hydrochloride Injection extracts with water until the washings are free from alkali, dry
Propranolol Injection is a sterile solution of Propranolol with anhydrous sodium sulphate, filter, evaporate the filtrate
Hydrochloride in Water for Injections containing Citric Acid. to dryness and dry the residue at 50º at a pressure of 2 kPa for
1 hour.
Propranolol Injection contains not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of On the residue, determine by infrared absorption
propranolol hydrochloride, C16H21NO2, HCl. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with propranolol hydrochloride RS treated in t he
Identification same manner or with the reference spectrum of propranolol.
A. Make alkaline with 1 M sodium hydroxide a volume B. When examined in the range 230 nm to 360 nm (2.4.7), the
containing 10 mg of Propranolol Hydrochloride and extract final solution obtained in the Assay shows absorption maxima
with three quantities, each of 5 ml, of ether. Wash the combined at about 290 nm, 306 nm and 319 nm.

996
IP 2007 PROPYLENE GLYCOL

Tests Description. A white or creamy white, crystalline powder;


odourless or almost odourless.
Uniformity of content. (For tablets containing 10 mg or less)
— Comply with the test stated under Tablets. Identification
Transfer one tablet to a 100-ml volumetric flask, add 5 ml of A. When examined in the range 230 nm to 360 nm (2.4.7), a
dilute hydrochloric acid and allow to stand, swirling 0.001 per cent w/v solution in methanol shows an absorption
occasionally, until it is disintegrated. Add about 70 ml of maximum only at about 275 nm; absorption at about 275 nm,
methanol and shake well for about 1 minute. Dilute to volume about 0.49.
with methanol, mix and centrifuge an aliquot of the solution.
Dilute a suitable volume of the clear solution with methanol B. Dissolve 10 mg in 50 ml of water, cool and add 5 ml of dilute
to produce a solution containing 20 µg of Propranolol ammonia solution; a red colour is produced which becomes
Hydrochloride per ml. Measure the absorbance of the resulting brown on standing and is restored on shaking.
solution at the maximum at about 290 nm (2.4.7), using C. Dissolve 5 mg in 50 ml of water and add 0.05 ml of ferric
methanol as the blank. Calculate the content of C16H21NO2, chloride test solution; a bluish black colour is produced.
HCl taking 206 as the specific absorbance at 290 nm.
Dissolution (2.5.2).
Tests
Apparatus. No 1 Chlorides (2.3.12). Shake 1.0 g with 50 ml of water for 5 minutes
Medium. 900 ml of 0.1 M hydrochloric acid and filter. 25 ml of the filtrate complies with the limit test for
Speed and time. 100 rpm and 45 minutes. chlorides (500 ppm).
Withdraw a suitable volume of the medium and filter. Dilute a Sulphates (2.3.17). Shake 0.8 g with 100 ml of water for
suitable volume of the filtrate with the same solvent. Measure 5 minutes and filter. 15 ml of the filtrate complies with the limit
the absorbance of the resulting solution at the maximum at test for sulphates (0.12 per cent).
about 290 nm (2.4.7). Calculate the content of C16H21NO2,HCl Sulphated ash (2.3.18). Not more than 0.1 per cent.
in the medium taking 206 as the specific absorbance at 290 nm.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
D. Not less than 75 per cent of the stated amount of C16H21NO2, on 1.0 g by drying in an oven at 105º.
HCl.
Storage. Store protected from light and moisture, in non-
Other tests. Comply with the tests stated under Tablets. metallic containers.
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 20 mg of Propranolol
Hydrochloride and shake with 20 ml of water for 10 minutes.
Add 50 ml of methanol, shake for a further 10 minutes, add Propylene Glycol
sufficient methanol to produce 100.0 ml and filter. Dilute
10.0 ml of the filtrate to 50.0 ml with methanol and measure the 1, 2-Propanediol
absorbance of the resulting solution at the maximum at about
290 nm (2.4.7). Calculate the content of C16H21NO2, HCl taking OH
206 as the specific absorbance at 290 nm. OH
H3 C
Storage. Store protected from light and moisture.
C3H8O2 Mol. Wt. 75.1
Propylene Glycol is (RS)-propane-1,2-diol.
Propyl Gallate Description. A clear, colourless, viscous liquid; practically
odourless; hygroscopic.
O
HO CH3 Identification
O
A. To 0.5 ml of a 0.01 per cent w/v solution, cooled in ice, add
HO 5 ml of a cooled mixture of 10 ml of water and 90 ml of sulphuric
OH acid. Heat for 10 minutes on a water-bath at 70º, cool and add
0.2 ml of a 3 per cent w/v solution of ninhydrin in a 2.5 per cent
C10H12O5 Mol. Wt. 212.2 w/v solution of sodium metabisulphite; a violet colour slowly
Propyl Gallate is propyl 3,4,5-trihydroxybenzoate. appears.

997
PROPYLPARABEN IP 2007

B. Heat 0.15 ml with 0.1 g of boric acid; a pleasant odour Inject 3 µl or other suitable volume of the test solution. Record
develops. the chromatograms adjusting the sensitivity so that the height
C. Add 1 ml to 0.5 g of potassium bisulphate and heat gently; of the peak due to propylene glycol is more than 50 per cent of
a fruity odour develops and when the solution is heated to the full-scale deflection in the chromatograms. Inject the same
dryness, no sharp, acrid smell of acrolein is perceptible. volume of reference solution and record the chromatograms.
The order of elution is propylene glycol, ethylene glycol and
Tests diethylene glycol. The test is not valid unless in the
chromatogram obtained with the reference solution, the
Appearance of solution. The substance under examination is resolution between the peaks due to propylene glycol (first
clear (2.4.1), and colourless (2.4.1). peak) and ethylene glycol (second peak) is not less than 1.0.
Acidity. Mix 10 ml with 40 ml of water and add 0.1 ml of No peaks corresponding to ethylene glycol and diethylene
bromothymol blue solution. The solution is greenish yellow glycol are obtained in the chromatogram obtained with the
and not more than 0.05 ml of 0.1 M sodium hydroxide is test solution.
required to change the colour to blue.
Sulphated ash (2.3.18). Not more than 0.01 per cent w/w,
Boiling range (2.4.8). 184º to 189º. determined by the following method. Heat 50 g until it burns,
and ignite. Allow to cool, moisten the residue with sulphuric
Relative density (2.4.29). 1.035 to 1.040.
acid and ignite; repeat the operations.
Refractive index (2.4.27). 1.431 to 1.433.
Water (2.3.43). Not more than 0.2 per cent, determined on
Heavy metals (2.3.13). Dilute 3 ml to 12 ml with water. The 5.0 g.
resulting solution complies with the limit test for heavy metals,
Storage. Store protected from moisture.
Method D (5 ppm). Use lead standard solution (1 ppm Pb) to
prepare the standard.
Oxidising substances. To 10 ml add 5 ml of water, 2 ml of
potassium iodide solution and 2 ml of 1 M sulphuric acid and Propylparaben
allow to stand in a ground-glass-stoppered flask protected
from light for 15 minutes. Titrate the liberated iodine with Propyl Hydroxybenzoate
0.05 M sodium thiosulphate using 1 ml of starch solution,
added towards the end of the titration, as indicator. Not more O
than 0.2 ml of 0.05 M sodium thiosulphate is required.
CH3
Reducing substances. Mix 1 ml with 1 ml of 6 M ammonia and O
heat in a water-bath at 60º for 5 minutes; the solution is not HO
yellow. Immediately add 0.15 ml of 0.1 M silver nitrate; the
solution does not change its appearance within 5 minutes.
C10H12O3 Mol. Wt. 180.2
Ethylene glycol and diethylene glycol. Determine by gas
Propylparaben is propyl 4-hydroxybenzoate.
chromatography (2.4.13).
Propylparaben contains not less than 99.0 per cent and not
Test solution. Dissolve 2 g of the substance under examination
more than 101.0 per cent of C10H12O3, calculated on the dried
in sufficient ethanol (95 per cent) to produce 100 ml.
basis.
Reference solution. Dissolve 2 g of the substance under Description. A white, crystalline powder; odourless.
examination, 0.02 g of ethylene glycol and 0.02 g of diethylene
glycol in ethanol (95 per cent) and dilute to 100 ml with the Identification
same solvent.
A. When examined in the range 230 nm to 360 nm (2.4.7), a
Chromatographic system
0.0005 per cent w/v solution in ethanol (95 per cent) shows
– a glass column 1.5 m x 3 mm, packed with 12 per cent
an absorption maximum at about 258 nm; absorption at
Sorbitol on untreated siliceous earth (such as
258 nm, 0.44 to 0.47.
Chromosorb W-NAW (SINS),
– temperature: B. To about 10 mg in a test-tube add 1 ml of sodium carbonate
column: 165º, solution, heat to boiling for 30 seconds and cool (solution A).
inlet port and detector at 260º, To a further 10 mg in a test-tube add 1 ml of sodium carbonate
– flow rate. 30 ml per minute of the carrier gas. solution; the substance partly dissolves (solution B). Add at

998
IP 2007 PROPYLTHIOURACIL

the same time to each of the solutions A and B 5 ml of stopper the flask and allow to stand for 15 minutes. Add 15 ml
aminophenazone solution and 1 ml of potassium ferricyanide of potassium iodide solution, mix and titrate the liberated
solution and mix. Solution B is yellow to orange-brown. iodine with 0.1 M sodium thiosulphate using 2 ml of starch
Solution A is orange to red and the colour is clearly more solution, added towards the end of the titration, as indicator.
intense than any similar colour that may be obtained with Repeat the operation without the substance under examination.
solution B. The difference between the titrations represents the amount
of potassium bromate required. The volume of 0.0333 M
Tests potassium bromate is equivalent to half of the volume of
0.1 M sodium thiosulphate required for the titration.
Appearance of solution. A 10.0 per cent w/v solution in ethanol
(95 per cent) is clear (2.4.1), and not more intensely coloured 1 ml of 0.0333 M potassium bromate is equivalent to
than reference solution BYS6 (2.4.1). 0.006007 g of C10H12O3.
Acidity. Dissolve 1.0 g in sufficient ethanol (95 per cent) to Storage. Store protected from moisture.
produce 10 ml. To 2 ml of the solution add 3 ml of ethanol
(95 per cent), 5 ml of carbon dioxide-free water and 0.1 ml of
bromocresol green solution. Not more than 0.1 ml of 0.1 M
sodium hydroxide is required to change the colour of the Propylthiouracil
solution.
Related substances. Determine by thin-layer chromatography H
(2.4.17), coating the plate with silica gel HF254. H3C N S
Mobile phase. A mixture of 88 volumes of dichloromethane, NH
10 volumes of ethyl acetate and 2 volumes of anhydrous
formic acid. O
Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml of methanol. C7H10N2OS Mol. Wt. 170.2
Reference solution. A 0.02 per cent w/v solution of the Propylthiouracil is 2,3-dihydro-6-propyl-2-thioxopyrimidin-
substance under examination in methanol. 4(1H)-one.
Apply to the plate 5 µl of each solution. After development, Propylthiouracil contains not less than 98.0 per cent and not
dry the plate in a current of hot air and examine in ultraviolet more than 100.5 per cent of C7H10N2OS, calculated on the dried
light at 254 nm. Any secondary spot in the chromatogram basis.
obtained with the test solution is not more intense than the Description. A white or pale cream-coloured crystals or
spot in the chromatogram obtained with the reference solution. crystalline powder; odourless.
Chlorides (2.3.12). Heat 2.0 g with 100 ml of water, cool, add
sufficient water to restore the original volume, and filter. 25 ml Identification
of the filtrate complies with the limit test for chlorides Test A may be omitted if tests B and C are carried out. Tests B
(500 ppm). and C may be omitted if test A is carried out.
Sulphates.To 10 ml of the filtrate obtained in the test for A. Determine by infrared absorption spectrophotometry (2.4.6).
Chlorides add 0.15 ml of dilute hydrochloric acid and 0.1 ml Compare the spectrum with that obtained with
of barium chloride solution; no turbidity is produced within propylthiouracil RS or with the reference spectrum of
10 minutes. propylthiouracil.
Sulphated ash (2.3.18). Not more than 0.1 per cent. B. Examine the chromatograms obtained in the test for Related
Loss on drying (2.4.19). Not more than 0.5 per cent, determined substances in ultraviolet light at 254 nm before exposure of
on 1.0 g by drying over silica gel for 5 hours. the plate to iodine vapour. The principal spot in the
chromatogram obtained with test solution (b) corresponds to
Assay. Weigh accurately about 80 mg, transfer to a glass-
that in the chromatogram obtained with reference solution
stoppered flask, add 25 ml of 2 M sodium hydroxide and boil
(b).
gently under a reflux condenser for 30 minutes. Cool and add
25.0 ml of 0.0333 M potassium bromate, 5 ml of a 12.5 per cent C. To about 20 mg add 8 ml of bromine water and shake for a
w/v solution of potassium bromide and 40 ml of glacial acetic few minutes. Boil until the mixture is decolorised, allow to cool
acid, cool in ice, add 10 ml of hydrochloric acid, immediately and filter. Add 2 ml of barium chloride solution; a white

999
PROPYLTHIOURACIL TABLETS IP 2007

precipitate is produced. Add 5 ml of 2 M sodium hydroxide; Propylthiouracil Tablets


the precipitate does not become violet.
Propylthiouracil Tablets contain not less than 92.5 per cent
Tests and not more than 107.5 per cent of the stated amount of
propylthiouracil, C7H10N2OS.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Identification
Mobile phase. A mixture of 100 volumes of chloroform, A. Shake a quantity of the powdered tablets containing 50 mg
12 volumes of 2-propanol and 0.2 volume of glacial acetic of Propylthiouracil with 20 ml of methanol for 10 minutes,
acid. filter and evaporate to dryness.
Test solution (a). Dissolve 0.1 g of the substance under On the residue, determine by infrared absorption
examination in 10 ml of methanol. spectrophotometry (2.4.6). Compare the spectrum with that
Test solution (b). Dissolve 0.1 g of the substance under obtained with propylthiouracil RS or with the reference
examination in 100 ml of methanol. spectrum of propylthiouracil.
Reference solution (a). A 0.01 per cent w/v solution of the B. Shake a quantity of the powdered tablets containing 50 mg
substance under examination in methanol. of Propylthiouracil with 60 ml of methanol for 20 minutes,
dilute to 100 ml with methanol and filter. Dilute 5 ml of the
Reference solution (b). A 0.1 per cent w/v solution of
filtrate to 250 ml with methanol. When examined in the range
propylthiouracil RS in methanol.
230 nm to 360 nm (2.4.7), the resulting solution shows an
Reference solution (c). A 0.0005 per cent w/v solution of absorption maximum only at about 274 nm.
thiourea in methanol. C. Extract a quantity of the powdered tablets in a continuous
Apply to the plate 10 µl of each solution. After development, extraction apparatus (2.1.8) with ether and evaporate the
dry the plate in air and examine in ultraviolet light at 254 nm. solution to dryness. The residue, after drying at 105º, melts at
Expose the plate to iodine vapour for 10 minutes. By both about 219º (2.4.21).
methods of visualisation, any spot corresponding to thiourea
in the chromatogram obtained with test solution (a) is not Tests
more intense than the spot in the chromatogram obtained
Related substances. Determine by thin-layer chromatography
with reference solution (c) and any other secondary spot is
(2.4.17), coating the plate with silica gel GF254.
not more intense than the spot in the chromatogram obtained
with reference solution (a). Mobile phase. A mixture of 100 volumes of chloroform,
12 volumes of 2-propanol and 0.2 volume of glacial acetic
Arsenic (2.3.10). Dissolve 2.0 g in 50 ml of water and add 10 ml
acid.
of stannated hydrochloric acid AsT. The resulting solution
complies with the limit test for arsenic (5 ppm). Test solution. Shake a quantity of the powdered tablets
containing 50 mg of Propylthiouracil with 5 ml of methanol for
Heavy metals (2.3.13). 1.0 g complies with the limit test for
15 minutes, filter and use the filtrate.
heavy metals, Method C (20 ppm).
Reference solution (a). Dilute 1 volume of the test solution to
Sulphated ash (2.3.18). Not more than 0.1 per cent.
100 volumes with methanol.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Reference solution (b). A 0.001 per cent w/v solution of
on 1.0 g by drying in oven at 105º.
thiourea in methanol.
Assay. Weigh accurately about 0.3 g, add 30 ml of water and
Apply to the plate 10 µl of each solution. After development,
30.0 ml (n1 ml) of 0.1 M sodium hydroxide, boil and shake until
dry the plate in air and examine in ultraviolet light at 254 nm.
solution is complete. Add 50 ml of 0.1 M silver nitrate with
Expose the plate to iodine vapour for 10 minutes. By both
stirring, boil gently for 5 minutes, cool and titrate with 0.1 M
methods of visualisation, any spot corresponding to thiourea
sodium hydroxide, determining the end-point
in the chromatogram obtained with the test solution is not
potentiometrically (2.4.25) (n2 ml). Record the total volume
more intense than the spot in the chromatogram obtained
(n1 + n2 ml) of 0.1 M sodium hydroxide added.
with reference solution (b) and any other secondary spot is
1 ml of 0.1 M sodium hydroxide is equivalent to 0.008511 g of not more intense than the spot in the chromatogram obtained
C7H10N2OS. with reference solution (a).
Storage. Store protected from light and moisture. Other tests. Comply with the tests stated under Tablets

1000
IP 2007 PROPYPHENAZONE

Assay. Weigh and powder 20 tablets. Weigh accurately a produce 50 ml (solution A). To 1 ml of solution A add 0.1 ml of
quantity of the powder containing about 0.15 g of ferric chloride solution; a brownish red colour is produced
Propylthiouracil, dissolve in a mixture of 20 ml of 0.1 M sodium which becomes yellow on addition of 1 ml of 2 M hydrochloric
hydroxide and 75 ml of water with the aid of gentle heat. Cool, acid.
add 4 g of sodium acetate, just acidify the solution to litmus
paper with 6 M acetic acid, add 0.5 ml of a freshly prepared Tests
0.5 per cent w/v solution of 1,5-diphenylcarbazone in ethanol Appearance of solution. Solution A is clear (2.4.1), and
(95 per cent) and titrate with 0.02 M mercuric nitrate until a colourless (2.4.1).
pinkish violet colour persists for 2 to 3 minutes.
Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of
1 ml of 0.02 M mercuric nitrate is equivalent to 0.006808 g of phenolphthalein solution; the solution is colourless. Add
C7H10N2OS. 0.2 ml of 0.01 M sodium hydroxide; the solution is pink. Add
Storage. Store protected from light and moisture. 0.4 ml of 0.01 M hydrochloric acid; the solution becomes
colourless. Add 0.2 ml of methyl red solution; the solution is
orange or red.
Related substances. Determine by thin-layer chromatography
Propyphenazone (2.4.17), coating the plate with silica gel HF254.
Mobile phase. A mixture of 45 volumes of cyclohexane,
45 volumes of ethyl acetate and 10 volumes of 1-butanol.
Test solution (a). An 8 per cent w/v solution of the substance
under examination in methanol.
O N CH3 Test solution (b). A 1.6 per cent w/v solution of the substance
under examination in methanol.
H3C CH3 Reference solution (a). A 0.016 per cent w/v solution of the
CH3 substance under examination in methanol.
Reference solution (b). A 1.6 per cent w/v solution of
C14H18N2O Mol. Wt. 230.3 propyphenazone RS in methanol.
Propyphenazone is 4-isopropyl-2,3-dimethyl-1-phenyl-3- Apply to the plate 5 µl of each solution. After development,
pyrazolin-5-one. dry the plate in a current of hot air for 15 minutes and examine
Propyphenazone contains not less than 99.0 per cent and not in ultraviolet light at 254 nm. Spray the plate with a mixture of
more than 101.0 per cent of C14H18N2O, calculated on the dried equal volumes of potassium ferricyanide solution and ferric
basis. chloride solution. By both methods of visualisation, any
secondary spot in the chromatogram obtained with test
Description. A white or slightly yellowish, crystalline powder; solution (a) is not more intense than the spot in the
odourless. chromatogram obtained with reference solution (a).

Identification Arsenic (2.3.10). Mix 1.0 g with 10 ml of a 2 per cent w/v


solution of magnesium nitrate in ethanol in a silica or platinum
Test A may be omitted if tests B and C are carried out. Tests B dish, evaporate on a water-bath and heat gradually in order to
and C may be omitted if test A is carried out. incinerate. If the material remains incompletely carbonised,
A. Determine by infrared absorption spectrophotometry (2.4.6). moisten with a small quantity of nitric acid and ignite again.
Compare the spectrum with that obtained with Cool, add 3 ml of hydrochloric acid and heat on a water-bath
propyphenazone RS or with the reference spectrum of to dissolve the residue. The resulting solution complies with
propyphenazone. the limit test for arsenic (10 ppm).

B. In the test for Related substances, the principal spot in the Heavy metals (2.3.13). 1.0 g complies with the limit test for
chromatogram obtained with test solution (b) corresponds to heavy metals, Method B (10 ppm).
that in the chromatogram obtained with reference solution Sulphated ash (2.3.18). Not more than 0.1 per cent.
(b).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
C. Dissolve 2 g in sufficient of a mixture of equal volumes of on 1.0 g by drying in an oven at 60º over phosphorus pentoxide
ethanol (95 per cent) and carbon dioxide-free water to at a pressure of 1.5 to 2.5 kPa for 4 hours.

1001
PROTAMINE SULPHATE IP 2007

Assay. Weigh accurately about 0.2 g of the dried material, Iron (2.3.14). Dissolve 2.0 g in water with the aid of heat and
dissolve in 30 ml of anhydrous glacial acetic acid. Titrate dilute to 20 ml with water. The resulting solution complies
with 0.1 M perchloric acid, determining the end-point with the limit test for iron (20 ppm).
potentiometrically (2.4.25). Carry out a blank titration. Mercury. Add 20 ml of a mixture of equal volumes of nitric
1 ml of 0.1 M perchloric acid is equivalent to 0.02303 g of acid and sulphuric acid to 2.0 g in a 250-ml flask fitted with a
C14H18N2O. ground-glass stopper, boil under a reflux condenser for 1 hour,
cool and carefully dilute with water. Boil until nitrous fumes
Storage. Store protected from moisture. are no longer evolved. Cool, carefully dilute the solution to
200 ml with water, mix and filter. Transfer 50 ml of the filtrate to
a separating funnel. Shake with successive small quantities of
chloroform until the chloroform layer remains colourless. To
Protamine Sulphate the aqueous layer add 25 ml of 1 M sulphuric acid, 115 ml of
water and 10 ml of a 20 per cent w/v solution of hydroxylamine
Protamine Sulphate is a purified mixture of the sulphates of hydrochloride. Titrate with dithizone solution; after each
basic peptides prepared from the sperm or mature testes of addition, shake the mixture 20 times and towards the end-
suitable species of fish. It binds with heparin in solution, point of the titration allow to separate and discard the
inhibiting its anticoagulant activity. It is prepared in conditions chloroform layer. Titrate until a greenish blue colour is
designed to minimise the degree of Microbial contamination. produced. Calculate the content of mercury using the
Each mg of Protamine Sulphate precipitates not less than equivalent of mercury in µg per ml of titrant determined in the
100 Units of heparin sodium RS, calculated on the dried basis. standardisation of the dithizone solution (10 ppm).
Description. A white or almost white powder; hygroscopic. Nitrogen (2.3.30). 21.0 to 26.0 per cent, calculated on the dried
basis, determined by Method C.
Identification Sulphates. 16 to 24 per cent, determined by the following
A. Produces a precipitate under the conditions of the Assay. method. Dissolve 0.15 g in 15 ml of water in a beaker, add 5 ml
of 2 M hydrochloric acid and heat to boiling. Slowly add to
B. Dissolve 0.2 g in 5 ml of water and dilute to 10 ml with the the boiling solution 10 ml of barium chloride solution. Cover
same solvent (solution A). To 0.5 ml of solution A add 4.5 ml of and heat on a water-bath for 1 hour. Filter and wash the
water, 1 ml of a 10 per cent w/v solution of sodium hydroxide precipitate several times with small quantities of hot water.
and 1 ml of a 0.02 per cent w/v solution of 1-naphthol and mix. Dry and ignite the residue to constant weight at 600º.
Cool to 5º and add 0.5 ml of alkaline sodium hypobromite
solution; an intense red colour is produced. 1 g of the residue is equivalent to 0.4117 g of SO4.
Abnormal toxicity (2.2.1). Complies with the test for abnormal
C. Heat 2 ml of solution A in a water-bath at 60º, add 0.1 ml of
toxicity, using 0.5 mg dissolved in 0.5 ml of water for injections.
mercuric sulphate solution and mix; no precipitate is produced.
Cool the mixture in ice; a white precipitate is produced. Loss on drying (2.4.19). Not more than 5.0 per cent, determined
on 1.0 g by drying in an oven at 105º for 3 hours.
D. Gives reaction A of sulphates (2.3.1).
Assay. Prepare solutions of the substance under examination
Tests in water containing (1) 0.015 per cent w/v (2) 0.01 per cent
w/v and (3) 0.005 per cent w/v. Titrate each of the solutions in
Appearance of solution. To 2.5 ml of solution A add 7.5 ml of duplicate with a 174 IU per ml or a suitable dilution of heparin
water. The resulting solution is not more opalescent than sodium RS using the following procedure. Introduce an
opalescence standard OS2 (2.4.1), and not more intensely accurately measured volume of the solution to be titrated, for
coloured than reference solution BYS6 (2.4.1). example 1.5 ml, into the cell of a suitable spectrophotometer,
Specific optical rotation (2.4.22). –65.0º to –85.0º, determined set the instrument at a suitable wavelength (none is critical) in
at 20º in a 1.0 per cent w/v solution in 0.1 M hydrochloric the visible range and add the titrant in small volumes until
acid. there is a sharp increase in the absorbance. Note the volume
of titrant added.
Light absorption (2.4.7). Dilute 2.5 ml of solution A to 5.0 ml
with water. Absorbance of the resulting solution at 260 to Carry out three independent assays. For each individual
280 nm is not more than 0.1. titration, calculate the number of Units of heparin titrated per
mg of the substance under examination. Calculate the result
Heavy metals (2.3.13). 1.0 g complies with the limit test for of the assay as the average of the 18 values. Test the linearity
heavy metals, Method B (20 ppm). of the response by standard statistical methods. The assay is

1002
IP 2007 PROTHIONAMIDE

not valid unless the standard deviations calculated for the Light absorption. Dilute the injection, if necessary, with water
results obtained with each test solution are less than 5 per to produce a solution containing 1 per cent w/v solution of
cent of the average result. Protamine Sulphate. Absorbance of the resulting solution at
Protamine Sulphate intended for use in the manufacture of 260 to 280 nm, not more than 0.1 (2.4.7).
Parenteral Preparations without a further appropriate Bacterial endotoxins (2.2.3). Not more than 7.0 Endotoxin Units
procedure for the removal of bacterial endotoxins complies per mg of protamine sulphate.
with the following additional requirement.
Abnormal toxicity. Complies with the test for abnormal toxicity
Bacterial endotoxins (2.2.3). Not more than 7.0 Endotoxin Units (2.2.1), using a volume containing 0.5 mg of Protamine
per mg of protamine sulphate. Sulphate.
Protamine Sulphate intended for use in the manufacture of Other tests. Complies with the tests stated under Parenteral
Parenteral Preparations without a further appropriate Preparations (Injections).
sterilisation procedure complies with the following Assay. Prepare solutions of the substance under examination
additional requirement. in water containing (1) 0.015 per cent w/v (2) 0.01 per cent
Sterility. Complies with the test for sterility (2.2.11). w/v and (3) 0.005 per cent w/v. Titrate each of the solutions in
duplicate with a 174 IU per ml or a suitable dilution of heparin
Storage. Store protected from moisture. If it is intended for
sodium RS using the following procedure. Introduce an
use in the manufacture of Parenteral Preparations, the container
accurately measured volume of the solution to be titrated, for
should be sterile and sealed so as to exclude micro-organisms.
example 1.5 ml, into the cell of a suitable spectrophotometer,
Labelling. The label states whether or not the contents are set the instrument at a suitable wavelength (none is critical) in
intended for use in the manufacture of Parenteral Preparations. the visible range and add the titrant in small volumes until
there is a sharp increase in the absorbance. Note the volume
of titrant added.
Protamine Sulphate Injection Carry out three independent assays. For each individual
Protamine Sulphate Injection is a sterile solution of Protamine titration, calculate the number of Units of heparin titrated per
Sulphate in Water for Injections. mg of the substance under examination. Calculate the result
of the assay as the average of the 18 values. Test the linearity
Protamine Sulphate Injection contains not less than 80.0 per of the response by standard statistical methods. The assay is
cent of the stated amount of protamine sulphate. not valid unless the standard deviations calculated for the
results obtained with each test solution are less than 5 per
Identification cent of the average result.
A. Produces a precipitate under the conditions of the Assay. Storage. Store protected from light, in single dose containers.
B. Dilute a suitable volume with water to give a solution Labelling. The label states (1) that the dose is calculated from
containing 0.2 per cent w/v solution of Protamine Sulphate. the results of determinations of the amount required to
To 5 ml of this solution add 1 ml of a 10 per cent w/v solution produce an acceptable blood-clotting time in the patient; (2)
of sodium hydroxide and 1 ml of a 0.02 per cent w/v solution the approximate number of Units of heparin activity 1 ml is
of 1-naphthol and mix. Cool to 5º and add 0.5 ml of alkaline capable of neutralising.
sodium hypobromite solution; an intense red colour is
produced.
C. Heat 2 ml in a water-bath at 60º, add 0.1 ml of mercuric
sulphate solution and mix; no precipitate is produced. Cool Prothionamide
the mixture in ice; a white precipitate is produced.
D. Gives reaction A of sulphates (2.3.1). N CH3

Tests
pH (2.4.24). 2.5 to 3.5.
S NH2
Optical rotation (2.4.22). –0.52º to –0.68º, determined in a
solution prepared by diluting the injection with 0.5 M
C9H12N2S Mol. wt. 180.3
hydrochloric acid so as to contain 0.8 per cent w/v of Protamine
Sulphate. Prothionamide is 2-propyl-4-pyridinecarbothioamide.

1003
PROTHIONAMIDE TABLETS IP 2007

Prothionamide contains not less than 99.0 per cent and not peak in the chromatogram obtained with the reference solution
more than 101.0 per cent of C8H10N2S ,calculated on the dried (1.0 per cent).
basis. Heavy Metals (2.3.13). 2.0 g complies with the limit test for
Description. A yellow, crystalline powder. heavy metals, Method B (10 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Identification
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
A. Determine by infrared absorption spectrophotometry (2.4.6). on 1.0 g by drying in an oven at 105º for 3 hours.
Compare the spectrum with that obtained with prothionamide
Assay. Determine by liquid chromatography (2.4.14) as
RS.
described under Related substances using the following
B. When examined in the range 230 nm to 350 nm (2.4.7), a solutions.
0.002 per cent w/v solution in ethanol (95 per cent) shows an
Test solution. Dissolve 50.0 mg of the substance under
absorption maximum only at about 291 nm. The absorbance at
examination in the mobile phase and dilute to 100.0 ml with the
291 nm is about 0.78.
mobile phase. Dilute 5.0 ml of this solution to a 50.0 ml with the
Tests mobile phase.
Reference solution. A 0.05 per cent w/v solution of
Acidity. Dissolve 2.0 g in 20 ml of methanol, heating to about
prothionamide RS in the mobile phase. Dilute 5.0 ml of the
50º, and add 20 ml of water. Cool slightly, shake until
solution to 50.0 ml with the mobile phase.
crystallisation occurs, if any and allow to cool to room
temperature. Add 60 ml of water and titrate with 0.1 M sodium Inject the reference solution. The tailing factor is not more
hydroxide using 0.2 ml of cresol red solution as indicator. Not than 2.0, the column efficiency in not less than 5000 theoretical
more than 0.2 ml of 0.1 M sodium hydroxide is required to plates and the relative standard deviation for replicate
change the colour of the indicator to red. injections is not more than 2.0 per cent.
Related substances. Determine by liquid chromatography Inject alternatively the test solution and the reference solution.
(2.4.14). Calculate the content of C9H12N2S.
Test solution. Dissolve 50 mg of the substance under Storage. Store protected from light and moisture.
examination in the mobile phase and dilute to 100 ml with the
mobile phase.
Reference solution. A 0.025 per cent w/v solution of Prothionamide Tablets
prothionamide RS in the mobile phase. Dilute 1 ml of the
Prothionamide Tablets contain not less than 90.0 per cent not
solution to 100 ml with the mobile phase.
more than 110.0 per cent of the stated amount of prothionamide,
Chromatographic system C9H12N2S. The tablets may be coated.
– a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm), Identification
– mobile phase: 60 volumes of a buffer solution prepared
A. Extract a quantity of the powdered tablets containing 25
by mixing 2.0 ml of triethylamine with 1000 ml of water,
mg of Prothionamide with 5 ml of methanol, filter and evaporate
adjusting the pH to 6.0 with dilute orthophosphoric
the filtrate to dryness. The residue complies with the following
acid and 40 volumes of acetonitrile,
test.
– flow rate.1 ml per minute,
– spectrophotometer set at 290 nm, Determine by infrared absorption spectrophotometry (2.4.6).
– a 20 µl loop injector. Compare the spectrum with that obtained with prothionamide
RS.
Inject the reference solution.The test is not valid unless the
relative standard deviation for replicate injections is not more B. In the Assay, the principal peak in the chromatogram
than 2.0 per cent. obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution.
Inject alternatively the test solution and the reference solution.
In the chromatogram obtained with the test solution the area Tests
of any individual impurity peak is not more than the area of
the principal peak in the chromatogram obtained with the Dissolution (2.5.2).
reference solution (0.5 per cent) and the sum of the areas of all Apparatus. No 2
such peaks is not more than twice the area of the principal Medium. 900 ml 0.1 M hydrochloric acid.

1004
IP 2007 PSEUDOEPHEDRINE HYDROCHLORIDE

Speed and time. 100 rpm and 30 minutes. – a 20 µl loop injector.


Withdraw a suitable volume of the medium and filter. Measure Inject the reference solution. The test is not valid unless the
the absorbance of the filtered solution, suitably diluted with tailing factor is not more than 2.0 the column efficiency is not
the dissolution medium if necessary, at the maximum at about less than 5000 theoretical plates and the relative standard
290 nm (2.4.7). Calculate the content of C9H12N2S in the medium deviation for replicate injections is not more than 2.0 per cent
from the absorbance obtained from a solution of known
Inject alternatively the test solution and the reference solution.
concentration of prothionamide RS in the same medium.
Calculate the content of C9H12N2S in the tablets.
D. Not less than 75 per cent of the stated amount of C9H12N2S.
Storage. Store protected from light and moisture.
Related substances. Determine by liquid chromatography
(2.4.14) as described under Assay using the following
solutions.
Test solution. Weigh accurately a quantity of the powdered Pseudoephedrine Hydrochloride
tablets containing 50 mg of Prothionamide disperse in the
mobile phase, shake, dilute to 100 ml with the mobile phase H OH
and filter. CH3
, HCl
Reference solution. A solution containing 0.025 per cent w/v H NHCH3
of prothionamide RS in the mobile phase. Dilute 1 ml of the
solution to 100 ml with the mobile phase.
C10H15NO,HCl Mol. Wt. 201.7
Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more Pseudoephedrine Hydrochloride is (1S,2S)-2-methylamino-
than 2.0 per cent 1-phenylpropan-1-ol hydrochloride.

Inject alternatively the test solution and the reference solution. Pseudoephedrine Hydrochloride contains not less than
In the chromatogram obtained with the test solution the area 99.0 per cent and not more than 101.0 per cent of C10H15NO,
of any individual impurity peak is not more than the area of HCl, calculated on the dried basis.
the peak in the chromatogram obtained with the reference Description. A white, crystalline powder; odourless or almost
solution (0.5 per cent) and the sum of the areas of all such odourless.
impurities is not more than twice the area of the peak in the
chromatogram obtained with the reference solution (1.0 per Identification
cent).
A. Determine by infrared absorption spectrophotometry (2.4.6).
Other tests. Comply with the test stated under the Tablets. Compare the spectrum with that obtained with
Assay. Determine by liquid chromatography (2.4.14). pseudoephedrine hydrochloride RS or with the reference
spectrum of pseudoephedrine hydrochloride.
Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing about 50 mg of B. When examined in the range 230 nm to 360 nm (2.4.7), a
Prothionamide, disperse in the mobile phase, shake and dilute 0.1 per cent w/v solution shows absorption maxima at about
to 100.0 ml with the mobile phase. Dilute 5.0 ml of the resulting 251 nm, 257 nm and 263 nm; absorbances at the maxima, about
solution to 50.0 ml with the mobile phase. 0.75, about 0.98 and about 0.78, respectively.

Reference solution. A 0.05 per cent w/v solution of C. A 5 per cent w/v solution gives the reactions of chlorides
prothionamide RS in the mobile phase. Dilute 5.0 ml of the (2.3.1).
solution to 50.0 ml with the mobile phase.
Tests
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Appearance of solution. A 5.0 per cent w/v solution is not
octadecylsilane bonded to porous silica (5 µm), more than very faintly opalescent and colourless (2.4.1).
– mobile phase: a mixture of 60 volumes of a buffer solution pH (2.4.24). 4.6 to 6.0, determined in a 5.0 per cent w/v solution.
prepared by mixing 2.0 ml of triethylamine with 1000 ml
Specific optical rotation (2.4.22). +61.0º to +62.5º, determined
of water and adjusting the pH to 6.0 with dilute
in a 5.0 per cent w/v solution using a 2-dm tube.
orthophosphoric acid, and 40 volumes of acetonitrile,
– flow rate. 1 ml per minute, Related substances. Determine by thin-layer chromatography
– spectrophotometer set at 290 nm, (2.4.17), coating the plate with silica gel G.

1005
PSEUDOEPHEDRINE SYRUP IP 2007

Mobile phase. A mixture of 40 volumes of butyl acetate, the saame manner or with the reference spectrum of
20 volumes of acetone, 20 volumes of 1-butanol, 10 volumes pseudoephedrine.
of 5 M ammonia and 10 volumes of methanol. B. The residue obtained in test A melts at about 118º (2.4.21).
Test solution. A 10 per cent w/v solution of the substance
C. Dissolve 50 mg of the residue obtained in test A in 10 ml of
under examination in ethanol (95 per cent).
0.1 M hydrochloric acid; it is dextro-rotatory.
Reference solution. A 0.1 per cent w/v solution of the
substance under examination in ethanol (95 per cent). Tests
Apply to the plate 10 µl of each solution. After development, Other tests. Complies with the tests stated under Oral Liquids.
dry the plate in a current of warm air, spray with a solution
Assay. Determine by liquid chromatography (2.4.14).
containing 0.3 g of ninhydrin in a mixture of 100 ml 1-butanol
and 3 ml of glacial acetic acid and heat at 120º for 20 minutes. Test solution. To an accurately measured volume of the syrup
Any secondary spot in the chromatogram obtained with the containing about 0.12 g of Pseudoephedrine Hydrochloride,
test solution is not more intense than the spot in the add 50 ml of 0.1 M hydrochloric acid mix, add sufficient
chromatogram obtained with the reference solution. Ignore 0.1 M hydrochloric acid to produce 100.0 ml, filter and use
any yellow spot near the line of application. the filtrate.
Sulphated ash (2.3.18). Not more than 0.1 per cent. Reference solution. A 0.12 per cent w/v solution of
pseudoephedrine hydrochloride RS in 0.1 M hydrochloric
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
acid.
on 1.0 g by drying in an oven at 105º for 3 hours.
Chromatographic system
Assay. Weigh accurately about 0.5 g, dissolve in a mixture of
– a stainless steel column 25 cm x 4.2 mm, packed with
50 ml of anhydrous glacial acetic acid and 10 ml of mercuric
octadecylsilane bonded to porous silica (5 µm),
acetate solution. Titrate with 0.1 M perchloric acid, using
– mobile phase: a mixture of 85 volumes of ethanol and
crystal violet solution as indicator. Carry out a blank titration.
15 volumes of a 0.4 per cent w/v solution of ammonium
1 ml of 0.1 M perchloric acid is equivalent to 0.02017 g of acetate,
C10H15NO, HCl. – flow rate. 1.5 ml per minute,
Storage. Store protected from light and moisture. – spectrophotometer set at 254 nm,
– a 20 µl loop injector.
Inject the reference solution and record the chromatogram.
The test is not valid unless the relative standard deviation is
Pseudoephedrine Syrup not more than 2.0 per cent and the tailing factor is not more
than 1.5.
Pseudoephedrine Hydrochloride Syrup
Inject alternately the test solution and the reference solution.
Pseudoephedrine Syrup is a solution of Pseudoephedrine
Hydrochloride in a suitable flavoured vehicle. Determine the weight per ml of the syrup (2.4.29), and
calculate the content of C10H15NO, HCl weight in volume.
Pseudoephedrine Syrup contains not less than 90.0 per cent
and not more than 110.0 per cent of the stated amount of Storage. Store protected from light and moisture.
pseudoephedrine hydrochloride, C10H15NO, HCl.

Identification Pseudoephedrine Tablets


A. Shake a quantity of the syrup containing 120 mg of Pseudoephedrine Hydrochloride Tablets
Pseudoephedrine Hydrochloride with two quantities, each of
30 ml, of ether, and discard the ether layer. Add 4 ml of Pseudoephedrine Tablets contain not less than 95.0 per cent
1 M sodium hydroxide to the aqueous layer and extract with and not more than 105.0 per cent of the stated amount of
two quantities, each of 10 ml, of ether. Dry the combined ether pseudoephedrine hydrochloride, C10H15NO, HCl. The tablets
extracts with anhydrous sodium sulphate, filter and evaporate may be coated.
the filtrate to dryness.
Identification
On the residue, determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that A. Shake a quantity of the powdered tablets containing 60 mg
obtained with pseudoephedrine hydrochloride RS treated in of Pseudoephedrine Hydrochloride with 10 ml of water and

1006
IP 2007 PSORALEN

filter. Shake the filtrate with 10 ml of ether, discard the ether Reference solution. A 0.12 per cent w/v solution of
layer. Add 1 ml of 1 M sodium hydroxide to the aqueous layer pseudoephedrine hydrochloride RS in methanol (50 per
and extract with two quantities, each of 10 ml, of ether. Dry the cent).
combined ether extracts with anhydrous sodium sulphate, Chromatographic system
filter and evaporate the filtrate to dryness. – a stainless steel column 20 cm x 4.6 mm, packed with
On the residue, determine by infrared absorption octadecylsilane bonded to porous silica (10 µm),
spectrophotometry (2.4.6). Compare the spectrum with that – mobile phase: 0.005 M dioctyl sodium sulphosuccinate
obtained with pseudoephedrine hydrochloride RS treated in in a mixture of 65 volumes of methanol, 35 volumes of
the same manner or with the reference spectrum of water and 1 volume of glacial acetic acid,
pseudoephedrine. – flow rate. 1.5 ml per minute,
– spectrophotometer set at 258 nm,
B. In the test for Related substances, the principal spot in the
– a 20 µl loop injector.
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution (b). Inject alternately the test solution and the reference solution.

Tests Calculate the content of C10H15NO,HCl in the tablets.


Storage. Store protected from light and moisture.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
Mobile phase. A mixture of 40 volumes of butyl acetate,
20 volumes of acetone, 20 volumes of 1-butanol, 10 volumes Psoralen
of 5 M ammonia and 10 volumes of methanol.
O O O
Test solution (a). Add 25 ml of methanol to a quantity of the
powdered tablets containing 0.5 g of Pseudoephedrine
Hydrochloride, shake for 5 minutes, filter, wash the filter with
methanol and evaporate the combined filtrate and washings C11H6O3 Mol. Wt. 186.1
to dryness. Dissolve the residue as completely as possible in Psoralen is 7H-furo[3,2-g][1]benzopyran-7-one, obtained
5 ml of methanol, centrifuge and use the supernatant liquid. from the fruits of Psoralea carylifolia Linn. (Fam. Legumi-
Test solution (b). Dilute 1 volume of the test solution to nosae) and from the leaves of Ficus carica (Fam.
10 volumes with methanol. Urticaceae) or prepared by synthesis.

Reference solution (a). Dilute 1 volume of the test solution to Psoralen contains not less than 95.0 per cent and not more
100 volumes with methanol. than 101.0 per cent of C11H6O3, calculated on the dried basis.

Reference solution (b). A 1.0 per cent w/v solution of Description. Colourless needles; odourless.
pseudoephedrine hydrochloride RS in methanol.
Identification
Apply to the plate 10 µl of each solution. After development,
dry the plate in a current of warm air, spray with a solution A. Dissolve 1 mg in 5 ml of ethanol (95 per cent) and add
containing 0.3 g of ninhydrin in a mixture of 100 ml 1-butanol 15 ml of a mixture containing 43 volumes of water, 5 volumes
and 3 ml of glacial acetic acid and heat at 120º for 20 minutes. of acetic acid and 3 volumes of propylene glycol; a blue
Any secondary spot in the chromatogram obtained with test fluorescence is visible in ultraviolet light at 365 nm.
solution (a) is not more intense than the spot in the B. Dissolve 1 mg in 2 ml of ethanol (95 per cent) and add
chromatogram obtained with reference solution (a). Ignore 0.1 ml of 0.1 M sodium hydroxide; a yellow fluorescence is
any yellow spot near the line of application. visible in ultraviolet light at 365 nm.
Other tests. Comply with the tests stated under Tablets. Tests
Assay. Determine by liquid chromatography (2.4.14).
Related substances. Determine by thin-layer chromatography
Test solution. Weigh and powder 20 tablets. To a quantity of (2.4.17), coating the plate with silica gel G.
the powdered tablets containing about 0.12 g of
Mobile phase. A mixture of 90 volumes of benzene and 10
Pseudoephedrine Hydrochloride, add 50 ml of 0.1 M
volumes of ethyl acetate.
hydrochloric acid mix with the aid of ultrasound for 15 minutes,
add sufficient methanol to produce 100.0 ml, filter and use the Test solution. Dissolve 0.2 g of the substance under
filtrate. examination in 10 ml of chloroform.

1007
PYRAZINAMIDE IP 2007

Reference solution. Dilute 1 volume of the test solution to 100 range 230 nm to 290 nm, the solution shows an absorption
volumes with chloroform. maximum at about 268 nm; absorbance at about 268 nm,
Apply to the plate 5 µl of each solution. After development, between 0.64 and 0.68.
dry the plate in air and examine in ultraviolet light at 365 nm. C. Boil 20 mg with 5 ml of sodium hydroxide solution; ammonia,
Any secondary spot in the chromatogram obtained with the recognisable by its odour, is evolved.
test solution is not more intense than the spot in the
chromatogram obtained with the reference solution. Tests
Sulphated ash (2.3.18). Not more than 0.1 per cent. Appearance of solution. A 1.0 per cent w/v solution in carbon
dioxide-free water (solution B) is clear (2.4.1), and colourless
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
(2.4.1).
on 1.0 g by drying in an oven at 105º.
Acidity or alkalinity. To 25 ml of solution B add 0.05 ml of
Assay. Weigh accurately about 0.1 g and dissolve in sufficient phenolphthalein solution and 0.2 ml of 0.01 M sodium
methanol to produce 100.0 ml. Dilute 2.0 ml of this solution to hydroxide; the solution is red. Add 1 ml of 0.01 M hydrochloric
100.0 ml with methanol and measure the absorbance of the acid; the solution is colourless. Add 0.15 ml of methyl red
resulting solution at the maximum at about 247 nm (2.4.7). solution; the solution is red.
Calculate the content of C11H6O3 from the absorbance obtained Related substances. Determine by thin-layer chromatography
by repeating the operation using a final solution of 20 µg of (2.4.17), coating the plate with silica gel GF254.
psoralen RS per ml in methanol in place of the substance
under examination. Mobile phase. A mixture of 60 volumes of 1-butanol,
20 volumes of glacial acetic acid and 20 volumes of water.
Storage. Store protected from light and moisture.
Test solution. Dissolve 0.1 g of the substance under
examination in 10 ml of a mixture of 90 volumes of chloroform
and 10 volumes of methanol.
Pyrazinamide Reference solution. A 0.002 per cent w/v solution of the
substance under examination in a mixture of 90 volumes of
O chloroform and 10 volumes of methanol.
N Apply to the plate 20 µl of each solution. After development,
NH2
dry the plate in air and examine in ultraviolet light at 254 nm.
N Any secondary spot in the chromatogram obtained with the
test solution is not more intense than the spot in the
C5H5N3O Mol. Wt. 123.1 chromatogram obtained with the reference solution.
Pyrazinamide is pyrazine-2-carboxamide. Heavy metals (2.3.13). 2.0 g complies with the limit test for
Pyrazinamide contains not less than 99.0 per cent and not heavy metals, Method B (10 ppm).
more than 100.5 per cent of C5H5N3O, calculated on the Sulphated ash (2.3.18). Not more than 0.1 per cent.
anhydrous basis.
Water (2.4.19). Not more than 0.5 per cent, determined on
Description. A white or almost white, crystalline powder; 5.0 g.
odourless or almost odourless.
Assay. Weigh accurately about 0.3 g and transfer to the flask
Identification of an ammonia distillation apparatus. Add 200 ml of water and
75 ml of sodium hydroxide solution. Boil gently for 20 minutes,
Test A may be omitted if tests B and C are carried out. Tests B collecting the distillate in 50.0 ml of 0.05 M sulphuric acid.
and C may be omitted if test A is carried out. Boil vigorously to complete the distillation of the ammonia
A. Determine by infrared absorption spectrophotometry (2.4.6). and titrate the excess of acid with 0.1 M sodium hydroxide,
Compare the spectrum with that obtained with pyrazinamide using methyl red solution as indicator. Repeat the operation
RS or with the reference spectrum of pyrazinamide. without the substance under examination. The difference
between the titrations represents the amount of acid required
B. Dissolve 50 mg in water and dilute to 100 ml with the same to neutralise the ammonia formed.
solvent (solution A). Dilute 1 ml of solution A to 10 ml. When
examined in the range 290 nm to 360 nm (2.4.7), the resulting 1 ml of 0.05 M sulphuric acid is equivalent to 0.01231 g of
solution shows an absorption maximum at about 310 nm. Dilute C5H5N3O.
2 ml of solution A to 100 ml with water. When examined in the Storage. Store protected from moisture.

1008
IP 2007 PYRIDOXINE HYDROCHLORIDE

Pyrazinamide Tablets occasionally, mix with the aid of ultrasound for 10 minutes and
dilute to 500.0 ml with water. Filter and discard the first 20 ml
Pyrazinamide Tablets contain not less than 95.0 per cent and of the filtrate. Dilute 5.0 ml of the filtrate to 100.0 ml with water
not more than 105.0 per cent of the stated amount of and measure the absorbance of the resulting solution at the
pyrazinamide, C5H5N3O. maximum at about 268 nm (2.4.7). Calculate the content of
C5H5N3O taking 650 as the specific absorbance at 268 nm.
Identification
Storage. Store protected from moisture.
A. Shake a quantity of the powdered tablets containing 0.25 g
of Pyrazinamide with 20 ml of ethanol, filter, evaporate the
filtrate to dryness and dry the residue at 105º for 30 minutes.
On the residue, determine by infrared absorption Pyridoxine Hydrochloride
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with pyrazinamide RS or with the reference spectrum
Vitamin B6
of pyrazinamide.
B. Shake a quantity of the powdered tablets containing 50 mg H3C N
of Pyrazinamide with 50 ml of water, dilute to 100 ml with , HCl
OH
water and filter (solution A). Dilute 1 ml of solution A to 10 ml HO
with water. When examined in the range 290 nm to 360 nm
(2.4.7), the resulting solution shows an absorption maximum OH
at about 310 nm. Dilute 2 ml of solution A to 100 ml with water.
When examined in the range 230 nm to 290 nm, the resulting C8H11NO3,HCl Mol. Wt. 205.6
solution shows an absorption maximum at about 268 nm; Pyridoxine Hydrochloride is 5-hydroxy-6-methylpyridine-
absorbance at 268 nm, between 0.64 and 0.68. 3,4-dimethanol hydrochloride.
C. Boil a quantity of the powdered tablets containing 20 mg of Pyridoxine Hydrochloride contains not less than 99.0 per cent
Pyrazinamide with 5 ml of sodium hydroxide solution; and not more than 101.0 per cent of C8H11NO3,HCl, calculated
ammonia, recognisable by its odour, is evolved. on the dried basis.
Tests Description. A white or almost white, crystalline powder;
odourless.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Identification
Mobile phase. A mixture of 60 volumes of 1-butanol,
Test A may be omitted if tests B, C and D are carried out. Tests
20 volumes of glacial acetic acid and 20 volumes of water.
B may be omitted if tests A, C and D are carried out.
Test solution. Shake a quantity of the powdered tablets
A. Determine by infrared absorption spectrophotometry (2.4.6).
containing 0.1 g of Pyrazinamide with 50 ml of a mixture of 90
Compare the spectrum with that obtained with pyridoxine
volumes of chloroform and 10 volumes of methanol, filter,
hydrochloride RS or with the reference spectrum of pyridoxine
evaporate to dryness and dissolve the residue in sufficient of
hydrochloride.
the same solvent mixture to produce 10 ml.
Reference solution. Dilute 1 volume of the test solution to B. When examined in the range 250 nm to 360 nm (2.4.7), a
500 volumes with a mixture of 90 volumes of chloroform and 0.001 per cent w/v solution in 0.1 M hydrochloric acid shows
10 volumes of methanol. an absorption maximum at 288 nm to 296 nm; absorbance at
the maximum, 0.420 to 0.445. A solution prepared by diluting
Apply to the plate 20 µl of each solution. After development, 1 ml of a 0.1 per cent w/v solution in 0.1 M hydrochloric acid
dry the plate in air and examine in ultraviolet light at 254 nm. to 100 ml with 0.025 M standard phosphate buffer shows
Any secondary spot in the chromatogram obtained with the absorption maxima at 248 nm to 256 nm and at 320 nm to
test solution is not more intense than the spot in the 327 nm; absorbances at the maxima, 0.175 to 0.195 and 0.345 to
chromatogram obtained with the reference solution. 0.365, respectively.
Other tests. Comply with the tests stated under Tablets. C. In the test for Related substances, the principal spot in the
Assay. Weigh and powder 20 tablets. Weigh accurately a chromatogram obtained with test solution (b) corresponds to
quantity of the powder containing about 0.1 g of Pyrazinamide, that in the chromatogram obtained with reference solution
add 200 ml of water, allow to stand for 10 minutes, swirling (b).

1009
PYRIDOXINE TABLETS IP 2007

D. A 5 per cent w/v solution gives reaction A of chlorides Pyridoxine Tablets


(2.3.1).
Pyridoxine Hydrochloride Tablets
Tests Pyridoxine Tablets contain not less than 90.0 per cent and not
Appearance of solution. A 5.0 per cent w/v solution is clear more than 110.0 per cent of the stated amount of pyridoxine
(2.4.1), and not more intensely coloured than reference solution hydrochloride, C8H11NO3, HCl.
YS7 (2.4.1).
Identification
pH (2.4.24). 2.4 to 3.0, determined in a 5.0 per cent w/v solution.
A. Shake a quantity of the powdered tablets containing 20 mg
Related substances. Determine by thin-layer chromatography of Pyridoxine Hydrochloride with 50 ml of 0.025 M standard
(2.4.17), coating the plate with silica gel G. phosphate buffer for 15 minutes and dilute to 100 ml with the
Mobile phase. A mixture of 65 volumes of acetone, 13 volumes same solvent. Mix, filter and dilute 5 ml of the filtrate to 100 ml
of dichloromethane, 13 volumes of tetrahydrofuran and with the same solvent. When examined in the range 230 nm to
9 volumes of strong ammonia solution. 360 nm (2.4.7), the resulting solution exhibits two maxima, at
about 254 nm and 324 nm.
Test solution (a). A 10 per cent w/v solution of the substance
B. Triturate a quantity of the powdered tablets containing
under examination in water.
20 mg of Pyridoxine Hydrochloride with 50 ml of water and
Test solution (b). A 1 per cent w/v solution of the substance allow to stand for 20 minutes. To 1 ml of the supernatant liquid
under examination water. add 10 ml of a 5 per cent w/v solution of sodium acetate, 1 ml
Reference solution (a). A 0.025 per cent w/v solution of the of water and 1 ml of a 0.5 per cent w/v solution of 2,6-di-
substance under examination in water. chloroquinone-4-chloroimide in ethanol (95 per cent) and
shake; a blue colour is produced which fades rapidly and
Reference solution (b). A 1 per cent w/v solution of pyridoxine becomes brown. Repeat the operation but adding 1 ml of a
hydrochloride RS in water. 0.3 per cent w/v solution of boric acid in place of 1 ml of
Apply to the plate 2 µl of each solution. After development, water; no blue colour is produced.
dry the plate in air and spray with a 5 per cent w/v solution of Tests
sodium carbonate in a mixture of 70 volumes of water and
30 volumes of ethanol (95 per cent). Dry in a current of air, Related substances. Determine by thin-layer chromatography
spray with a 0.1 per cent w/v solution of 2,6-dichloroquinone- (2.4.17), coating the plate with silica gel G.
4-chloroimide in ethanol (95 per cent) and examine Mobile phase. A mixture of 65 volumes of acetone, 13 volumes
immediately. Any secondary spot in the chromatogram of dichloromethane, 13 volumes of tetrahydrofuran and
obtained with test solution (a) is not more intense than the 9 volumes of strong ammonia solution.
spot in the chromatogram obtained with reference solution
Test solution. Shake a quantity of the powdered tablets
(a). Ignore any spots remaining on the line of application.
containing 40 mg of Pyridoxine Hydrochloride with 10 ml of
Heavy metals (2.3.13). 12 ml of a 5.0 per cent w/v solution water for 15 minutes, filter and use the filtrate.
complies with the limit test for heavy metals, Method D
Reference solution. Dilute 1 ml of test solution to 200 ml with
(20 ppm). Use lead standard solution (1 ppm Pb) to prepare
water.
the standard.
Apply to the plate 10 µl of each solution. After development,
Sulphated ash (2.3.18). Not more than 0.1 per cent. dry the plate in air and spray with a 5 per cent w/v solution of
Loss on drying (2.4.19). Not more than 0.5 per cent, determined sodium carbonate in a mixture of 70 volumes of water and
on 1.0 g by drying in an oven at 105º. 30 volumes of ethanol (95 per cent). Dry it in a current of air,
spray with a 0.1 per cent w/v solution of 2,6-dichloroquinone-
Assay. Weigh accurately about 0.15 g, dissolve in a mixture of
4-chloroimide in ethanol (95 per cent) and examine
5 ml of anhydrous glacial acetic acid and 6 ml of mercuric
immediately. Any secondary spot in the chromatogram
acetate solution. Titrate with 0.1 M perchloric acid, using
obtained with the test solution is not more intense than the
crystal violet solution as indicator, until a green colour is
spot in the chromatogram obtained with the reference solution.
produced. Carry out a blank titration.
Uniformity of content. Comply with the test stated under
1 ml of 0.1 M perchloric acid is equivalent to 0.02056 g of Tablets.
C8H11NO3, HCl.
Powder one tablet, add 50 ml of 0.1 M hydrochloric acid and
Storage. Store protected from moisture. heat on a water-bath for 15 minutes, swirling occasionally.

1010
IP 2007 PYRIMETHAMINE

Cool, dilute to 100.0 ml with 0.1 M hydrochloric acid, filter, C. Dissolve 0.14 g in sufficient ethanol to produce 100 ml,
discarding the first 20 ml of the filtrate. If necessary, dilute dilute 10 ml of this solution to 100 ml with 0.1 M hydrochloric
quantitatively and stepwise with 0.1 M hydrochloric acid to acid and dilute 10 ml of the solution to 100 ml with the same
produce a solution containing 10 µg of the pyridoxine solvent. When examined in the range 230 nm to 360 nm (2.4.7),
hydrochloride per ml and measure the absorbance of the the resulting solution shows an absorption maximum at about
resulting solution at the maximum at about 290 nm (2.4.7). 272 nm; absorbance at about 272 nm, 0.43 to 0.46.
Calculate the content of C8H11NO3, HCl taking 430 as the
specific absorbance at 290 nm. Tests
Other tests. Comply with the tests stated under Tablets. Appearance of solution. Dissolve 0.25 g in sufficient of a mixture
Assay. Weigh and powder 20 tablets. Weigh accurately a of 3 volumes of dichloromethane and 1 volume of methanol
quantity of the powder containing about 20 mg of Pyridoxine to produce 10 ml. The resulting solution is clear (2.4.1), and
Hydrochloride, add 50 ml of 0.1 M hydrochloric acid and not more intensely coloured than reference solution BYS6
heat on a water-bath for 15 minutes, swirling occasionally. (2.4.1).
Cool, dilute to 100.0 ml with 0.1 M hydrochloric acid and Acidity or alkalinity. Shake 1.0 g with 50 ml of water for
filter, discarding the first 20 ml of the filtrate. Dilute 5.0 ml of 2 minutes and filter (solution A). To 10 ml of solution A add
the filtrate to 100.0 ml with 0.1 M hydrochloric acid and 0.05 ml of phenolphthalein solution; the solution is colourless
measure the absorbance of the resulting solution at the and not more than 0.2 ml of 0.01 M sodium hydroxide is
maximum at about 290 nm (2.4.7). Calculate the content of required to change the colour to pink. Add 0.4 ml of 0.01 M
C8H11NO3, HCl taking 430 as the specific absorbance at hydrochloric acid and 0.05 ml of methyl red solution; the
290 nm. solution is red or orange.
Storage. Store protected from light and moisture. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 76 volumes of toluene, 12 volumes
Pyrimethamine of glacial acetic acid, 8 volumes of 1-propanol and 4 volumes
of chloroform.
N NH2
H3C Prepare the following solutions immediately before use.
N Test solution (a). Dissolve 0.1 g of the substance under
examination in 10 ml of a mixture of 90 volumes of chloroform
NH2 and 10 volumes of methanol.
Cl
C12H13ClN4 Mol. Wt. 248.7 Test solution (b). Dissolve 0.1 g of the substance under
examination in 100 ml of the same solvent mixture.
Pyrimethamine is 5-(4-chlorophenyl)-6-ethylpyrimidine-2,4-
diamine. Reference solution (a). A 0.0025 per cent w/v solution of the
substance under examination in the same solvent mixture.
Pyrimethamine contains not less than 99.0 per cent and not
more than 101.0 per cent of C12H13ClN4, calculated on the dried Reference solution (b). A 0.1 per cent w/v solution of
basis. pyrimethamine RS in the same solvent mixture.

Description. Colourless crystals or an almost white, crystalline Apply to the plate 20 µl of each solution. After development,
powder; odourless. dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with test
Identification solution (a) is not more intense than the spot in the
chromatogram obtained with reference solution (a).
Test A may be omitted if tests B and C are carried out. Tests B
and C may be omitted if test A is carried out. Sulphates (2.3.17). 25 ml of Solution A complies with the limit
test for sulphates. Use a mixture of 5.0 ml of sulphate standard
A. Determine by infrared absorption spectrophotometry (2.4.6).
solution (10 ppm SO4) and 10 ml of distilled water to prepare
Compare the spectrum with that obtained with pyrimethamine
the standard (100 ppm).
RS or with the reference spectrum of pyrimethamine.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Loss on drying (2.4.19). Not more than 0.5 per cent, determined
that in the chromatogram obtained with reference solution (b). on 1.0 g by drying in an oven at 105º for 4 hours.

1011
PYRIMETHAMINE AND SULPHADOXINE TABLETS IP 2007

Assay. Weigh accurately about 0.2 g, dissolve in 25 ml of 2.0 ml of solution A prepared by dissolving 0.1 g of phenacetin
anhydrous glacial acetic acid, heating gently, cool. Titrate (internal standard) in 100 ml of acetonitrile and sufficient of
with 0.1 M perchloric acid, determining the end-point the mobile phase to produce 50.0 ml.
potentiometrically (2.4.25). Carry out a blank titration. Reference solution. Weigh accurately about 25 mg of
1 ml of 0.1 M perchloric acid is equivalent to 0.02487 g of pyrimethamine RS and 500 mg of sulphadoxine RS, add 35 ml
C12H13ClN4. of acetonitrile and sufficient of the mobile phase to produce
100.0 ml and mix. To 25.0 ml add 2.0 ml of solution A and add
Storage. Store protected from light and moisture.
sufficient of the mobile phase to produce 50.0 ml and mix well.
Chromatographic system
– a stainless steel column 30 cm x 4 mm, packed with
Pyrimethamine and Sulphadoxine octadecylsilane bonded to porous silica (5 µm),
Tablets – mobile phase: a mixture of 4 volumes of a 1 per cent v/v
solution of glacial acetic acid and 1 volume of
Pyrimethamine and Sulphadoxine Tablets contain not less than acetonitrile,
90.0 per cent and not more than 110.0 per cent of the stated – flow rate. 2 ml per minute,
amounts of pyrimethamine, C12H13ClN4, and of sulphadoxine, – spectrophotometer set at 254 nm,
C12H14N4O4S. – a 20 µl loop injector.
Identification Inject the reference solution. The relative standard deviation
for replicate injections is not more than 2.0 percent.
Determine by thin-layer chromatography (2.4.17), coating the
plate with silica gel G. Inject alternately the test solution and the reference solution.
The relative retention times should be about 1.3 for
Mobile phase. A mixture of 4 volumes of chloroform, pyrimethamine, 1.0 for phenacetin and 0.7 for sulphadoxine.
4 volumes of n-heptane, 1 volume of glacial acetic acid and
4 volumes of a mixture of 1 volume of methanol and 19 volumes Calculate the content of C12H13ClN4 and the content of
of ethanol. C12H14N4O4S in the tablets.
Test solution. Shake a quantity of the powdered tablets Storage. Store protected from light and moisture.
containing 25 mg of Pyrimethamine with 50 ml of a 2 per cent
w/v solution of strong ammonia solution in methanol.
Reference solution (a). A 0.05 per cent w/v solution of
pyrimethamine RS in methanol.
Reference solution (b). A 1.0 per cent w/v solution of
sulphadoxine RS in methanol.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
One of the principal spots in the chromatogram obtained with
the test solution corresponds to the spot in the chromatogram
obtained with reference solution (a) and the other corresponds
to that in the chromatogram obtained with reference solution
(b).

Tests
Other tests. Complies with the tests stated under Tablets.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh accurately a quantity of the powdered
tablets containing about 25 mg of Pyrimethamine and 500 mg
of Sulphadoxine and shake with 35 ml of acetonitrile for
30 minutes in a 100-ml volumetric flask. Dilute to volume with
the mobile phase, mix and filter. To 25.0 ml of the filtrate add

1012
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

Q
Quinalbarbitone Sodium ....
Quinalbarbitone Tablets ....
Quinidine Sulphate ....
Quinidine Tablets ....
Quinine Bisulphate ....
Quinine Bisulphate Tablets ....
Quinine Dihydrochloride ....
Quinine Dihydrochloride Injection ....
Quinine Sulphate ....
Quinine Tablets ....
Quiniodochlor ....
Quiniodochlor Tablets ....

1013
IP 2007 QUINALBARBITONE SODIUM

Quinalbarbitone Sodium Tests


Quinalbarbital Sodium; Secobarbital Sodium; Appearance of solution. A freshly prepared 10.0 per cent w/v
Secobarbitone Sodium; Soluble Quinalbarbitone solution in ethanol (95 per cent) is clear (2.4.1), and not more
intensely coloured than reference solution YS7 (2.4.1).
H pH (2.4.24). Not more than 11.0, determined on a 10.0 per cent
O N ONa w/v solution.
H2C
N Heavy metals (2.3.13). 0.67 g dissolved in a mixture of 5 ml of
H3C 1 M sodium hydroxide and 20 ml of water complies with the
O limit test for heavy metals, Method C (30 ppm).
H3C
Related substances. Complies with the test for related
C12H17N2NaO3 Mol.Wt.260.3 substances in barbiturates (2.3.4).
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
Quinalbarbitone Sodium is sodium (RS)-5-allyl-5-(1- on 0.5 g by drying in an oven at 105º.
methylbutyl)barbiturate.
Assay. Weigh accurately about 0.5 g, dissolve in 10 ml of
Quinalbarbitone Sodium contains not less than 98.5 per cent ethanol, add 10 ml of silver nitrate-pyridine reagent and titrate
and not more than 102.0 per cent of C12H17N2NaO3, calculated with 0.1 M ethanolic sodium hydroxide using 0.5 ml of
on the dried basis. thymolphthalein solution as indicator, until a pure blue colour
Description. A white powder; odourless; hygroscopic. is obtained. Carry out a blank titration.
1 ml of 0.1 M ethanolic sodium hydroxide is equivalent to
Identification 0.02603 g of C12H17N2NaO3.
Test A may be omitted if tests B, C, D and E are carried out. Storage. Store protected from moisture.
Tests B, C and D may be omitted if tests A and E are carried
out.
A. To 10 ml of a 10.0 per cent w/v solution in ethanol (95 per Quinalbarbitone Tablets
cent) add 120 ml of water and 5 ml of 2 M acetic acid, stir
vigorously, add 200 ml of water and boil until the precipitate Quinalbarbital Sodium Tablets; Quinalbarbitone
dissolves and no oily particles remain on the surface of the Sodium Tablets; Secobarbital Sodium Tablets;
liquid. Allow to cool until a haziness begins to appear in the Secobarbitone Sodium Tablets
solution, induce crystallisation, if necessary and allow the
solution to stand for 12 hours. Wash the crystals with three Quinalbarbitone Tablets contain not less than 92.5 per cent
quantities, each of 10 ml, of water and dry the residue at 80º. and not more than 107.5 per cent of the stated amount of
quinalbarbitone sodium, C12H17N2NaO3. The tablets are coated.
On the residue determine by infrared absorption
spectrophotometry (2.4.6). Compare the spectrum with that Identification
obtained with quinalbarbitone RS treated in the same manner.
A. Shake a quantity of the powdered tablets containing about
B. To 0.5 g add 5 ml of sodium carbonate solution and 10 ml of
0.5 g of Quinalbarbitone Sodium with 10 ml of water and filter.
nitrobenzyl chloride solution. Heat for 30 minutes on a water-
To 10 ml of the filtrate add 5 ml of sodium carbonate solution
bath under reflux and allow to stand for 1 hour. Filter, wash the
and 10 ml of nitrobenzyl chloride solution. Heat for 30 minutes
precipitate successively with 10 ml of dilute sodium hydroxide
on a water-bath under reflux and allow to stand for 1 hour.
solution and 50 ml of water; recrystallise from a mixture of
Filter, wash the precipitate successively with 10 ml of dilute
equal volumes of chloroform and ethanol (95 per cent) and
sodium hydroxide solution and 50 ml of water; recrystallise
dry at 105º. The crystals melt at about 156º (2.4.21)
from a mixture of equal volumes of chloroform and ethanol
C. Complies with the test for identification of barbiturates (95 per cent) and dry at 105º. The crystals melt at about
(2.3.2). 156º (2.4.21)
D. Gives the reaction of non-nitrogen substituted barbiturates B. Triturate a quantity of the powdered tablets containing
(2.3.1). 0.5 g of Quinalbarbitone Sodium with 10 ml of water, filter,
E. Ignite 1 g; the residue gives the reactions of sodium salts acidify the filtrate with acetic acid; oily drops are formed
(2.3.1). which may eventually crystallise.

1015
IP 2007

C. The powdered tablets give the reactions of sodium salts Mobile phase. A mixture of 60 volumes of toluene, 36 volumes
(2.3.1). of ether and 15 volumes of diethylamine.
Tests Test solution. Dissolve 1 g of the substance under examination
in 100 ml of methanol.
Other tests. Comply with the tests stated under Tablets.
Reference solution (a). A 1 per cent w/v solution of quinidine
Assay. Weigh and digest 20 tablets with 50 ml of water until
sulphate RS in methanol.
completely disintegrated and not more than a small residue
remains. Add 5 ml of 1 M sodium hydroxide, filter and wash Reference solution (b). A 1 per cent w/v solution of each of
the residue with sufficient water to produce 100.0 ml. Extract a quinidine sulphate RS and quinine sulphate RS in methanol.
volume of the solution containing 0.5 g of Quinalbarbitone
Apply to the plate 4 µl of each solution. After development,
Sodium with two quantities, each of 10 ml, of ether, washing
dry the plate in air for 15 minutes and repeat the development.
each ether extract with the same 3 ml of water. Add the water
Dry the plate at 105º for 30 minutes, allow to cool and spray
to the aqueous liquid, acidify with hydrochloric acid and
with potassium iodoplatinate solution. The principal spot in
extract with successive quantities, each of 15 ml, of ether until
the chromatogram obtained with the test solution corresponds
complete extraction is effected. Wash the combined extracts
to the spot in the chromatogram obtained with reference
with two quantities, each of 2 ml, of water and extract the
solution (a). The test is not valid unless the chromatogram
combined washings with 10 ml of ether. Add the ether to the
obtained with reference solution (b) shows two clearly
main ether layer, filter and wash the filter with ether. Evaporate
separated spots.
the solvent and dry the residue to constant weight at 50º.
B. To 5 ml of a 0.1 per cent w/v solution add 0.2 ml of bromine
1 g of residue is equivalent to 1.092 g of C12H17N2NaO3.
solution and 1 ml of dilute ammonia solution; an emerald-
Storage. Store protected from moisture. green colour is produced.
C. To a 0.5 per cent w/v solution add an equal volume of dilute
sulphuric acid; a strong blue fluorescence is produced.
Quinidine Sulphate
D. To 5 ml of a 1 per cent w/v solution add 1 ml of silver nitrate
Quinidine Bisulphate solution and stir with a glass rod; after a short interval, a
white precipitate soluble in nitric acid is produced (distinction
from many other alkaloids).
OCH3
E. A 1 per cent w/v solution gives the reactions of sulphates
H (2.3.1).
HO H H
CH2 , H2SO4, 2H2O
N
Tests
N H H
Appearance of solution. A 2.0 per cent w/v solution in 0.1 M
2
hydrochloric acid is clear (2.4.1), and not more intensely
coloured than reference solution GYS4 (2.4.1).
(C20H24N2O2)2,H2SO4,2H2O Mol. Wt. 783.0 pH (2.4.24). 6.0 to 6.8, determined in a 1.0 per cent w/v solution.
Quinidine Sulphate is (8R,9S)-6′-methoxycinchonan-9-ol
Specific optical rotation (2.4.22). +275º to +290º, determined
sulphate dihydrate. The alkaloid is obtained from the bark of
in a 2.0 per cent w/v solution in 0.1 M hydrochloric acid.
various species of Cinchona and from Remijia pedunculata
Fluckiger (Fam. Rubiaceae) or prepared from quinine. Dihydroquinidine sulphate. Not more than 15.0 per cent,
calculated on the dried basis and determined by the following
Quinidine Sulphate contains not less than 99.0 per cent and
method. Dissolve 0.2 g in 20 ml of water, add 0.5 g of potassium
not more than 101.5 per cent of alkaloid monosulphates,
bromide and 15 ml of 2 M hydrochloric acid. Titrate slowly
calculated as (C20H24N2O2)2,H2SO4 on the dried basis.
with 0.0167 M potassium bromate using methyl red solution
Description. A white or almost white, crystalline powder or as indicator until a yellow colour is obtained. Add a solution
needle-like crystals; odourless. of 0.5 g of potassium iodide in 200 ml of water and stopper the
flask immediately. Allow to stand in the dark for 5 minutes and
Identification titrate with 0.1 M sodium thiosulphate using starch solution,
A. Determine by thin-layer chromatography (2.4.17), coating added towards the end of the titration, as indicator. Repeat
the plate with silica gel G. the operation without the substance under examination.

1016
IP 2007

1 ml of 0.0167 M potassium bromate is equivalent to with a retention time relative to quinine of about 1.4. The
0.01867 g of (C20H24N2O2)2, H2SO4. chromatogram obtained with reference solution (b) shows a
principal peak due to quinidine and a peak due to
Calculate the content of dihydroquinidine sulphate by
dihydroquinidine, with a retention time relative to quinidine
subtracting the result from the assay result.
of about 1.2. The chromatogram obtained with reference
Other cinchona alkaloids. Determine by liquid solution (c) shows four peaks due to quinine, dihydroquinine,
chromatography (2.4.14). quinidine and dihydroquinidine which are identified by
Test solution. Dissolve 20 mg of the substance under comparison of their retention times with those of the
examination in 5 ml of the mobile phase. Heat gently, if corresponding peaks in the chromatograms obtained with
necessary to dissolve the powder as completely as possible, reference solutions (a) and (b).
cool, dilute to 10 ml with the mobile phase and mix. The test is not valid unless (a) in the chromatogram obtained
with reference solution (c) the resolution between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate
due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile
between the peaks due to dihydroquinidine and quinine is at
phase and dilute to 10 ml with the mobile phase.
least 1.0 and (b) the signal-to-noise ratio of the principal peak
Reference solution (b). Prepare in the same manner as in the chromatogram obtained with reference solution (d) is at
reference solution (a) but using quinidine sulphate RS in least 5.
place of quinine sulphate RS.
Inject the test solution and allow the chromatography to
Reference solution (c). Mix equal volumes of reference proceed for 2.5 times the retention time of the principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
Reference solution (d). Dilute 1 volume of reference solution normalisation, ignoring any peaks the areas of which are less
(a) to 10 volumes with the mobile phase and dilute 1 volume of than that of the peak in the chromatogram obtained with
the resulting solution to 50 volumes with the mobile phase. reference solution (d) (0.2 per cent). The content of
dihydroquinidine is not greater than 15 per cent, the content
Reference solution (e). A solution containing 0.1 per cent of any related substance eluting before quinidine is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance
Chromatographic system is not greater than 2.5 per cent.
– a stainless steel column 25 cm x 4.6 mm packed with Sulphated ash (2.3.18). Not more than 0.1 per cent.
octadecylsilane bonded to porous silica (5 µm) (such as
Loss on drying (2.4.19). 3.0 per cent to 5.0 per cent, determined
Hypersil ODS 5 µm),
on 1.0 g by drying in an oven at 130º.
– mobile phase: a solution prepared by dissolving 6.8 g of
potassium dihydrogen orthophosphate and 3.0 g of Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
hexylamine in 700 ml of water, adjusting the pH to 2.8 10 ml of chloroform and 20 ml of acetic anhydride. Titrate
with 1 M orthophosphoric acid, adding 60 ml of with 0.1 M perchloric acid, determining the end-point
acetonitrile and diluting to 1000 ml with water, potentiometrically (2.4.25). Carry out a blank titration.
– flow rate. 1.5 ml per minute, 1 ml of 0.1 M perchloric acid is equivalent to 0.02490 g of
– spectrophotometer set at 250 nm for reference solution (C20H24N2O2)2,H2SO4.
(e) and 316 nm for the other solutions,
– a 10 µl loop injector. Storage. Store protected from light.
Inject separately reference solutions (b) and (e). If necessary,
adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b) Quinidine Tablets
the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0
(the distance along the baseline between the point of injection Quinidine Sulphate Tablets
and the perpendicular dropped from the maximum of the peak Quinidine Tablets contain not less than 95.0 per cent and not
of an unretained component) being calculated from the peak more than 105.0 per cent of the stated amount of quinidine
due to thiourea in the chromatogram obtained with reference sulphate, (C20H24N2O2)2,H2SO4,2H2O.
solution (e).
Identification
Inject reference solutions (a), (b), (c) and (d). The
chromatogram obtained with reference solution (a) shows a A. Determine by thin-layer chromatography (2.4.17), coating
principal peak due to quinine and a peak due to dihydroquinine the plate with silica gel G.

1017
IP 2007

Mobile phase. A mixture of 80 volumes of toluene, 20 volumes Reference solution (b). Prepare in the same manner as
of acetone and 10 volumes of diethylamine. reference solution (a) but using quinidine sulphate RS in
place of quinine sulphate RS.
Test solution. Extract a quantity of the powdered tablets
containing 0.1 g of Quinidine Sulphate with 10 ml of a mixture Reference solution (c). Mix equal volumes of reference
of 2 volumes of chloroform and 1 volume of ethanol (95 per solutions (a) and (b).
cent) and filter. Reference solution (d). Dilute 1 volume of reference solution
Reference solution. 1.0 per cent w/v solution of quinidine (a) to 10 volumes with the mobile phase and dilute 1 volume of
sulphate RS in a mixture of 2 volumes of chloroform and the resulting solution to 50 volumes with the mobile phase.
1 volume of ethanol (95 per cent). Reference solution (e). A solution containing 0.1 per cent
Apply to the plate 2 µl of each solution. After development, w/v of thiourea in the mobile phase.
dry the plate in air and spray with 0.05 M ethanolic sulphuric Chromatographic system
acid and then with dilute potassium iodobismuthate solution. – a stainless steel column 25 cm x 4.6 mm packed with
The principal spot in the chromatogram obtained with the test octadecylsilane bonded to porous silica (5 µm) (such as
solution corresponds to that in the chromatogram obtained Hypersil ODS 5 µm),
with the reference solution. – mobile phase: a solution prepared by dissolving 6.8 g of
B. Extract a quantity of the powdered tablets containing 0.1 g potassium dihydrogen orthophosphate and 3.0 g of
of Quinidine Sulphate with 20 ml of water and filter. The filtrate hexylamine in 700 ml of water, adjusting the pH to 2.8
(solution A) is dextro-rotatory. with 1 M orthophosphoric acid, adding 60 ml of
acetonitrile and diluting to 1000 ml with water,
C. To 1 ml of solution A add 4 ml of water, 2 or 3 drops of – flow rate. 1.5 ml per minute,
bromine solution and 1 ml of dilute ammonia solution; an – spectrophotometer set at 250 nm for reference solution
emerald green colour is produced. (e) and 316 nm for the other solutions,
D. Solution A gives the reactions of sulphates (2.3.1). – a 10 µl loop injector.
Inject separately reference solutions (b) and (e). If necessary,
Tests adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b)
Dissolution (2.5.2). the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0
Apparatus. No 2 (the distance along the baseline between the point of injection
Medium. 900 ml of 0.1 M hydrochloric acid and the perpendicular dropped from the maximum of the peak
Speed and time. 100 rpm and 30 minutes. of an unretained component) being calculated from the peak
due to thiourea in the chromatogram obtained with reference
Withdraw a suitable volume of the medium and filter. Measure solution (e).
the absorbance of the filtrate, suitably diluted if necessary, at
the maximum at about 248 nm (2.4.7). Calculate the content of Inject reference solutions (a), (b), (c) and (d). The
(C20H24N2O2)2,H2SO4,2H2O in the medium from the absorbance chromatogram obtained with reference solution (a) shows a
obtained from a solution of known concentration of quinidine principal peak due to quinine and a peak due to dihydroquinine
sulphate RS. with a retention time relative to quinine of about 1.4. The
chromatogram obtained with reference solution (b) shows a
D. Not less than 70 per cent of the stated amount of principal peak due to quinidine and a peak due to
(C20H24N2O2)2,H2SO4,2H2O. dihydroquinidine, with a retention time relative to quinidine
Other cinchona alkaloids. Determine by liquid of about 1.2. The chromatogram obtained with reference
chromatography (2.4.14). solution (c) shows four peaks due to quinine, dihydroquinine,
quinidine and dihydroquinidine which are identified by
Test solution. Mix a quantity of the powdered tablets comparison of their retention times with those of the
containing 50 mg of Quinidine Sulphate with 20 ml of the corresponding peaks in the chromatograms obtained with
mobile phase. Heat gently to dissolve the powder as completely reference solutions (a) and (b).
as possible, cool, dilute to 25 ml with the mobile phase and
The test is not valid unless (a) in the chromatogram obtained
filter, discarding the first few ml of filtrate.
with reference solution (c) the resolution between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile between the peaks due to dihydroquinidine and quinine is at
phase and dilute to 10 ml with the mobile phase. least 1.0 and (b) the signal-to-noise ratio of the principal peak

1018
IP 2007

in the chromatogram obtained with reference solution (d) is at Mobile phase. A mixture of 60 volumes of toluene, 36 volumes
least 5. of ether and 15 volumes of diethylamine.
Inject the test solution and allow the chromatography to Test solution. Dissolve 1 g of the substance under examination
proceed for 2.5 times the retention time of the principal peak. in 100 ml methanol.
Calculate the percentage content of related substances by
normalisation, ignoring any peaks the areas of which are less Reference solution (a). A 1 per cent w/v solution of quinine
than that of the peak in the chromatogram obtained with sulphate RS in methanol.
reference solution (d) (0.2 per cent). The content of Reference solution (b). A 1 per cent w/v solution of each of
dihydroquinidine is not greater than 15 per cent, the content quinidine sulphate RS and quinine sulphate RS in methanol.
of any related substance eluting before quinidine is not greater
than 5 per cent and the content of any other related substance Apply to the plate 4 µl of each solution. After development,
is not greater than 2.5 per cent. dry the plate in air for 15 minutes and repeat the development.
Dry the plate at 105º for 30 minutes, allow to cool and spray
Other tests. Comply with the tests stated under Tablets. with potassium iodoplatinate solution. The principal spot in
Assay. Weigh and powder 20 tablets. Weigh accurately a the chromatogram obtained with the test solution corresponds
quantity of the powder containing about 0.4 g of Quinidine to the spot in the chromatogram obtained with reference
Sulphate, dissolve as completely as possible in 40 ml of acetic solution (a). The test is not valid unless the chromatogram
anhydride with the aid of heat and cool. Titrate with 0.1 M obtained with reference solution (b) shows two clearly
perchloric acid, using crystal violet solution as indicator. separated spots.
Carry out a blank titration. B. A 5 per cent w/v solution has a blue fluorescence.
1 ml of 0.1 M perchloric acid is equivalent to 0.02610 g of C. A 5 per cent w/v solution gives the reactions of sulphates
(C20H24N2O2)2,H2SO4,2H2O. (2.3.1).
Storage. Store protected from light.
Tests
pH (2.4.24). 2.8 to 3.4, determined in a 1.0 per cent w/v solution.
Quinine Bisulphate Specific optical rotation (2.4.22). –208º to –216º, determined
at 20º in a 3.0 per cent w/v solution in 0.1 M hydrochloric
Quinine Acid Sulphate acid.
Other cinchona alkaloids. Determine by liquid
OCH3 chromatography (2.4.14).
H
HO H H Test solution. Dissolve 20 mg of the substance under
CH2 , H2SO4, 7H2O examination in 5 ml of the mobile phase. Heat gently, if
N
necessary to dissolve the powder as completely as possible,
N H H
cool, dilute to 10 ml with the mobile phase and mix.
Reference solution (a). Dissolve 20 mg of quinine sulphate
C20H24N2O2,H2SO4,7H2O Mol. Wt. 548.6
RS, with gentle heating if necessary, in 5 ml of the mobile
Quinine Bisulphate is (8S,9R)-6′-methoxycinchonan-9-ol phase and dilute to 10 ml with the mobile phase.
hydrogen sulphate heptahydrate. The alkaloid is obtained
from the bark of various species of Cinchona. Reference solution (b). Prepare in the same manner as
reference solution (a) but using quinidine sulphate RS in
Quinine Bisulphate contains not less than 98.5 per cent and place of quinine sulphate RS.
not more than 101.5 per cent of alkaloid hydrogen sulphates,
calculated as C20H24N2O2,H2SO4 on the anhydrous basis. Reference solution (c). Mix equal volumes of reference
solutions (a) and (b).
Description. Colourless or faintly yellow crystals or a white
or faintly yellow, crystalline powder; efflorescent in dry air. Reference solution (d). Dilute 1 volume of reference solution
(a) to 10 volumes with the mobile phase and dilute 1 volume of
Identification the resulting solution to 50 volumes with the mobile phase.
A. Determine by thin-layer chromatography (2.4.17), coating Reference solution (e). A solution containing 0.1 per cent
the plate with silica gel G. w/v of thiourea in the mobile phase.

1019
IP 2007

Chromatographic system Titratable cation. 75.3 to 79.6 per cent, calculated on the
– a stainless steel column 25 cm x 4.6 mm packed with anhydrous basis, determined by the following method. Titrate
octadecylsilane bonded to porous silica (5 µm) (such as the combined aqueous solutions reserved in the Assay with
Hypersil ODS 5 µm), 0.1 M hydrochloric acid using 0.1 ml of phenolphthalein
– mobile phase: a solution prepared by dissolving 6.8 g of solution as indicator.
potassium dihydrogen orthophosphate and 3.0 g of 1 ml of 0.1 M sodium hydroxide is equivalent to 0.01632 g of
hexylamine in 700 ml of water, adjusting the pH to 2.8 [C20H26N2O2]2+.
with 1 M orthophosphoric acid, adding 60 ml of
acetonitrile and diluting to 1000 ml with water, Sulphated ash (2.3.18). Not more than 0.1 per cent.
– flow rate. 1.5 ml per minute, Water (2.3.43). 19.0 to 25.0 per cent, determined on 0.2 g.
– spectrophotometer set at 250 nm for reference solution
Assay. Weigh accurately about 0.45 g, dissolve in 15 ml of
(e) and 316 nm for the other solutions,
water, add 25 ml of 0.1 M sodium hydroxide and extract with
– a 10 µl loop injector.
three quantities, each of 25 ml, of chloroform. Wash each
Inject separately reference solutions (b) and (e). If necessary, chloroform extract with the same 20 ml of water. Combine the
adjust the concentration of acetonitrile in the mobile phase so aqueous solutions and reserve for the test for Titratable cation.
that in the chromatogram obtained with reference solution (b) Dry the chloroform extracts with anhydrous sodium sulphate,
the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0 evaporate to dryness at a pressure of 2 kPa and dissolve the
(the distance along the baseline between the point of injection residue in 50 ml of anhydrous glacial acetic acid. Titrate with
and the perpendicular dropped from the maximum of the peak 0.1 M perchloric acid, using crystal violet solution as
of an unretained component) being calculated from the peak indicator. Carry out a blank titration.
due to thiourea in the chromatogram obtained with reference
1 ml of 0.1 M perchloric acid is equivalent to 0.02113 g of
solution (e).
C20H24N2O2,H2SO4.
Inject reference solutions (a), (b), (c) and (d). The
chromatogram obtained with reference solution (a) shows a Storage. Store protected from light.
principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 1.4. The
chromatogram obtained with reference solution (b) shows a Quinine Bisulphate Tablets
principal peak due to quinidine and a peak due to
Quinine Acid Sulphate Tablets
dihydroquinidine, with a retention time relative to quinidine
of about 1.2. The chromatogram obtained with reference Quinine Bisulphate Tablets contain not less than 95.0 per cent
solution (c) shows four peaks due to quinine, dihydroquinine, and not more than 105.0 per cent of the stated amount of
quinidine and dihydroquinidine which are identified by quinine bisulphate, C20H24N2O2,H2SO4,7H2O. The tablets are
comparison of their retention times with those of the coated.
corresponding peaks in the chromatograms obtained with
reference solutions (a) and (b). Identification
The test is not valid unless (a) in the chromatogram obtained A. Determine by thin-layer chromatography (2.4.17), coating
with reference solution (c) the resolution between the peaks the plate with silica gel G.
due to quinine and quinidine is at least 1.5 and the resolution Mobile phase. A mixture of 80 volumes of toluene, 20 volumes
between the peaks due to dihydroquinidine and quinine is at of acetone and 10 volumes of diethylamine.
least 1.0 and (b) the signal-to-noise ratio of the principal peak
in the chromatogram obtained with reference solution (d) is at Test solution. Extract a quantity of the powdered tablets
least 5. containing 0.1 g of Quinine Bisulphate with 10 ml of a mixture
of 2 volumes of chloroform and 1 volume of ethanol (95 per
Inject the test solution and allow the chromatography to cent) and filter.
proceed for 2.5 times the retention time of the principal peak.
Calculate the percentage content of related substances by Reference solution. A 1.0 per cent w/v solution of quinine
normalisation, ignoring any peaks the areas of which are less sulphate in the same solvent mixture.
than that of the peak in the chromatogram obtained with Apply to the plate 2 µl of each solution. After development,
reference solution (d) (0.2 per cent). The content of dry the plate in air and spray with 0.05 M ethanolic sulphuric
dihydroquinine is not greater than 10 per cent, the content of acid and then with dilute potassium iodobismuthate solution.
any related substance eluting before quinine is not greater The principal spot in the chromatogram obtained with the test
than 5 per cent and the content of any other related substance solution corresponds to that in the chromatogram obtained
is not greater than 2.5 per cent. with the reference solution.

1020
IP 2007

B. Extract a quantity of the powdered tablets containing 0.1 g hexylamine in 700 ml of water, adjusting the pH to 2.8
of Quinine Bisulphate with 20 ml of water and filter (solution with 1 M orthophosphoric acid, adding 60 ml of
A). To 5 ml of solution A add 0.2 ml of bromine solution and acetonitrile and diluting to 1000 ml with water,
1 ml of dilute ammonia solution; an emerald-green colour is – flow rate. 1.5 ml per minute,
produced. – spectrophotometer set at 250 nm for reference solution
C. Solution A is laevo-rotatory. (e) and 316 nm for the other solutions,
– a 10 µl loop injector.
D. Solution A gives the reactions of sulphates (2.3.1).
Inject separately reference solutions (b) and (e). If necessary,
Tests adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b)
Dissolution (2.5.2). the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0
Apparatus. No 2 (the distance along the baseline between the point of injection
Medium. 900 ml of 0.1 M hydrochloric acid and the perpendicular dropped from the maximum of the peak
of an unretained component) being calculated from the peak
Speed and time. 100 rpm and 45 minutes.
due to thiourea in the chromatogram obtained with reference
Withdraw a suitable volume of the medium and filter. Measure solution (e).
the absorbance of the filtrate, suitably diluted if necessary, at
the maximum at about 348 nm (2.4.7). Calculate the content of Inject reference solutions (a), (b), (c) and (d). The
C20H24N2O2,H2SO4,7H2O in the medium taking 99 as the specific chromatogram obtained with reference solution (a) shows a
absorbance at 348 nm. principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 1.4. The
D. Not less than 70 per cent of the stated amount of chromatogram obtained with reference solution (b) shows a
C20H24N2O2,H2SO4,7H2O. principal peak due to quinidine and a peak due to
Other cinchona alkaloids. Determine by liquid dihydroquinidine, with a retention time relative to quinidine
chromatography (2.4.14). of about 1.2. The chromatogram obtained with reference
solution (c) shows four peaks due to quinine, dihydroquinine,
Test solution. Remove any coating from the tablets and mix a quinidine and dihydroquinidine which are identified by
quantity of the powdered tablet cores containing 50 mg of comparison of their retention times with those of the
Quinine Bisulphate with 20 ml of the mobile phase. Heat gently corresponding peaks in the chromatograms obtained with
to dissolve the powder as completely as possible, cool, dilute reference solutions (a) and (b).
to 25 ml with the mobile phase and filter, discarding the first
few ml of the filtrate. The test is not valid unless (a) in the chromatogram obtained
with reference solution (c) the resolution f between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate
due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile
between the peaks due to dihydroquinidine and quinine is at
phase and dilute to 10 ml with the mobile phase.
least 1.0 and (b) the signal-to-noise ratio of the principal peak
Reference solution (b). Prepare in the same manner as in the chromatogram obtained with reference solution (d) is at
reference solution (a) but using quinidine sulphate RS in least 5.
place of quinine sulphate RS.
Inject the test solution and allow the chromatography to
Reference solution (c). Mix equal volumes of reference proceed for 2.5 times the retention time of the principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
Reference solution (d). Dilute 1 volume of reference solution normalisation, ignoring any peaks the areas of which are less
(a) to 10 volumes with the mobile phase and dilute 1 volume of than that of the peak in the chromatogram obtained with
the resulting solution to 50 volumes with the mobile phase. reference solution (d) (0.2 per cent). The content of
dihydroquinine is not greater than 10 per cent, the content of
Reference solution (e). A solution containing 0.1 per cent any related substance eluting before quinine is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance
Chromatographic system is not greater than 2.5 per cent.
– a stainless steel column 25 cm x 4.6 mm packed with Other tests. Comply with the tests stated under Tablets.
octadecylsilane bonded to porous silica (5 µm) (such as
Hypersil ODS 5 µm), Assay. Weigh and powder 20 tablets. Weigh accurately a
– mobile phase: a solution prepared by dissolving 6.8 g of quantity of the powder containing about 0.6 g of Quinine
potassium dihydrogen orthophosphate and 3.0 g of Bisulphate, dissolve as completely as possible in 40 ml of

1021
IP 2007

acetic anhydride with the aid of heat and cool. Titrate with obtained with reference solution (b) shows two clearly
0.1 M perchloric acid, using crystal violet solution as separated spots.
indicator. Carry out a blank titration. B. To a 0.5 per cent w/v solution add an equal volume of dilute
1 ml of 0.1 M perchloric acid is equivalent to 0.05486 g of sulphuric acid; a strong blue fluorescence is produced.
C20H24N2O2,H2SO4,7H2O.
C. A 1 per cent w/v solution gives the reactions of chlorides
Storage. Store protected from light. (2.3.1).

Tests
pH (2.4.24). 2.0 to 3.0, determined in a 3.0 per cent w/v solution.
Quinine Dihydrochloride Specific optical rotation (2.4.22). –223º to –229º, determined
in a 3.0 per cent w/v solution in 0.1 M hydrochloric acid.
Quinine Acid Hydrochloride
Barium. To 15 ml of a 2 per cent w/v solution add 1 ml of
dilute sulphuric acid; the solution remains clear for at least
OCH3 15 minutes.
H
HO H H Dihydroquinine dihydrochloride. Not more than 10.0 per cent,
CH2 , 2HCl calculated on the dried basis and determined by the following
N
method. Dissolve 0.2 g in 20 ml of water, add 0.5 g of potassium
N H H bromide and 15 ml of 2 M hydrochloric acid. Titrate slowly
with 0.0167 M potassium bromate using methyl red solution
as indicator until a yellow colour is obtained. Add a solution
C20H24N2O2,2HCl Mol. Wt. 397.3 of 0.5 g of potassium iodide in 200 ml of water and stopper the
Quinine Dihydrochloride is the (8S,9R)-6′− flask immediately. Allow to stand in the dark for 5 minutes and
methoxycinchonan-9-ol dihydrochloride. The alkaloid is titrate with 0.1 M sodium thiosulphate using starch solution,
obtained from the bark of various species of Cinchona. added towards the end of the titration, as indicator. Repeat
the operation without the substance under examination.
Quinine Dihydrochloride contains not less than 99.0 per cent
and not more than 101.5 per cent of alkaloid dihydrochlorides, 1 ml of 0.0167 M potassium bromate is equivalent to
calculated as C20H24N2O2,2HCl on the dried basis. 0.01987 g of C20H24N2O2,2HCl. Calculate the content of
dihydroquinine dihydrochloride by subtracting the result from
Description. A white or almost white powder; odourless. the assay result.
Identification Other cinchona alkaloids. Determine by liquid
chromatography (2.4.14).
A. Determine by thin-layer chromatography (2.4.17), coating
Test solution. Dissolve 20 mg of the substance under
the plate with silica gel G.
examination in 5 ml of the mobile phase. Heat gently, if
Mobile phase. A mixture of 60 volumes of toluene, 36 volumes necessary to dissolve the powder as completely as possible,
of ether and 15 volumes of diethylamine. cool, dilute to 10 ml with the mobile phase and mix.
Test solution. Dissolve 1 g of the substance under examination Reference solution (a). Dissolve 20 mg of quinine sulphate
in 100 ml of methanol. RS, with gentle heating if necessary, in 5 ml of the mobile
phase and dilute to 10 ml with the mobile phase.
Reference solution (a). A 1 per cent w/v solution of quinidne
sulphate RS in methanol. Reference solution (b). Prepare in the same manner as
reference solution (a) but using quinidine sulphate RS in
Reference solution (b). A 1 per cent w/v solution of each of place of quinine sulphate RS.
quinidine sulphate RS and quinine sulphate RS in methanol.
Reference solution (c). Mix equal volumes of reference
Apply to the plate 4 µl of each solution. After development, solutions (a) and (b).
dry the plate in air for 15 minutes and repeat the development.
Dry the plate at 105º for 30 minutes, allow to cool and spray Reference solution (d). Dilute 1 volume of reference solution
with potassium iodoplatinate solution. The principal spot in (a) to 10 volumes with the mobile phase and dilute 1 volume of
the chromatogram obtained with the test solution corresponds the resulting solution to 50 volumes with the mobile phase.
to the spot in the chromatogram obtained with reference Reference solution (e). A solution containing 0.1 per cent
solution (a). The test is not valid unless the chromatogram w/v of thiourea in the mobile phase.

1022
IP 2007

Chromatographic system Titratable cation. 79.7 to 84.2 per cent, calculated on the dried
– a stainless steel column 25 cm x 4.6 mm packed with basis, determined by the following method. Weigh accurately
octadecylsilane bonded to porous silica (5 µm) (such as about 0.4 g, dissolve in 10 ml of water, add 40 ml of methanol
Hypersil ODS 5 µm), and titrate with 0.1 M sodium hydroxide using
– mobile phase: a solution prepared by dissolving 6.8 g of phenolphthalein solution as indicator.
potassium dihydrogen orthophosphate and 3.0 g of 1 ml of 0.1 M sodium hydroxide is equivalent to 0.01632 g of
hexylamine in 700 ml of water, adjusting the pH to 2.8 [C20H26N2O2]2+.
with 1 M orthophosphoric acid, adding 60 ml of
acetonitrile and diluting to 1000 ml with water Sulphates (2.3.17). 0.125 g complies with the limit test for
– flow rate. 1.5 ml per minute, sulphates ( 0.12 per cent).
– spectrophotometer set at 250 nm for reference solution Sulphated ash (2.3.18). Not more than 0.1 per cent.
(e) and 316 nm for the other solutions, Loss on drying (2.4.19). Not more than 3.0 per cent, determined
– a 10 µl loop injector. on 1.0 g by drying in an oven at 105º.
Inject separately reference solutions (b) and (e). If necessary, Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of
adjust the concentration of acetonitrile in the mobile phase so anhydrous glacial acetic acid and add 20 ml of acetic
that in the chromatogram obtained with reference solution (b) anhydride and 10 ml of mercuric acetate solution. Titrate
the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0 with 0.1 M perchloric acid, using crystal violet solution as
(the distance along the baseline between the point of injection indicator. Carry out a blank titration.
and the perpendicular dropped from the maximum of the peak
of an unretained component) being calculated from the peak 1 ml of 0.1 M perchloric acid is equivalent of 0.01987 g of
due to thiourea in the chromatogram obtained with reference C20H24N2O2,2HCl.
solution (e). Storage. Store protected from light.
Inject reference solutions (a), (b), (c) and (d). The
chromatogram obtained with reference solution (a) shows a
principal peak due to quinine and a peak due to dihydroquinine Quinine Dihydrochloride Injection
with a retention time relative to quinine of about 1.4. The
Quinine Acid Hydrochloride Injection
chromatogram obtained with reference solution (b) shows a
principal peak due to quinidine and a peak due to Quinine Dihydrochloride Injection is a sterile solution of
dihydroquinidine, with a retention time relative to quinidine Quinine Dihydrochloride in Water for Injections.
of about 1.2. The chromatogram obtained with reference Quinine Dihydrochloride Injection contains not less than
solution (c) shows four peaks due to quinine, dihydroquinine, 95.0 per cent and not more than 105.0 per cent of the stated
quinidine and dihydroquinidine which are identified by amount of quinine dihydrochloride, C20H24N2O2,2HCl.
comparison of their retention times with those of the
corresponding peaks in the chromatograms obtained with Description. A clear, almost colourless to light yellow solution.
reference solutions (a) and (b). Identification
The test is not valid unless (a) in the chromatogram obtained
A. Determine by thin-layer chromatography (2.4.17), coating
with reference solution (c) the resolution between the peaks
the plate with silica gel G.
due to quinine and quinidine is at least 1.5 and the resolution
between the peaks due to dihydroquinidine and quinine is at Mobile phase. A mixture of 80 volumes of toluene, 20 volumes
least 1.0 and (b) the signal-to-noise ratio of the principal peak of acetone and 10 volumes of diethylamine.
in the chromatogram obtained with reference solution (d) is at Test solution. Extract a volume of the injection containing
least 5. 0.1 g of Quinine Dihydrochloride Bisulphate with 10 ml of a
Inject the test solution and allow the chromatography to mixture of 2 volumes of chloroform and 1 volume of ethanol
proceed for 2.5 times the retention time of the principal peak. (95 per cent) and filter.
Calculate the percentage content of related substances by Reference solution (a). A 1 per cent w/v solution of quinine
normalisation, ignoring any peaks the areas of which are less sulphate in the same solvent mixture.
than that of the peak in the chromatogram obtained with
reference solution (d) (0.2 per cent). The content of Reference solution (b). A solution of 1 per cent w/v of each of
dihydroquinine is not greater than 10 per cent, the content of quinidine sulphate RS and quinine sulphate RS in the same
any related substance eluting before quinine is not greater solvent mixture.
than 5 per cent and the content of any other related substance Apply to the plate 2 µl of each solution. After development,
is not greater than 2.5 per cent. dry the plate in air and spray with 0.05 M ethanolic sulphuric

1023
IP 2007

acid and then with dilute potassium iodobismuthate solution. that in the chromatogram obtained with reference solution (b)
The principal spot in the chromatogram obtained with the test the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0
solution corresponds to that in the chromatogram obtained (the distance along the baseline between the point of injection
with the reference solution (a). The test is not valid unless the and the perpendicular dropped from the maximum of the peak
chromatogram obtained with reference solution (b) shows two of an unretained component) being calculated from the peak
clearly separated spots. due to thiourea in the chromatogram obtained with reference
B. Dilute 1 ml to 20 ml with water, add 0.1 M sodium hydroxide solution (e).
dropwise until the solution becomed turbid. Add 1 or 2 drops Inject reference solutions (a), (b), (c) and (d). The chromatogram
of 0.05 M sulphuric acid to obtain a clear solution. To this obtained with reference solution (a) shows a principal peak
solution add an equal volume of dilute sulphuric acid; a due to quinine and a peak due to dihydroquinine with a
strong blue fluorescence is produced. retention time relative to quinine of about 1.4. The
chromatogram obtained with reference solution (b) shows a
C. Solution A gives the reactions of chlorides (2.3.1).
principal peak due to quinidine and a peak due to
Tests dihydroquinidine, with a retention time relative to quinidine
of about 1.2. The chromatogram obtained with reference
pH (2.4.24).1.5 to 3.0. solution (c) shows four peaks due to quinine, dihydroquinine,
Other cinchona alkaloids. Determine by liquid quinidine and dihydroquinidine which are identified by
chromatography (2.4.14). comparison of their retention times with those of the
corresponding peaks in the chromatograms obtained with
Test solution. Dilute, if necessary a suitable volume of the reference solutions (a) and (b).
injection with the mobile phase to produce a solution
containing 0.2 per cent w/v of Quinine Dihydrochloride. The test is not valid unless (a) in the chromatogram obtained
with reference solution (c) the resolution between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile factor between the peaks due to dihydroquinidine and quinine
phase and dilute to 10 ml with the mobile phase. is at least 1.0 and (b) the signal-to-noise ratio of the principal
Reference solution (b). Prepare in the same manner as peak in the chromatogram obtained with reference solution
reference solution (a) but using quinidine sulphate RS in (d) is at least 5.
place of quinine sulphate RS. Inject the test solution and allow the chromatography to
Reference solution (c). Mix equal volumes of reference proceed for 2.5 times the retention time of the principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
normalisation, ignoring any peaks the areas of which are less
Reference solution (d). Dilute 1 volume of reference solution than that of the peak in the chromatogram obtained with
(a) to 10 volumes with the mobile phase and dilute 1 volume of reference solution (d) (0.2 per cent). The content of
the resulting solution to 50 volumes with the mobile phase. dihydroquinine is not greater than 10 per cent, the content of
Reference solution (e). A solution containing 0.1 per cent any related substance eluting before quinine is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance
is not greater than 2.5 per cent.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with Other tests. Complies with the tests stated under Parenteral
octadecylsilane bonded to porous silica (5 µm) (such as Preparations (Injections).
Hypersil ODS 5 µm), Assay. To an accurately measured volume containing about
– mobile phase: a solution prepared by dissolving 6.8 g of 0.5 g of Quinine Dihydrochloride add 20 ml of water and 5 ml
potassium dihydrogen orthophosphate and 3.0 g of of sodium hydroxide solution. Extract with successive
hexylamine in 700 ml of water, adjusting the pH to 2.8 quantities, each of 10 ml, of chloroform until complete extraction
with 1 M orthophosphoric acid, adding 60 ml of of the alkaloid is effected, washing each extract with the same
acetonitrile and diluting to 1000 ml with water, two quantities, each of 5 ml, of water. Remove the chloroform
– flow rate. 1.5 ml per minute, from the combined extracts, dissolve the residue in 50 ml of
– spectrophotometer set at 250 nm for reference solution anhydrous glacial acetic acid and add 20 ml of acetic
(e) and 316 nm for the other solutions, anhydride. Titrate with 0.1 M perchloric acid, using crystal
– a 10 µl loop injector. violet solution as indicator. Carry out a blank titration.
Inject separately reference solutions (b) and (e). If necessary, 1 ml of 0.1 M perchloric acid is equivalent to 0.01987 g of
adjust the concentration of acetonitrile in the mobile phase so C20H24N2O2,2HCl.

1024
IP 2007

Storage. Store protected from light. C. To a 0.5 per cent w/v solution add an equal volume of dilute
Labelling. The label states that the solution must be diluted sulphuric acid; a strong blue fluorescence is produced.
to a strength not exceeding 30 mg per ml before administration D. To 5 ml of a 1 per cent w/v solution add 1 ml of silver nitrate
and that care should be taken to ensure slow intravenous solution and stir with a glass rod; after a short interval, a
injection. white precipitate soluble in nitric acid is produced (distinction
from many other alkaloids).
E. A 1 per cent w/v solution gives the reactions of sulphates
(2.3.1).
Quinine Sulphate
Tests
Appearance of solution. A 2.0 per cent w/v solution in 0.1 M
OCH3 hydrochloric acid is clear (2.4.1), and not more intensely
H coloured than reference solution GYS4 (2.4.1).
HO H H
CH2 , H2SO4, 2H2O pH (2.4.24). 5.7 to 6.6, determined in a 1.0 per cent w/v
N suspension in water.
N H H
Specific optical rotation (2.4.22). –237º to –245º, determined
in a 2.0 per cent w/v solution in 0.1 M hydrochloric acid.
2
Other cinchona alkaloids. Determine by liquid
(C20H24N2O2)2,H2SO4,2H2O Mol. Wt.783.0 chromatography (2.4.14).
Test solution. Dissolve 20 mg of the substance under
Quinine Sulphate is (8S,9R)-6′-methoxycinchonan-9-ol
examination in 5 ml of the mobile phase. Heat gently, if
sulphate dihydrate. The alkaloid is obtained from the bark of
necessary to dissolve the powder as completely as possible,
various species of Cinchona.
cool, dilute to 10 ml with the mobile phase and mix.
Quinine Sulphate contains not less than 99.0 per cent and not Reference solution (a). Dissolve 20 mg of quinine sulphate
more than 101.0 per cent of alkaloid monosulphates, calculated RS, with gentle heating if necessary, in 5 ml of the mobile
as (C20H24N2O2)2,H2SO4 on the dried basis. phase and dilute to 10 ml with the mobile phase.
Description. White or almost white, needle-like crystals or a Reference solution (b). Prepare in the same manner as
crystalline powder. reference solution (a) but using quinidine sulphate RS in
place of quinine sulphate RS.
Identification
Reference solution (c). Mix equal volumes of reference
A. Determine by thin-layer chromatography (2.4.17), coating solutions (a) and (b).
the plate with silica gel G.
Reference solution (d). Dilute 1 volume of reference solution
Mobile phase. A mixture of 40 volumes of toluene, 24 volumes (a) to 10 volumes with the mobile phase and dilute 1 volume of
of ether and 10 volumes of diethylamine. the resulting solution to 50 volumes with the mobile phase.
Test solution. Dissolve 1 g of the substance under examination Reference solution (e). A solution containing 0.1 per cent
in 100 ml of methanol. w/v of thiourea in the mobile phase.
Reference solution. A 1 per cent w/v solution of quinine Chromatographic system
sulphate RS in methanol. – a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm) (such as
Apply to the plate 5 µl of each solution. After development, Hypersil ODS 5 µm),
dry the plate in air for 15 minutes and repeat the development. – mobile phase: a solution prepared by dissolving 6.8 g of
Dry the plate at 105º for 30 minutes, allow to cool and spray potassium dihydrogen orthophosphate and 3.0 g of
with potassium iodoplatinate solution. The principal spot in hexylamine in 700 ml of water, adjusting the pH to 2.8
the chromatogram obtained with the test solution corresponds with 1 M orthophosphoric acid, adding 60 ml of
to the spot in the chromatogram obtained with the reference acetonitrile and diluting to 1000 ml with water,
solution. – flow rate. 1.5 ml per minute,
B. To 5 ml of a 0.1 per cent w/v solution add 0.2 ml of bromine – spectrophotometer set at 250 nm for reference solution
solution and 1 ml of dilute ammonia solution; an emerald- (e) and 316 nm for the other solutions,
green colour is produced. – a 10 µl loop injector.

1025
IP 2007

Inject separately reference solutions (b) and (e). If necessary, Quinine Tablets
adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b) Quinine Sulphate Tablets
the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0 Quinine Sulphate Tablets contain not less than 95.0 per cent
(the distance along the baseline between the point of injection and not more than 105.0 per cent of the stated amount of
and the perpendicular dropped from the maximum of the peak quinine sulphate, (C20H24N2O2)2,H2SO4,2H2O. The tablets are
of an unretained component) being calculated from the peak coated.
due to thiourea in the chromatogram obtained with reference
solution (e). Identification
Inject reference solutions (a), (b), (c) and (d). The A. Determine by thin-layer chromatography (2.4.17), coating
chromatogram obtained with reference solution (a) shows a the plate with silica gel G.
principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 1.4. The Mobile phase. A mixture of 80 volumes of toluene, 20 volumes
chromatogram obtained with reference solution (b) shows a of acetone and 10 volumes of diethylamine.
principal peak due to quinidine and a peak due to Test solution. Extract a quantity of the powdered tablets
dihydroquinidine, with a retention time relative to quinidine containing 0.1 g of Quinine Sulphate with 10 ml of a mixture of
of about 1.2. The chromatogram obtained with reference 2 volumes of chloroform and 1 volume of ethanol (95 per
solution (c) shows four peaks due to quinine, dihydroquinine, cent) and filter.
quinidine and dihydroquinidine which are identified by
Reference solution. A 1 per cent w/v solution of quinine
comparison of their retention times with those of the
sulphate in the same solvent mixture.
corresponding peaks in the chromatograms obtained with
solutions (a) and (b). Apply to the plate 2 µl of each solution. After development,
dry the plate in air and spray with 0.05 M ethanolic sulphuric
The test is not valid unless (a) in the chromatogram obtained
acid and then with dilute potassium iodobismuthate solution.
with reference solution (c) the resolution between the peaks
The principal spot in the chromatogram obtained with the test
due to quinine and quinidine is at least 1.5 and the resolution
solution corresponds to that in the chromatogram obtained
between the peaks due to dihydroquinidine and quinine is at
with the reference solution.
least 1.0 and (b) the signal-to-noise ratio of the principal peak
in the chromatogram obtained with reference solution (d) is at B. Extract a quantity of the powdered tablets containing 0.1 g
least 5. of Quinine Sulphate with 20 ml of water and filter (solution A).
To 5 ml of solution A add 0.2 ml of bromine solution and 1 ml
Inject the test solution and allow the chromatography to of dilute ammonia solution; an emerald-green colour is
proceed for 2.5 times the retention time of the principal peak. produced.
Calculate the percentage content of related substances by
normalisation, ignoring any peaks the areas of which are less C. Solution A is laevo-rotatory.
than that of the peak in the chromatogram obtained with D. Solution A gives the reactions of sulphates (2.3.1).
reference solution (d) (0.2 per cent). The content of
dihydroquinine is not greater than 10 per cent, the content of Tests
any related substance eluting before quinine is not greater
than 5 per cent and the content of any other related substance Dissolution (2.5.2).
is not greater than 2.5 per cent. Apparatus. No 2
Sulphated ash (2.3.18). Not more than 0.1 per cent. Medium. 900 ml of 0.1 M hydrochloric acid
Speed and time. 100 rpm and 30 minutes.
Loss on drying (2.4.19). 3.0 to 5.0 per cent, determined on
1.0 g by drying in an oven at 105º. Withdraw a suitable volume of the medium and filter. Measure
the absorbance of the filtrate, suitably diluted if necessary, at
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of the maximum at about 348 nm (2.4.7). Calculate the content of
10 ml of chloroform and add 20 ml of acetic anhydride. Titrate C20H24N2O2, H2SO4, 2H2O in the medium from a solution of
with 0.1 M perchloric acid, determining the end-point known concentration of quinine sulphate RS.
potentiometrically (2.4.25). Carry out a blank titration.
D. Not less than 70 per cent of the stated amount of
1 ml of 0.1 M perchloric acid is equivalent to 0.02490 g of C20H24N2O2, H2SO4, 2H2O.
(C20H24N2O2)2,H2SO4.
Other cinchona alkaloids. Determine by liquid
Storage. Store protected from light. chromatography (2.4.14).

1026
IP 2007

Test solution. Remove any coating from the tablets and mix a quinidine and dihydroquinidine which are identified by
quantity of the powdered tablet cores containing 50 mg of comparison of their retention times with those of the
Quinine Sulphate with 20 ml of the mobile phase. Heat gently corresponding peaks in the chromatograms obtained with
to dissolve the powder as completely as possible, cool, dilute reference solutions (a) and (b).
to 25 ml with the mobile phase and filter, discarding the first The test is not valid unless (a) in the chromatogram obtained
few ml of the filtrate. with reference solution (c) the resolution between the peaks
Reference solution (a). Dissolve 20 mg of quinine sulphate due to quinine and quinidine is at least 1.5 and the resolution
RS, with gentle heating if necessary, in 5 ml of the mobile between the peaks due to dihydroquinidine and quinine is at
phase and dilute to 10 ml with the mobile phase. least 1.0 and (b) the signal-to-noise ratio of the principal peak
Reference solution (b). Prepare in the same manner as in the chromatogram obtained with reference solution (d) is at
reference solution (a) but using quinidine sulphate RS in least 5.
place of quinine sulphate RS. Inject the test solution and allow the chromatography to
Reference solution (c). Mix equal volumes of reference proceed for 2.5 times the retention time of the principal peak.
solutions (a) and (b). Calculate the percentage content of related substances by
normalisation, ignoring any peaks the areas of which are less
Reference solution (d). Dilute 1 volume of reference solution than that of the peak in the chromatogram obtained with
(a) to 10 volumes with the mobile phase and dilute 1 volume of reference solution (d) (0.2 per cent). The content of
the resulting solution to 50 volumes with the mobile phase. dihydroquinine is not greater than 10 per cent, the content of
Reference solution (e). A solution containing 0.1 per cent any related substance eluting before quinine is not greater
w/v of thiourea in the mobile phase. than 5 per cent and the content of any other related substance
Chromatographic system is not greater than 2.5 per cent.
– a stainless steel column 25 cm x 4.6 mm packed with Other tests. Comply with the tests stated under Tablets.
octadecylsilane bonded to porous silica (5 µm) (such as
Assay. Weigh and powder 20 tablets. Weigh accurately a
Hypersil ODS 5 µm),
quantity of the powder containing about 0.4 g of Quinine
– mobile phase: a solution prepared by dissolving 6.8 g of
Sulphate, dissolve as completely as possible in 40 ml of acetic
potassium dihydrogen orthophosphate and 3.0 g of
anhydride with the aid of heat and cool. Filter, if necessary
hexylamine in 700 ml of water, adjusting the pH to 2.8
through Whatman No.1 filter paper and rinse with an additional
with 1 M orthophosphoric acid, adding 60 ml of
40 ml of acetic anhydride in small volumes. Titrate with 0.1 M
acetonitrile and diluting to 1000 ml with water,
perchloric acid, using crystal violet solution as indicator.
– flow rate. 1.5 ml per minute,
Carry out a blank titration.
– spectrophotometer set at 250 nm for reference solution
(e) and 316 nm for the other solutions, 1 ml of 0.1 M perchloric acid is equivalent to 0.02610 g of
– a 10 µl loop injector. (C20H24N2O2)2,H2SO4,2H2O.
Inject separately reference solutions (b) and (e). If necessary, Storage. Store protected from light.
adjust the concentration of acetonitrile in the mobile phase so
that in the chromatogram obtained with reference solution (b)
the capacity factor of the peak due to quinidine is 3.5 to 4.5, V0
(the distance along the baseline between the point of injection Quiniodochlor
and the perpendicular dropped from the maximum of the peak Clioquinol; Iodochlorhydroxyquinoline;
of an unretained component) being calculated from the peak Iodochlorhydroxyquin
due to thiourea in the chromatogram obtained with reference
solution (e).
OH
Inject reference solutions (a), (b), (c) and (d). The
chromatogram obtained with reference solution (a) shows a I N
principal peak due to quinine and a peak due to dihydroquinine
with a retention time relative to quinine of about 1.4. The
chromatogram obtained with reference solution (b) shows a Cl
principal peak due to quinidine and a peak due to
dihydroquinidine, with a retention time relative to quinidine
C9H5CIINO Mol. Wt. 305.5
of about 1.2. The chromatogram obtained with reference
solution (c) shows four peaks due to quinine, dihydroquinine, Quiniodochlor is 5-chloro-7-iodoquinolin-8-ol.

1027
IP 2007

Quiniodochlor contains not less than 97.0 per cent and not Reference solution (a). Add 0.5 ml of N,O-bis
more than 103.0 per cent of C9H5ClINO, calculated on the dried (trimethylsilyl)acetamide to a mixture of 0.1 g of the substance
basis. under examination and 0.5 ml of pyridine, mix, allow to stand
Description. A yellowish white to brownish yellow powder; for 15 minutes and add 5 ml of hexane.
odour, faint and characteristic. Reference solution (b). Treat a mixture of 0.1 g of the substance
under examination and 0.5 ml of pyridine as described for the
Identification test solution.
Chromatographic system
A. Determine by infrared absorption spectrophotometry (2.4.6).
– a glass column 1.5 m x 4 mm, packed with silanised
Compare the spectrum with that obtained with quiniodochlor
diatomaceous support (100 to 120 mesh) coated with 3
RS or with the reference spectrum of quiniodochlor.
per cent w/w of methyl silicone gum,
B. When examined in the range 230 nm to 360 nm (2.4.7), a – temperature:
0.0005 per cent w/v solution in 3 M hydrochloric acid shows column.190º,
an absorption maximum at about 267 nm. inlet port and detector. 240º,
C. Burn 20 mg by the oxygen-flask method (2.3.34), using 5 ml – flame ionisation detector,
of 2 M sodium hydroxide as the absorbing liquid and dilute to – nitrogen as carrier gas.
25 ml with water. To 5 ml add 1 ml of silver nitrate solution; a In the chromatogram obtained with the test solution the peaks
yellow precipitate is produced. Add 5 ml of 5 M ammonia, following the solvent peak, in order of emergence, are due to
shake, filter and acidify the filtrate with nitric acid; a white (a) 5-chloro-8-hydroxyquinoline, (b) 5,7-dichloro-8-hydroxy-
precipitate is produced. quinoline, (c) the internal standard, (d) quiniodochlor and (e)
5,7-diiodo-8-hydroxyquinoline.
Tests In the chromatogram obtained with reference solution (b)
Acidity or alkalinity. Shake 0.5 g with 10 ml of water previously calculate the content of 5-chloro-8-hydroxy-quinoline, 5,7-
neutralised to phenolphthalein solution. The solution is dichloro-8-hydroxyquinoline and 5,7-diiodo-8-
colourless and not more than 0.05 ml of 0.1 M sodium hydroxyquinoline by reference to the corresponding peaks in
hydroxide is required to change the colour of the solution to the chromatogram obtained with the test solution.
pink. The total content of the named impurities does not exceed
3.0 per cent w/w, the content of any other impurity does not
Free iodine. Shake 1.0 g with a solution of 1 g of potassium
exceed 0.2 per cent w/w and the sum of the contents is not
iodide in 20 ml of water for 30 seconds, allow to stand for
more than 4.0 per cent w/w.
5 minutes and filter. To 10 ml of the filtrate add 1 ml of 1 M
sulphuric acid and 2 ml of chloroform and shake. Any colour Sulphated ash (2.3.18). Not more than 0.2 per cent.
in the chloroform layer is discharged on the addition of 0.1 ml Loss on drying (2.4.19). Not more than 0.5 per cent, determined
of 0.005 M sodium thiosulphate. on 1.0 g by drying over phosphorus pentoxide at a pressure
Halide ions. Shake 0.5 g with 25 ml of water for 1 minute and not exceeding 0.7 kPa for 24 hours.
filter. To the filtrate add 0.5 ml of 2 M nitric acid and 0.5 ml of Assay. Weigh accurately about 0.3 g and dissolve in 25 ml of
0.1 M silver nitrate and allow to stand for 5 minutes. Any anhydrous pyridine. Titrate with 0.1 M tetrabutylammonium
opalescence produced is not more intense than that obtained hydroxide, determining the end-point potentiometrically
by adding 0.5 ml of 0.1 M silver nitrate to 25 ml of water (2.4.25). Carry out a blank titration.
containing 0.5 ml of 2 M nitric acid and 0.2 ml of 0.01 M 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
hydrochloric acid and allowing to stand for 5 minutes. 0.03055 g of C9H5ClINO.
Related substances. Determine by gas chromatography Storage. Store protected from light.
(2.4.13).
Test solution. Add 0.5 ml of N,O-bis (trimethylsilyl)acetamide
to 0.5 ml of a solution in pyridine containing 0.4 per cent w/v Quiniodochlor Tablets
of each of 5-chloro-8- hydroxyquinoline, 5,7-dichloro-8-
hydroxy- quinoline and 5-chloro-7-iodo-8-hydroxyquinoline
Clioquinol Tablets; Iodochlorhydroxyquinoline Tablets;
and 0.04 per cent w/v of the substance under examination, Iodochlorhydroxyquin Tablets
mix, allow to stand for 15 minutes and add 5 ml of a 0.05 per Quiniodochlor Tablets contain not less than 90.0 per cent and
cent w/v solution of dibutylphthalate (internal standard) in not more than 110.0 per cent of the stated amount of
hexane. quiniodochlor, C9H5ClINO.

1028
IP 2007

Identification 50.0 ml with 2-methoxyethanol. To 5.0 ml of this solution add


1 ml of water and sufficient of a mixture of 24 volumes of
Triturate a quantity of the powdered tablets containing about 2-methoxyethanol and 6 volumes of water to produce 50.0 ml.
250 mg of Quiniodochlor with 20 ml of acetone, filter and add To 10.0 ml of the solution add 10 ml of 2-methoxyethanol and
20 ml of water to the filtrate. Collect the precipitate formed on 2 ml of a solution prepared by dissolving 0.5 g of ferric chloride
a filter and dry at 105º. The residue complies with the following hexahydrate in 80 ml of 2-methoxyethanol and adding 0.1 ml
tests. of hydrochloric acid and sufficient 2-methoxyethanol to
A. Determine by infrared absorption spectrophotometry (2.4.6). produce 100 ml. Dilute the solution to 25.0 ml with
Compare the spectrum with that obtained with quiniodochlor 2-methoxyethanol and measure the absorbance of the
RS or with the reference spectrum of quiniodochlor. resulting solution at the maximum at about 650 nm (2.4.7),
using as blank a solution prepared by treating 10 ml of the
B. When examined in the range 230 nm to 360 nm (2.4.7), a
aqueous 2-methoxyethanol in the same manner beginning at
0.0005 per cent w/v solution in 3 M hydrochloric acid shows
the words “add 10 ml of 2-methoxyethanol.....”.
an absorption maximum at about 267 nm.
Calculate the content of C9H5ClINO from the absorbance
Tests obtained using 10.0 ml of a solution prepared in the following
manner. Dissolve 0.125 g of quiniodochlor RS in sufficient 2-
Disintegration (2.5.1). Maximum time, 30 minutes.
methoxyethanol to produce 50.0 ml, warming to effect solution;
Other tests. Comply with the tests stated under Tablets. add 1 ml of water to 5.0 ml of the solution and add sufficient of
Assay. Weigh and finely powder 20 tablets. Weigh accurately the mixture of 24 volumes of 2-methoxyethanol and 6 volumes
a quantity of the powder containing 0.125 g of Quiniodochlor, of water to produce 50.0 ml. Using 10.0 ml of this solution
shake with 20 ml of hot 2-methoxyethanol, decant the hot repeat the operation beginning at the words “add 10 ml of
supernatant liquid through a fine filter. Repeat the extraction 2-methoxyethanol....”
with two further quantities, of 20 ml and 10 ml, of Storage. Store protected from light.
2-methoxyethanol, combine the filtered extracts and dilute to

1029
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

R
Rabeprazole Sodium ....
Rabeprazole Tablets ....
Ramipril ....
Ramipril Capsules ....
Ramipril Tablets ....
Ranitidine Hydrochloride ....
Ranitidine Injection ....
Ranitidine Tablets ....
Purifid Rayon ....
Reserpine ....
Reserpine Injection ....
Reserpine Tablets ....
Riboflavine ....
Riboflavine Sodium Phosphate ....
Riboflavine Tablets ....
Rifampicin ....
Rifampicin Capsules ....
Rifampicin Oral Suspension ....
Rifampicin Tablets ....
Rifampicin and Isoniazid Tablets ....
Rifampicin, Isoniazid And Ethambutol Tablets ....
Rifampicin, Isoniazid and Pyrazinamide Tablets ....
Rifampicin, Isoniazid, Pyrazinamide And Ethambutol Tablets ....
Ritonavir ....
Ritonavir Capsules ....
Ritonavir Tablets ....
Rosiglitazone Maleate ....
Rosiglitazone Tablets ....
Rosuvastatin Calcium ....
Rosuvastatin Tablets ....

1031
MONOGRAPHS INDIAN PHARMACOPOEIA 2007

Roxithromycin ....
Roxithromycin Tablets ....

1032
IP 2007 RABEPRAZOLE SODIUM

Rabeprazole Sodium Water (2.3.43). Not more than 7.0 per cent, determined on
0.3 g.
Na O N Assay. Determine by liquid chromatography (2.4.14).
N S Test solution. Dissolve 0.1 g of the substance under
O OCH3 examination in 100.0 ml of mobile phase. Dilute 5.0 ml of the
N CH3 solution to 100.0 ml with the same solvent.
C18H20N3O3S.Na Mol. Wt. 381.4 Reference solution. A 0.005 per cent w/v solution of
rabeprazole sodium RS in the mobile phase.
Rabeprazole sodium is 2-({[4-(3-methoxypropoxy)-3-methyl-
2-pyridinyl]methyl}sulphenyl)-1H-benzimidazole sodium. Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with
Rabeprazole sodium contains not less than 98.0 per cent and octylsilane bonded to porous silica (5 µm) (such as
not more than 102.0 per cent of C18H20N3O3S.Na, calculated on Hypersil keystone betabasic C8),
the anhydrous basis. – column temperature 40°,
Description. A white to light yellow, crystalline powder, – mobile phase: a mixture of 72 volumes of 0.1 M
hygroscopic. phosphate buffer pH 7.0 and 28 volumes of acetonitrile,
– flow rate. 1.4 ml per minute,
Identification – spectrophotometer set at 282 nm,
A. Determine by infrared absorption spectrophotometry (2.4.6). – a 10 µl loop injector.
Compare the spectrum with that obtained with rabeprazole Inject the reference solution. The test is not valid unless the
sodium RS. tailing factor is not more than 2.0 and the relative standard
B. A 10 per cent w/v solution in carbon dioxide-free water deviation for replicate injections is not more than 2.0 per cent.
gives reaction of sodium (2.3.1). Inject the test solution and the reference solution.

Tests Calculate the percentage content of C18H20N3O3S.Na.


Storage. Store protected from light and moisture.
Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Dissolve 50 mg of the substance under
examination in 100 ml with the mobile phase. Rabeprazole Tablets
Reference solution (a). A 0.05 per cent w/v solution of Rabeprazole Sodium Tablets
rabeprazole sodium RS in the mobile phase.
Rabeprazole Tablets contain Rabeprazole Sodium.
Reference solution (b). Dilute 1 ml of reference solution (a) to
Rabeprazole Tablets contain not less than 90.0 per cent and
100 ml with mobile phase.
not more than 110.0 per cent of the stated amount of
Chromatographic system as described under Assay. rabeprazole sodium, C18H20N3O3SNa.
Inject reference solution (a). The test is not valid unless the
Identification
tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates. In the Assay, the principal peak in the chromatogram obtained
Inject the test solution and reference solution (b). Run the with the test solution corresponds to the peak in the
chromatogram three times of the principal peak. In the chromatogram obtained with the reference solution.
chromatogram obtained with the test solution, the area of any
Tests
secondary peak is not more than 0.5 times the area of the peak
in the chromatogram obtained with reference solution (b) (0.5 Dissolution (2.5.2).
per cent) and the sum of areas of all the secondary peaks is
Apparatus. No 1
not more than 1.5 times the area of the peak in the chromatogram
obtained with the reference solution (b) (1.5 per cent). Medium. 900 ml of 0.1 M hydrochloric acid.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Speed and time. 50 rpm for 120 minutes.
heavy metals, Method A (20 ppm). Replace the 0.1 M hydrochloric acid with phosphate buffer
Sulphated ash (2.3.18). Not more than 0.1 per cent. pH 7.4. Run the apparatus at 75 rpm for 45 minutes. Withdraw

1033
RAMIPRIL IP 2007

a suitable volume of the medium and filter. Measure the Rabeprazole Sodium, disperse in 20 ml of 0.1 M sodium
absorbance of the filtered solution immediately, suitably diluted hydroxide and dilute to 100.0 ml with solvent mixture, filter.
with phosphate buffer pH 10.4 if necessary, at the maximum at Reference solution. Weigh accurately about 25 mg of
about 291 nm (2.4.7). Calculate the content of C18H20N3O3SNa rabeprazole sodium RS, dissolve in 10 ml of 0.1 M sodium
in the medium from the absorbance obtained from a solution hydroxide and dilute to 50.0 ml with solvent mixture.
of known concentration of rabeprazole sodium RS, prepared
by dissolving in minimum quantity of a mixture of 75 volumes Chromatographic system
of acetonitrile and 25 volumes of methanol and suitably – a stainless steel column 25 cm x 4.6 mm packed with
diluted with phosphate buffer pH 10.4. octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 65 volumes of 0.15 per cent
D. Not less than 70 per cent of the stated amount of
w/v solution of potassium dihydrogen phosphate
C18H20N3O3SNa.
previously adjusted pH to 6.0 with orthophosphoric
Related substances. Determine by liquid chromatography acid and 35 volumes of acetonitrile,
(2.4.14). – flow rate. 1 ml per minute,
Solvent mixture. 80 volumes of methanol, 20 volumes of water – spectrophotometer set at 280 nm,
and 0.1 volume of diethylamine. – a 10 µl loop injector.
Test solution. Weigh accurately a quantity of the powdered Inject the reference solution. The test is not valid unless the
tablet containing 50 mg of Rabeprazole Sodium, disperse in tailing factor is not more than 2.0 and the relative standard
100 ml of solvent mixture and filter. deviation for replicate injections is not more than 2.0 per cent.
Reference solution (a). A 0.05 per cent w/v solution of Inject the test solution and the reference solution.
rabeprazole sodium RS in the solvent mixture.
Calculate the content of C18H20N3O3SNa.
Reference solution (b). Dilute 1 ml of reference solution (a) to
Storage. Store protected from light and moisture.
100 ml with solvent mixture.
Chromatographic system as described under Assay.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
Ramipril
not less than 2000 theoretical plates. C23H32N2O5 Mol. Wt. 416.5
Inject the test solution and reference solution (b). Run the Ramipril is (2S,3aS,6aS)-1-[(S)-2-[[(S)-1-(ethoxycarbonyl)-3-
chromatogram three times of the principal peak. In the phenylpropyl]amino]propanoyl]
chromatogram obtained with the test solution, the area of any octahydrocyclopenta[b]pyrrole-2-carboxylic acid.
secondary peak is not more than twice the area of the peak in
Ramipril contains not less than 98.0 per cent and not more
the chromatogram obtained with reference solution (b) (2.0
than 101.0 per cent of C23H32N2O5, calculated on the dried basis.
per cent) and the sum of areas of all the secondary peaks is
not more than 6 times the area of the peak in the chromatogram Description. A white or almost white, crystalline powder.
obtained with the reference solution (b) (6.0 per cent).
Identification
Uniformity of content (For tablets containing 10 mg or less).
Comply with the tests stated under Tablets. Determine by infrared absorption spectrophotometry (2.4.6).
Disperse 1 tablet in sufficient 0.1 M sodium hydroxide to Compare the spectrum with that obtained with ramipril RS.
produce 0.0015 per cent w/v solution. Measure the absorbance
Tests
of the resulting solution at the maximum at about 292 nm (2.4.7).
Calculate the content of C18H20N3O3SNa from the absorbance Appearance of solution. A 1.0 per cent w/v solution in methanol
obtained from same concentration of rabeprazole sodium RS is clear (2.4.1) and colourless (2.4.1).
in the same medium.
Specific optical rotation (2.4.22). + 32.0° to + 38.0°, determined
Other tests. Comply with the tests stated under Tablets. in 1.0 per cent w/v solution in 0.1 M methanolic hydrochloric
Assay. Determine by liquid chromatography (2.4.14). acid.
Solvent mixture. 80 volumes of methanol, 20 volumes of water Related substances. Determine by liquid chromatography
and 0.1 volume of diethylamine. (2.4.14).
Test solution. Weigh and powder 20 tablets. Weigh accurately Test solution. Dissolve 25 mg of the substance under
a quantity of the powdered tablet containing 50 mg of examination in 25 ml of mobile phase B.

1034
IP 2007 RAMIPRIL CAPSULES

Reference solution (a). A 0.1 per cent w/v solution of ramipril 1 ml of 0.1 M sodium hydroxide is equivalent to 0.04165 gm of
RS in the mobile phase B. C23H32N2O5.
Reference solution (b). Dilute 1 ml of reference solution (a) to Storage. Store protected from light.
100 ml with mobile phase B.
Chromatographic system
– a stainless steel column 25 cm x 4.0 mm packed with
octadecylsilyl bonded to porous silica (5 µm),
Ramipril Capsules
– column temperature 65°, Ramipril Capsules contain not less than 90.0 per cent and not
– mobile phase: A. dissolve 2.0 g of sodium perchlorate more than 110.0 per cent of the stated amount of ramipril,
in a mixture of 0.5 ml of triethylamine and 800 ml of C23H32N2O5.
water; adjust pH to 3.6 with orthophosphoric acid and
add 200 ml of acetonitrile, Identification
B. dissolve 2.0 g of sodium perchlorate in a mixture
Shake a quantity of the content of the capsules containing 25
of 0.5 ml of triethylamine and 300 ml of water; adjust
mg of Ramipril with 50 ml of acetone, centrifuge for
pH to 2.6 with orthophosphoric acid and add 700 ml of
10 minutes, filter. Evaporate the filtrate to dryness at 60° for 3
acetonitrile,
hours. The residue complies with the following test.
– a linear gradient programme using the conditions given
below, Determine by infrared absorption spectrophotometry (2.4.6).
– flow rate. 1 ml per minute, Compare the spectrum with that obtained with ramipril RS.
– spectrophotometer set at 210 nm,
– a 10 µl loop injector. Tests
Time Mobile phase A Mobile phase B Dissolution (2.5.2).
(in min.) (per cent v/v) (per cent v/v)
Apparatus. No 1
0 90 10 Medium. 500 ml of 0.1 M hydrochloric acid.
6 90 10 Speed and time. 75 rpm and 45 minutes.
7 75 25
Withdraw a suitable volume of the medium and filter.
20 65 35
Determine by liquid chromatography (2.4.14).
30 25 75
40 25 75 Test solution. Dilute the filtrate to get 0.00025 per cent w/v
45 90 10 solution of Ramipril with 0.1 M hydrochloric acid.
55 90 10 Reference solution. A 0.00025 per cent w/v solution of ramipril
RS in 0.1 M hydrochloric acid.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in Chromatographic system as described under Assay.
not less than 2000 theoretical plates. Inject the test solution and the reference solution.
Inject the test solution and reference solution (b). In the Calculate the content of C23H32N2O5.
chromatogram obtained with the test solution, the area of any
secondary peak is not more than 0.5 times the area of the peak D. Not less than 70 per cent of the stated amount of
in the chromatogram obtained with reference solution (b) (0.5 C23H32N2O5.
per cent) and the sum of areas of all the secondary peaks is Uniformity of content (For capsules containing 10 mg or
not more than the area of the peak in the chromatogram less). Comply with the test stated under Capsules.
obtained with the reference solution (b) (1.0 per cent).
Determine by liquid chromatography (2.4.14).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Test solution. Disperse one capsule in 100 ml of 0.1 M
Loss on drying (2.4.19). Not more than 0.2 per cent, determined hydrochloric acid, sonicate for 15 minutes. Dilute if necessary
on 1.0 g by drying in an oven at 60°, under vaccume, for to produce 0.025 per cent w/v solution of Ramipril in 0.1 M
4 hours. hydrochloric acid.
Assay. Weigh accurately about 0.3 gm, dissolve in 25 ml of
Reference solution. A 0.025 per cent w/v solution of
methanol and add 25 ml of water. Titrate with 0.1 M sodium
ramipril RS in 0.1 M hydrochloric acid.
hydroxide. Determine the end-point potentiometrically (2.4.25).
Carry out a blank titration. Chromatographic system as described under Assay.

1035
RAMIPRIL TABLETS IP 2007

Inject the test solution and the reference solution. Determine by liquid chromatography (2.4.14).
Calculate the content of C23H32N2O5. Test solution. Dilute the filtrate to get 0.00025 per cent w/v
Assay. Determine by liquid chromatography (2.4.14). solution of Ramipril with 0.1 M hydrochloric acid.

Test solution. Weigh a quantity of the content of capsules Reference solution. A 0.00025 per cent w/v solution of ramipril
containing 25 mg of Ramipril, disperse in 100.0 ml of 0.1 M RS in 0.1 M hydrochloric acid.
hydrochloric acid, mix and centrifuge. Chromatographic system as described under Assay.
Reference solution. A 0.025 per cent w/v solution of ramipril Inject the test solution and the reference solution.
RS in 0.1 M hydrochloric acid.
Calculate the content of C23H32N2O5.
Chromatographic system
– a stainless steel column 12.5 cm x 4.6 mm packed with D. Not less than 70 per cent of the stated amount of
octadecylsilyl bonded to porous silica (5 µm), C23H32N2O5.
– mobile phase: a mixture of 42 volumes of acetonitrile
Uniformity of content (For tablets containing 10 mg or less).
and 58 volumes of a solution containing 1.4 per cent w/
Comply with the test stated under Tablets.
v solution of sodium perchlorate and 0.58 per cent w/
v solution of orthophosphoric acid adjusted to pH 2.5 Determine by liquid chromatography (2.4.14), as described
with triethylamine, adjust the pH of the mixture to 2.1 under Assay.
with orthophosphoric acid,
Test solution. Take one tablet, add 5 ml of 0.1 M hydrochloric
– flow rate. 1 ml per minute,
acid, sonicate for 10 minutes, dilute, if necessary, with
– spectrophotometer set at 210 nm,
sufficient 0.1 M hydrochloric acid to produce a solution
– a 50 µl loop injector.
containing 0.025 per cent w/v of Ramipril, centrifuge and use
Inject the reference solution. The test is not valid unless the the supernatant liquid.
relative standard deviation for replicate injections is not more
Calculate the content of C23H32N2O5.
than 2.0 per cent.
Inject the test solution and the reference solution. Assay. Determine by liquid chromatography (2.4.14).

Calculate the content of C23H32N2O5. Test solution. Weigh and powder 20 tablets. Weigh a quantity
of powdered tablets containing 25 mg of Ramipril, disperse in
100.0 ml of 0.1 M hydrochloric acid and centrifuge.
Reference solution. A 0.025 per cent w/v solution of ramipril
Ramipril Tablets RS in 0.1 M hydrochloric acid.
Ramipril Tablets contain not less than 90.0 per cent and not
Chromatographic system
more than 110.0 per cent of the stated amount of ramipril,
– a stainless steel column 12.5 cm x 4.6 mm packed with
C23H32N2O5.
octadecylsilyl bonded to porous silica (5 µm),
– mobile phase: a mixture of 42 volumes of acetonitrile
Identification
and 58 volumes of a solution containing 1.4 per cent
Shake a quantity of the powdered tablets containing 25 mg of w/v solution of sodium perchlorate and 0.58 per cent
Ramipril with 50 ml of acetone, centrifuge for 10 minutes, filter. w/v solution of orthophosphoric acid adjusted to pH
Evaporate the filtrate to dryness at 60° for 3 hours. The residue 2.5 with triethylamine, adjust the pH of the mixture to
complies with the following test. 2.1 with orthophosphoric acid,
– flow rate. 1 ml per minute,
Determine by infrared absorption spectrophotometry (2.4.6).
– spectrophotometer set at 210 nm,
Compare the spectrum with that obtained with ramipril RS.
– a 50 µl loop injector.
Tests Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
Dissolution (2.5.2).
than 2.0 per cent.
Apparatus. No 1
Inject the test solution and the reference solution.
Medium. 500 ml of 0.1 M hydrochloric acid.
Speed and time. 75 rpm and 45 minutes. Calculate the content of C23H32N2O5.
Withdraw a suitable volume of the medium and filter. Storage. Store protected from moisture.

1036
IP 2007 RANITIDINE HYDROCHLORIDE

Ranitidine Hydrochloride Reference solution (c). Dilute 30.0 ml of reference solution (a)
to 100 ml with methanol.
H Reference solution (d). Dilute 5.0 ml of reference solution (a)
O N CHNO2 to 100 ml with methanol.
H3C N S , HCl
NHCH3 Reference solution (e). Weigh accurately a quantity of
CH3
ranitidine hydrochloride related compound A RS in
C13H22N4O3S,HCl Mol. Wt. 350.9 methanol, and dilute with methanol to obtain a solution
containing a known concentration of about 0.127 per cent
Ranitidine Hydrochloride is N-[2-[[[5-[(dimethylamino)
w/v.
methyl]furan-2-yl]methyl]thio]ethyl]-N-methyl-2-
nitroethene-1,1-diamine hydrochloride. Reference solution (f). Weigh accurately a quantity of
ranitidine hydrochloride related compound B RS in
Ranitidine Hydrochloride contains not less than 97.5 per cent
methanol, and dilute with methanol to obtain a solution
and not more than 102.0 per cent of C13H22N4O3S, HCl,
containing a known concentration of about 0.1 per cent w/v.
calculated on the dried basis.
Apply to the plate 10 µl of each solution except reference
Description. A white to pale yellow, crystalline powder.
solution (e). Apply separately an additional 10 µl of the test
Identification solution and on top of this application, apply 10 µl of reference
solution (e). After development, dry the plate in air and expose
A. Determine by infrared absorption spectrophotometry (2.4.6). it to iodine vapours in a closed chamber until the spots are
Compare the spectrum with that obtained with ranitidine revealed. Any spot in the chromatogram obtained with the
hydrochloride RS or with the reference spectrum of ranitidine test solution corresponding to the principal spot in the
hydrochloride. chromatogram obtained with reference solution (f) is not more
B. In the test for Related substances, the principal spot in the intense than that of the principal spot in the chromatogram
chromatogram obtained with test solution (b) corresponds to obtained with reference solution (b) (0.5 per cent) and no
that in the chromatogram obtained with reference solution (a). other spot in the chromatogram obtained with the test solution
is more intense than the principal spot in the chromatogram
C. A 5 per cent w/v solution gives the reactions of chlorides obtained with reference solution (c) (0.3 per cent). The sum of
(2.3.1). the intensities of all the secondary spots in the chromatogram
Tests obtained with the test solution does not exceed 1.0 per cent.
The test is not valid unless the chromatogram obtained with
Appearance of solution. A 1.0 per cent w/v solution is clear
the combined test solution and reference solution (e) shows
(2.4.1), and not more intensely coloured than reference solution
two clearly separated principal spots and the chromatogram
BYS5 (2.4.1).
obtained with reference solution (d) shows a clearly visible
pH (2.4.24). 4.5 to 6.0, determined in a 1.0 per cent w/v solution spot.
in carbon dioxide-free water.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Loss on drying (2.4.19). Not more than 0.75 per cent,
determined on 1.0 g by drying in an oven at 60º at a pressure
Mobile phase. A mixture of 25 volumes of ethyl acetate,
not exceeding 2.75 kPa for 3 hours.
15 volumes of 2-propanol, 4 volumes of strong ammonia
solution and 2 volumes of water. Assay. Determine by liquid chromatography (2.4.14)
Test solution (a). Dissolve 0.22 g of the substance under Test solution. A 0.0112 per cent w/v of the substance under
examination in 10 ml of methanol. examination in the mobile phase.
Test solution (b). Dilute 1 ml of test solution (a) to 100 ml with Reference solution. 0.0112 per cent w/v of ranitidine
methanol. hydrochloride RS in the mobile phase.
Reference solution (a). Weigh accurately a quantity of Chromatographic system
ranitidine hydrochloride RS in methanol, and dilute with – a stainless steel column 25 cm x 4.0 mm, packed with
methanol to obtain a solution containing a known octadecylsilane bonded to porous silica (5 µm),
concentration of about 0.022 per cent w/v. – mobile phase: a mixture of 85 volumes of methanol and
Reference solution (b). Dilute 10.0 ml of reference solution (a) 15 volumes of 0.1 M ammonium acetate,
to 20 ml with methanol. – flow rate. 2 ml per minute,

1037
RANITIDINE INJECTION IP 2007

–– spectrophotometer set at 322 nm, Reference solution (a). Weigh accurately a quantity of
– a 20 µl loop injector. ranitidine hydrochloride RS in water, and dilute with water
Inject the reference solution. The test is not valid unless the to obtain a solution containing a known concentration of
relative standard deviation for replicate injections is not more about 0.056 per cent w/v.
than 2.0 per cent. Reference solution (b). Dilute 10.0 ml of reference solution (a)
to 20 ml with water.
Inject alternately the test solution and the reference solution.
Reference solution (c). Dilute 5.0 ml of reference solution (a)
Calculate the content of C13H22N4O3S, HCl.
to 20 ml with water.
Storage. Store protected from light and moisture.
Reference solution (d). Dilute 6.0 ml of reference solution (c)
to 10 ml with water.
Reference solution (e). Dilute 5.0 ml of reference solution (b)
Ranitidine Injection to 50 ml with water.
Ranitidine Hydrochloride Injection Reference solution (f). Dilute 5 ml of reference solution (e) to
10 ml with water.
Ranitidine Injection is a sterile solution of Ranitidine
Hydrochloride in Water for Injections and may be suitably Reference solution (g). A 0.127 per cent w/v of solution of
buffered. ranitidine impurity A RS in methanol.
Ranitidine Hydrochloride Injection contains not less than Apply to the plate 10 µl of each solution. Apply separately an
90.0 per cent and not more than 110.0 per cent of the stated additional 10 µl of the test solution and on top of this
amount of ranitidine, C13H22N4O3S. application, apply 10 µl of reference solution (g). After
development, dry the plate in air and expose it to iodine vapours
Identification in a closed chamber until the spots are revealed. The major
secondary spot in the chromatogram obtained with the test
A. To a volume of the injection containing 25 mg of ranitidine solution is not more intense than that of the principal spot in
add 20 ml of methanol, mix and evaporate to dryness. Add the chromatogram obtained with reference solution (a)
1 ml of light petroleum (60º to 80º) to the resulting residue, (2.0 per cent) and no other secondary spot in the chromatogram
scratch the side of the vessel with a glass rod to induce obtained with the test solution is more intense than the
crystallisation, evaporate to dryness and dry the residue at principal spot in the chromatogram obtained with reference
60º for 10 minutes.The residue complies with the following solution (b) (1.0 per cent). The sum of the intensities of all the
test. secondary spots in the chromatogram obtained with the test
Determine by infrared absorption spectrophotometry (2.4.6). solution does not exceed 5.0 per cent.
Compare the spectrum with that obtained with ranitidine The test is not valid unless the chromatogram obtained with
hydrochloride RS or with the reference spectrum of ranitidine the combined test solution and reference solution (g) shows
hydrochloride. two clearly separated principal spots and the chromatogram
B. In the Assay, the principal peak in the chromatogram obtained with reference solution (f) shows a clearly visible
obtained with the test solution corresponds to the peak in the spot.
chromatogram obtained with the reference solution. Other tests. Comply with the tests stated under Parenteral
Preparations (Injections).
Tests
Assay. Determine by liquid chromatography (2.4.14)
pH (2.4.24). 6.7 to 7.3, if the preparation is buffered; 4.5 to 7.0,
Test solution. Dilute a volume of the injection containing
if the preparation is unbuffered.
10.0 mg of ranitidine to 100.0 ml with the mobile phase.
Related substances. Determine by thin-layer chromatography
Reference solution. A 0.0112 per cent w/v solution of
(2.4.17), coating the plate with silica gel GF254.
ranitidine hydrochloride RS in the mobile phase.
Mobile phase. A mixture of 25 volumes of ethyl acetate,
Chromatographic system
15 volumes of 2-propanol, 4 volumes of strong ammonia
– a stainless steel column 25 cm x 4.0 mm packed with
solution and 2 volumes of water.
octadecylsilane bonded to porous silica (5 µm),
Test solution. Dilute suitably a volume of the injection with – mobile phase: a mixture of 85 volumes of methanol and
water to produce a solution containing the equivalent of 15 volumes of 0.1 M ammonium acetate,
2.5 per cent w/v of ranitidine in methanol. – flow rate. 2 ml per minute,

1038
IP 2007 RANITIDINE TABLETS

– spectrophotometer set at 322 nm, methanol to obtain a solution containing a known


– a 20 µl loop injector. concentration of about 0.022 per cent w/v.
Inject the reference solution. The test is not valid unless the Reference solution (b). Dilute 10 ml of reference solution (a)
relative standard deviation for replicate injections is not more to 20 ml with methanol.
than 2.0 per cent. Reference solution (c). Dilute 30 ml of reference solution (a)
Inject alternately the test solution and the reference solution. to 100 ml with methanol.
Calculate the content of C13H22N4O3S in the injection. Reference solution (d). Dilute 5 ml of reference solution (a) to
50 ml with methanol.
Storage. Store protected from light.
Reference solution (e). Dilute 5 ml of reference solution (a) to
Labelling. The label states (1) the strength in terms of the 100 ml with methanol.
equivalent amount of ranitidine; (2) where appropriate, that
Reference solution (f). Weigh accurately a quantity of
the injection is buffered.
ranitidine hydrochloride related compound A RS in
methanol, and dilute with methanol to obtain a solution
containing a known concentration of about 0.127 per cent
Ranitidine Tablets w/v.

Ranitidine Hydrochloride Tablets Apply to the plate 10 µl of each solution. Apply separately an
additional 10 µl of the test solution and on top of this
Ranitidine Tablets contain not less than 90.0 per cent and not application, apply 10 µl of reference solution (f). After
more than 110.0 per cent of the stated amount of the ranitidine, development, dry the plate in air and expose it to iodine vapours
C13H22N4O3S. The tablets are coated. in a closed chamber until the spots are revealed. Any spot in
the chromatogram obtained with the test solution
Identification corresponding to the principal spot in the chromatogram
obtained with reference solution (f) is not more intense than
A. Shake a quantity of the powdered tablets containing 25 mg
that of the principal spot in the chromatogram obtained with
of ranitidine with 5 ml of methanol for 5 minutes, filter and
reference solution (b) (0.5 per cent) and no other spot in the
evaporate the filtrate to dryness. Add 1 ml of light petroleum
chromatogram obtained with the test solution is more intense
(60º to 80º) to the resulting residue, scratch the side of the
than the principal spot in the chromatogram obtained with
vessel with a glass rod to induce crystallisation, evaporate to
reference solution (c) (0.3 per cent). The sum of the intensities
dryness and dry the residue at 60º for 10 minutes. The residue
of all the secondary spots in the chromatogram obtained with
complies with the following test.
the test solution does not exceed 2.0 per cent.
Determine by infrared absorption spectrophotometry (2.4.6).
The test is not valid unless the chromatogram obtained with
Compare the spectrum with that obtained with ranitidine
the combined test solution and reference solution (f) shows
hydrochloride RS or with the reference spectrum of ranitidine
two clearly separated principal spots and the chromatogram
hydrochloride.
obtained with reference solution (e) shows a clearly visible
B. In the Assay, the principal peak in the chromatogram spot.
obtained with the test solution corresponds to the peak in the Other tests. Comply with the tests stated under Tablets.
chromatogram obtained with the reference solution.
Assay. Determine by liquid chromatography (2.4.14)
Tests Test solution. Weigh and powder 20 tablets. Shake 1.5 g of the
Related substances. Determine by thin-layer chromatography powder with 400 ml of the mobile phase, dilute to 500.0 ml with
(2.4.17), coating the plate with silica gel GF254. the mobile phase, filter and dilute the filtrate with the mobile
phase to obtain a solution containing the equivalent of
Mobile phase. A mixture of 25 volumes of ethyl acetate, 0.01 per cent w/v of ranitidine.
15 volumes of 2-propanol, 4 volumes of strong ammonia
Reference solution. A 0.0112 per cent w/v solution of
solution and 2 volumes of water.
ranitidine hydrochloride RS in the mobile phase.
Test solution. Shake a quantity of the powdered tablets
Chromatographic system
containing 0.45 g of ranitidine with 20 ml of methanol and
– a stainless steel column 25 cm x 4.0 mm, packed with
filter.
octadecylsilane bonded to porous silica (5 µm),
Reference solution (a). Weigh accurately a quantity of – mobile phase: a mixture of 85 volumes of methanol and
ranitidine hydrochloride RS in methanol, and dilute with 15 volumes of 0.1 M ammonium acetate,

1039
PURIFIED RAYON IP 2007

– flow rate. 2 ml per minute, allow to macerate for 2 hours. Decant the solution, squeeze
– spectrophotometer set at 322 nm, the residual liquid carefully from the sample with a glass rod
– a 20 µl loop injector. mix and filter. The filtered extract is colourless. Compare the
Inject the reference solution. The test is not valid unless the colour of the extract with water using identical tubes of
relative standard deviation for replicate injections is not more colourless, transparent, neutral glass 12 mm in diameter
than 2.0 per cent. measuring 2 ml. Compare the colours in diffused daylight,
viewing horizontally against a white background.
Inject alternately the test solution and the reference solution.
Acidity or alkalinity. To 25 ml of filtered extract obtained in
Calculate the content of C13H22N4O3S in the tablets. Colour of extract, add 0.1 ml of dilute phenolphthalein
Storage. Store protected from light and moisture. solution; to another 25 ml add 0.05 ml of methyl orange
Labelling. The label states the strength in terms of the solution. Neither solution shows a pink colour.
equivalent amount of ranitidine. Foreign fibres. When examined under a microscope, it is seen
to consist exclusively of viscose fibres, except that
occasionally a few isolated foreign fibres may be present.
Purified Rayon Fluorescence. Examine a layer about 5 mm in thickness under
ultraviolet light at 365 nm. It displays only a slight, brownish-
Viscose Fibre; absorbent viscose violet fluorescence and a few yellow particles. Not more than
Description. White or very slightly yellow, purified rayon is a a few isolated fibres show an intense blue fluorescence.
fibrous form of bleached regenerated cellulose, which can be
produced with a lustrous or matt appearance, and is soft to Absorbency
the touch. The fibres can be produced with average staple A. Sinking time. Not more than 10 seconds, determined by
length between 32 mm to 80 mm, and are practically odourless. the following method.
Identification Apparatus
A. When examined under a microscope in the dry state, or A dry, cylindrical copper wire basket, 80 mm high and 50 mm in
when mounted in ethanol (95 per cent) and water, the diameter, fabricated from wire of diameter 0.4 mm and having a
following characteristics are observed. They are usually of mesh aperture of 15 to 20 mm; the basket weighs 2.4 to 3.0 g.
more or less uniform width. Many longitudinal parallel lines
are distributed unequally over the width in the case of standard Method
viscose fibres, but such lines are absent or very few in fibres Weigh the basket to the nearest 10 mg. Take five samples,
produced through the zinc-free process. The ends are cut each of approximately 1 g, from different places in the material
more or less straight. Matt fibres contain numerous granular under examination, place loosely in the basket and weigh the
particles of approximately 1µ average diameter. packed basket to the nearest 10 mg. Hold the basket with its
B. Treat with iodinated zinc chloride solution; the fibres long axis in the horizontal position and drop it from a height of
become violet. about 10 mm into water at 25o contained in a beaker at least 12
cm in diameter and filled to a depth of 10 cm. Measure with a
C. To 0.1 g add 10 ml of zinc chloride-formic acid solution, stopwatch the time taken by the basket to sink below the
heat to 40º and allow to stand for 2 hours 30 minutes, shaking surface of the water. Repeat the procedure on two further
occasionally. The fibres dissolve completely except for the samples and calculate the average value.
matt variety where titanium dioxide particles remain.
B. Water-holding capacity. Not less than 18.0 g per g,
D. Dissolve the residue obtained in the test for Sulphated ash determined by the following method.
in 5 ml of sulphuric acid with slight warming, allow to cool,
and carefully add 0.2 ml of hydrogen peroxide solution (10 After the sinking time has been recorded in test A, remove the
volumes). The solution does not undergo colour change in basket from the water, allow it to drain for 30 seconds with its
case of lustrous variety of fibre, but for matt variety an orange- long axis in the horizontal position over the beaker, transfer it
yellow colour is obtained, the intensity of which depends on to a tared beaker and weigh to the nearest 10 mg. Calculate the
the quantity of titanium dioxide present. weight of water retained by the sample. Repeat the procedure
on two further samples and calculate the average value.
Tests
Colouring matter. Slowly extract 10 g in a narrow percolator
Colour of extract. Take 15 g of material under examination in a with ethanol (95 per cent) until 50 ml of extract is obtained.
suitable vessel, add 150 ml of water, close the vessel and The extract is not more intensely coloured than reference

1040
IP 2007 RESERPINE

solution YS5 or GYS6, (2.4.1) or a solution prepared in the Description. White to slightly yellow small crystals or a
following manner. To 3.0 ml of CSS add 7.0 ml of a solution of crystalline powder which darkens slowly on exposure to light.
hydrochloric acid containing 1 per cent w/v of hydrochloric
acid and dilute 0.5 ml of the resulting solution to 10 ml with Identification
the same solution of hydrochloric acid.
Test A may be omitted if tests B, C, D and E are carried out.
Ether-soluble substances. Not more than 0.5 per cent, Tests B, C, D and E may be omitted if test A is carried out.
determined by the following method. Extract 5 g with ether in A. Determine by infrared absorption spectrophotometry (2.4.6).
a continuous extraction apparatus such as a Soxhlet apparatus, Compare the spectrum with that obtained with reserpine RS
for 4 hours in such a way that the rate is at least four extractions or with the reference spectrum of reserpine.
per hour. Evaporate the ether and dry the residue to constant
weight at 105º. B. Dilute 1 ml of a 0.2 per cent w/v solution in chloroform to
100.0 ml with ethanol (95 per cent). When examined
Water-soluble substances. Not more than 0.7 per cent, immediately after preparation, in the range 230 nm to 360 nm
determined by the following method. Boil 5 g with 500 ml of (2.4.7), the resulting solution shows an absorption maximum
water for 30 minutes, stirring frequently and replacing the at about 268 nm; absorbance at about 268 nm, about 0.55.
water lost by evaporation. Decant the liquid into a beaker, Over the range 288 nm to 295 nm, the spectrum exhibits a
squeeze the residual liquid from the material carefully with a slight minimum and then a shoulder or a slight maximum;
glass rod, mix the liquids and filter the extract whilst hot. absorbance over this range, about 0.34.
Evaporate 400 ml of the filtrate and dry the residue to constant
weight at 105º. C. To about 1 mg add 0.1 ml of a 0.1 per cent w/v solution of
sodium molybdate in sulphuric acid; a yellow colour is
Hydrogen sulphide. To 10 ml of the filtered extract obtained produced which changes to blue within 2 minutes.
in the Colour of extract, add 1.9 ml of water, 0.15 ml of dilute
acetic acid and 1 ml of lead acetate solution. After 2 minutes, D. To 1 mg add 0.2 ml of a freshly prepared 1 per cent w/v
the solution is not more intensely coloured than a reference solution of vanillin in hydrochloric acid; a pink colour
solution prepared at the same time using 0.15 ml of dilute develops within 2 minutes.
acetic acid, 1.2 ml of thioacetamide reagent, 1.7 ml of lead E. Mix about 0.5 mg with 5 mg of 4-dimethylaminobenzal-
standard solution (10 ppm Pb) and 10 ml of filtered extract. dehyde and 0.2 ml of glacial acetic acid and add 0.2 ml of
Sulphated ash (2.3.18). Not more than 1.5 per cent. sulphuric acid; a green colour is produced. Add 1 ml of
glacial acetic acid; the colour changes to red.
Loss on drying (2.4.19). Not more than 13.0 per cent, determined
on 5 g by drying in an oven at 105o. Tests
Specific optical rotation (2.4.22). –116º to –128º, determined
in a solution prepared immediately before use by dissolving
Reserpine 0.25 g in sufficient chloroform to produce 25 ml.
Oxidation products. Absorbance of a 0.02 per cent w/v solution
in glacial acetic acid at about 388 nm, measured immediately
H3CO
after preparation, not more than 0.10 (2.4.7).
N
N H Sulphated ash (2.3.18). Not more than 0.1 per cent.
H H OCH3
O Loss on drying (2.4.19). Not more than 0.5 per cent, determined
H on 0.5 g by drying in an oven over phosphorus pentoxide at
O C OCH3
H3COOC 60º at a pressure not exceeding 0.7 kPa for 3 hours.
OCH3
OCH3 Assay. For total alkaloids — Weigh accurately about 0.5 g
and dissolve in a mixture of 40 ml of anhydrous glacial acetic
C33H40N2O9 Mol. Wt. 608.7 acid and 6 ml of acetic anhydride. Titrate with 0.1 M
perchloric acid, determining the end-point potentiometrically
Reserpine is methyl 11,17α-dimethoxy-18β-[(3,4,5-
(2.4.25). Carry out a blank titration.
trimethoxybenzoyl)oxy]-3β,20α-yohimbane-16β-carboxylate.
1 ml of 0.1 M perchloric acid is equivalent to 0.06087 g of
Reserpine contains not less than 99.0 per cent and not more
total alkaloids.
than 101.0 per cent of total alkaloids and not less than 98.0 per
cent and not more than 102.0 per cent of reserpine, C33H40N2O9, For reserpine, C 33H 40N2O 9 — Carry out the following
both calculated on the dried basis. procedure protected from light. Weigh accurately about

1041
RESERPINE INJECTION IP 2007

25.0 mg, moisten with 2 ml of ethanol (95 per cent), add 2 ml D. To 1 mg add 0.2 ml of a freshly prepared 1 per cent w/v
of 0.25 M sulphuric acid and 10 ml of ethanol (95 per cent) solution of vanillin in hydrochloric acid; a pink colour
and warm gently to dissolve. Cool, dilute to 100.0 ml with develops within 2 minutes.
ethanol (95 per cent) and dilute 5.0 ml to 50.0 ml with the E. Mix about 0.5 mg with 5 mg of 4-dimethylaminobenzal-
same solvent (solution A). Transfer 10.0 ml to a boiling tube, dehyde and 0.2 ml of glacial acetic acid and add 0.2 ml of
add 2 ml of 0.25 M sulphuric acid and 2 ml of a freshly prepared sulphuric acid; a green colour is produced. Add 1 ml of
0.3 per cent w/v solution of sodium nitrite, mix and heat in a glacial acetic acid; the colour changes to red.
water-bath at 55º for 35 minutes. Cool, add 1 ml of a freshly
prepared 5 per cent w/v solution of sulphamic acid and dilute Tests
to 25.0 ml with ethanol (95 per cent). Measure the absorbance
of the resulting solution at the maximum at about 388 nm (2.4.7), pH (2.4.24). 3.0 to 4.0.
using as the blank a solution prepared by treating a further Other tests. Comply with the tests stated under Parenteral
10.0 ml of solution A in the same manner and at the same time Preparations (Injections).
but omitting the sodium nitrite solution.
Assay. Protect the solutions from light throughout the assay.
Calculate the content of C33H40N2O9 from the absorbance Measure accurately a volume of the injection containing about
obtained by repeating the operation using reserpine RS in 10 mg of Reserpine and dilute with a 2 per cent w/v solution of
place of the substance under examination. citric acid to 100.0 ml. Extract 10.0 ml of this solution for
Storage. Store protected from light. 2 minutes with three quantities, each of 15 ml, of chloroform.
Wash the combined extracts with 10 ml of a 1 per cent w/v
solution of sodium bicarbonate, add sufficient chloroform to
produce 50.0 ml, mix and evaporate 10.0 ml to dryness on a
Reserpine Injection water-bath. Dissolve the residue in 10 ml of ethanol (95 per
Reserpine Injection is a sterile solution of Reserpine in Water cent), add 2 ml of 0.25 M sulphuric acid and 10 ml of ethanol
for Injections prepared with the aid of a suitable acid. It may (95 per cent) and warm gently to dissolve. Cool, dilute to
contain suitable antioxidants. 100.0 ml with ethanol (95 per cent) and dilute 5.0 ml to 50.0 ml
with the same solvent (solution A). Transfer 10.0 ml to a boiling
Reserpine Injection contains not less than 90.0 per cent and
tube, add 2 ml of 0.25 M sulphuric acid and 2 ml of a freshly
not more than 110.0 per cent of the stated amount of reserpine,
prepared 0.3 per cent w/v solution of sodium nitrite, mix and
C33H40N2O9.
heat in a water-bath at 55º for 35 minutes. Cool, add 1 ml of a
Identification freshly prepared 5 per cent w/v solution of sulphamic acid
and dilute to 25.0 ml with ethanol (95 per cent). Measure the
Extract a suitable volume of the injection containing 10 mg of absorbance of the resulting solution at the maximum at about
Reserpine with 10 ml of chloroform and evaporate the 388 nm (2.4.7), using as the blank a solution prepared by
chloroform layer to dryness. The residue complies with the treating a further 10.0 ml of solution A in the same manner and
following tests. at the same time but omitting the sodium nitrite solution.
Test A may be omitted if tests B, C, D and E are carried out. Calculate the content of C33H40N2O9 from the absorbance
Tests B, C, D and E may be omitted if test A is carried out. obtained by repeating the operation using reserpine RS in
A. Determine by infrared absorption spectrophotometry (2.4.6). place of the substance under examination.
Compare the spectrum with that obtained with reserpine RS Storage. Store protected from light in single dose (or if
or with the reference spectrum of reserpine. stabilising agents are present, in multiple dose) containers.
B. Dilute 1 ml of a 0.2 per cent w/v solution in chloroform to
100 ml with ethanol (95 per cent). When examined immediately
after preparation, in the range 230 nm to 360 nm (2.4.7), the Reserpine Tablets
resulting solution shows an absorption maximum at about
268 nm; absorbance at about 268 nm, about 0.55. Over the Reserpine Tablets contain not less than 90.0 per cent and not
range 288 nm to 295 nm, the spectrum exhibits a slight minimum more than 110.0 per cent of the stated amount of reserpine,
and then a shoulder or a slight maximum; absorbance over C33H40N2O9.
this range, about 0.34.
Identification
C. To about 1 mg add 0.1 ml of a 0.1 per cent w/v solution of
sodium molybdate in sulphuric acid; a yellow colour is A. Powder a few tablets and extract with chloroform. Evaporate
produced which changes to blue within 2 minutes. the extract to dryness, add 0.1 ml of a 0.1 per cent w/v solution

1042
IP 2007 RIBOFLAVINE

of sodium molybdate in sulphuric acid; a yellow colour is cent w/v solution of sodium nitrite, mix and heat in a water-
produced which changes to blue within 2 minutes. bath at 55º for 35 minutes. Cool, add 1 ml of a freshly prepared
B. Powder a few tablets and extract with chloroform. Evaporate 5 per cent w/v solution of sulphamic acid and dilute to 25.0 ml
the extract to dryness, add 0.2 ml of a freshly prepared 1 per with ethanol (95 per cent). Measure the absorbance of the
cent w/v solution of vanillin in hydrochloric acid; a pink resulting solution at the maximum at about 388 nm (2.4.7),
colour develops within 2 minutes. using as the blank a solution prepared by treating a further
10.0 ml of solution A in the same manner and at the same time
Tests but omitting the sodium nitrite solution.

Uniformity of content. Protect the solutions from light Calculate the content of C33H40N2O9 from the absorbance
throughout the test. obtained by repeating the operation using reserpine RS in
place of the substance under examination.
Comply with the test stated under Tablets.
Storage. Store protected from light.
Powder one tablet, disperse in 10 ml of a 2 per cent w/v solution
of citric acid and extract for 2 minutes with three quantities,
each of 5 ml, of chloroform, filter the extracts through a plug
of cotton moistened with chloroform. Wash the chloroform Riboflavine
extracts with 10 ml of a 1 per cent w/v solution of sodium
Lactoflavin; Vitamin B2
bicarbonate and evaporate the chloroform extracts to dryness
on a water-bath. For tablets containing upto 250 µg of
Reserpine per tablet, dissolve the residue in 10.0 ml of ethanol CH2OH
(95 per cent). For tablets containing more than 250 µg of HO C H
Reserpine per tablet, dissolve the residue in a suitable volume HO C H
of ethanol (95 per cent) to give a concentration of 250 µg of HO C H
Reserpine per 10.0 ml; add 2 ml of 0.25 M sulphuric acid and
CH2
2 ml of a freshly prepared 0.3 per cent w/v solution of sodium
nitrite, mix and heat in a water-bath at 55º for 35 minutes. Cool, H3C N N O
add 1 ml of a freshly prepared 5 per cent w/v solution of
NH
sulphamic acid and dilute to 25.0 ml with ethanol (95 per H3C N
cent). Measure the absorbance of the resulting solution at O
the maximum at about 388 nm (2.4.7), using as the blank a
solution prepared by treating a further 10.0 ml of solution A in C17H20N4O6 Mol. Wt. 376.4
the same manner and at the same time but omitting the sodium
nitrite solution. Riboflavine is 3,10-dihydro-7,8-dimethyl-10-[(2S,3S,4R)-
2,3,4,5-tetrahydroxypentyl]benzopteridine-2,4-dione.
Calculate the content of C33H40N2O9 in the tablet from the
absorbance obtained by repeating the operation using Riboflavine contains not less than 98.0 per cent and not more
reserpine RS in place of the substance under examination. than 101.0 per cent of C17H20N4O6, calculated on the dried
basis.
Other tests. Comply with the tests stated under Tablets.
Description. A yellow to orange-yellow, crystalline powder;
Assay. Protect the solutions from light throughout the assay. odour, slight.
Weigh accurately a quantity of the powdered tablets
containing about 1 mg of Reserpine, add 10 ml of a 2 per cent Identification
w/v solution of citric acid and extract for 2 minutes with three A. Determine by infrared absorption spectrophotometry (2.4.6).
quantities, each of 15 ml, of chloroform. Wash the combined Compare the spectrum with that obtained with riboflavine RS
extracts with 10 ml of a 1 per cent w/v solution of sodium or with the reference spectrum of riboflavine.
bicarbonate, add sufficient chloroform to produce 50.0 ml
and evaporate 10.0 ml to dryness on a water-bath. Dissolve B. Dissolve about 1 mg in 100 ml of water. The solution has a
the residue in 10 ml of ethanol (95 per cent) and add 2 ml of pale greenish yellow colour by transmitted light and an intense
0.25 M sulphuric acid and 10 ml of ethanol (95 per cent) and yellowish green fluorescence by reflected light, which
warm gently to dissolve. Cool, dilute to 100.0 ml with ethanol disappears on addition of mineral acids or alkalis.
(95 per cent) and dilute 5.0 ml to 50.0 ml with the same solvent
Tests
(solution A). Transfer 10.0 ml to a boiling tube, add 2 ml of
0.25 M sulphuric acid and 2 ml of a freshly prepared 0.3 per pH (2.4.24). 5.5 to 7.2, determined in a saturated solution.

1043
RIBOFLAVINE SODIUM PHOSPHATE IP 2007

Specific optical rotation (2.4.22). –115º to –135º, determined Riboflavin Sodium Phosphate is monosodium 3,10-dihydro-
in a 0.5 per cent w/v solution in carbonate-free 0.05 M sodium 7,8-dimethyl-10-[(2S,3S,4R)-2,3,4-trihydroxypentyl]
hydroxide. Measure the angle of rotation within 30 minutes of benzopteridine-2,4-dione 5-phosphate dihydrate.
preparing the solution. Riboflavine Sodium Phosphate contains the equivalent of not
Light absorption (2.4.7). Dilute a suitable volume of the final less than 73.0 per cent and not more than 79.0 per cent of
solution obtained in the Assay with an equal volume of water. C17H20N4O6, calculated on the dried basis.
When examined in the range 210 nm to 460 nm, the resulting Description. A yellow to orange-yellow, crystalline powder;
solution exhibits maxima at about 223 nm, 267 nm, 373 nm and hygroscopic.
444 nm; the ratio of the absorbance at the maximum at about
373 nm to that at about 267 nm, 0.31 to 0.33 and the ratio of the Identification
absorbance at the maximum at about 444 nm to that at about
267 nm, 0.36 to 0.39. A. When examined in the range 230 nm to 360 nm (2.4.7), a
0.001 per cent w/v solution in phosphate buffer pH 7.0 shows
Lumiflavine. Shake 25 mg with 10 ml of chloroform for 5 minutes
an absorption maximum at about 267 nm; absorbance at about
and filter. The filtrate is not more intensely coloured than
267 nm, 0.58 to 0.64.
reference solution BYS6 (2.4.1).
Sulphated ash (2.3.18). Not more than 0.1 per cent. B. Dissolve about 10 mg in sufficient 2 M sodium hydroxide
to produce 100 ml, expose 1 ml to ultraviolet light at 254 nm for
Loss on drying (2.4.19). Not more than 1.5 per cent, determined 5 minutes, add sufficient 5 M acetic acid to make the solution
on 1.0 g by drying in an oven at 105º. acidic to litmus paper and shake the mixture with 2 ml of
Assay. Carry out the procedure in subdued light. dichloromethane; the lower layer exhibits a yellow
Weigh accurately about 65 mg and transfer to an amber-glass fluorescence.
500-ml volumetric flask, suspend in 5 ml of water, ensuring C. To 0.5 g add 10 ml of nitric acid, evaporate the mixture to
that it is completely wetted. Dissolve in 5 ml of 2 M sodium dryness on a water-bath, ignite the residue until the carbon is
hydroxide. As soon as dissolution is complete add 100 ml of removed, dissolve the final residue in 5 ml of water and filter.
water and 2.5 ml of glacial acetic acid and dilute to 500.0 ml The filtrate gives the reactions of sodium salts and reaction B
with water. To 20.0 ml of this solution add 3.5 ml of a 1.4 per of phosphates (2.4.1).
cent w/v solution of sodium acetate and dilute to 200.0 ml
with water. Measure the absorbance of the resulting solution Tests
at the maximum at about 444 nm (2.4.7).
pH (2.4.24). 4.0 to 6.3, determined in a 2.0 per cent w/v solution.
Calculate the content of C17H20N4O6 taking 328 as the specific
absorbance at 444 nm. Specific optical rotation (2.4.22). +38.0º to +42.0º, determined
in a 1.5 per cent w/v solution in 5 M hydrochloric acid.
Storage. Store protected from light.
Heavy metals (2.3.13). To 2.0 g in a silica crucible add 2 ml of
nitric acid dropwise followed by 0.25 ml of sulphuric acid.
Riboflavine Sodium Phosphate Heat cautiously until white fumes are evolved and ignite. Extract
the cooled residue with two quantities, each of 2 ml, of
Riboflavine-5-phosphate (Sodium Salt); Vitamin B2 hydrochloric acid and evaporate the extracts to dryness.
Sodium Phosphate Dissolve the residue in 2 ml of 2 M acetic acid and dilute to
20 ml with water. 12 ml of the solution complies with the limit
O test for heavy metals, Method D (10 ppm). Use 1.0 ml of lead
H2C O P ONa standard solution (10 ppm Pb) to prepare the standard.
HO C H OH Lumiflavine. Shake 35 mg with 10 ml of dichloromethane for
HO C H 5 minutes and filter. The filtrate is not more intensely coloured
HO C H than reference solution BYS6 (2.4.1).
CH2
Inorganic phosphate. Not more than 1.5 per cent, determined
H3C N N O , 2H2O by the following method. Dissolve 0.1 g in sufficient water to
NH produce 100 ml. Dilute 5 ml with 10 ml of water and add 5 ml of
H3C N buffered cupric sulphate solution pH 4.0, 2 ml of a 3 per cent
O w/v solution of ammonium molybdate, 1 ml of a freshly
prepared solution containing 2 per cent w/v of 4-methyl-
C17H20N4NaO9P, 2H2O Mol. Wt. 514.4 aminophenol sulphate and 5 per cent w/v of sodium meta-

1044
IP 2007 RIFAMPICIN

bisulphite and 1 ml of a 3 per cent v/v solution of perchloric Other tests. Comply with the tests stated under Tablets.
acid. Add sufficient water to produce 25 ml, mix and measure Assay. Carry out the procedure in subdued light.
the absorbance of the resulting solution at the maximum at
about 800 nm (2.4.7), within 15 minutes of its preparation, Weigh and powder 20 tablets. Weigh accurately a quantity of
using as the blank a solution prepared in the same manner but the powder containing about 10 mg of Riboflavine, add a
omitting the substance under examination. The absorbance is mixture of 5 ml of glacial acetic acid and 100 ml of water and
not greater than that produced by repeating the operation heat on a water-bath for 1 hour with occasional shaking. Dilute
using a solution prepared in the same manner using 15 ml of with 50 ml of water, cool, add 30 ml of 1 M sodium hydroxide
phosphate standard solution (5 ppm PO4) and beginning at with continuous stirring. Add sufficient water to produce
the words “add 5 ml of buffered cupric sulphate solution pH 1000.0 ml, mix and filter, discarding the first few ml of the filtrate.
4.0,” Measure the absorbance of the filtrate at the maximum at about
444 nm (2.4.7).
Loss on drying (2.4.19). Not more than 8.0 per cent, determined
on 0.5 g by drying in an oven at 100º at a pressure not exceeding Calculate the content of C17H20N4O6 taking 328 as the specific
0.7 kPa for 5 hours. absorbance at 444 nm.
Storage. Store protected from light.
Assay. Carry out the procedure protected from light.
Weigh accurately about 0.1 g, dissolve in 150 ml of water, add
2 ml of glacial acetic acid and dilute to 1000.0 ml with water. Rifampicin
To 10.0 ml add 3.5 ml of a 1.4 per cent w/v solution of sodium
acetate, dilute to 50.0 ml with water and measure the Rifampin
absorbance of the resulting solution at the maximum at about
444 nm (2.4.7). Calculate the percentage content of C17H20N4O6
CH3 CH3
taking 328 as the specific absorbance at 444 nm.
HO
Storage. Store protected from light.
H3C O OH O
CH3 CH3
O NH
Riboflavine Tablets H3CO CH3 OH OH
Lactoflavin Tablets; Vitamin B2 Tablets H3C
Riboflavine Tablets contain not less than 90.0 per cent and O N
not more than 115.0 per cent of the stated amount of O N
riboflavine, C17H20N4O6. OH N
O CH3
Identification CH3

Shake a quantity of the powdered tablets containing 1 mg of


C43H58N4O12 Mol. Wt. 823.0
Riboflavine with 100 ml of water and filter; the filtrate has a
pale greenish yellow colour by transmitted light and an intense Rifampicin is (12Z,14E,24E)-(2S,16S,17S,18R,19R,20R,
yellowish green fluorescence by reflected light, which 21S,22R,23S)-1,2-dihydro-5,6,9,17,19-pentahydroxy-23-
disappears on addition of mineral acids or alkalis. methoxy-2,4,12,16,18,20,22-heptamethyl-8-(4-methylpiperazin-
1-yliminomethyl)-1,11-dioxo-2,7-(epoxypentadeca-1,11,13-
Tests trienimino)naphtho[2,1-b]furan-21-yl acetate.
Uniformity of content. Comply with the test stated under Rifampicin contains not less than 97.0 per cent and not more
Tablets. than 102.0 per cent of C43H58N4O12, calculated on the dried
basis.
Powder one tablet, add a mixture of 2.5 ml of glacial acetic
acid and 50 ml of water and heat on a water-bath for 1 hour Description. A brick-red to reddish brown, crystalline powder;
with occasional stirring. Dilute with 50 ml of water, add 15 ml practically odourless.
of 1 M sodium hydroxide with continuous stirring and then
Identification
sufficient water to produce a solution containing 10 µg of
Riboflavine per ml. Filter and discard the first few ml of the A. Determine by infrared absorption spectrophotometry (2.4.6).
filtrate. On the clear filtrate carry out the Assay beginning at Compare the spectrum with that obtained with rifampicin RS
the words “Measure the absorbance....”. or with the reference spectrum of rifampicin.

1045
RIFAMPICIN CAPSULES IP 2007

B. In the Assay, the principal peak in the chromatogram Heavy metals (2.3.13). 1.0 g complies with the limit test for
obtained with the test solution corresponds to the peak in the heavy metals, Method B (20 ppm).
chromatogram obtained with the reference solution. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Tests Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 80º at a pressure not exceeding
pH (2.4.24). 4.5 to 6.5, determined in a 1.0 per cent w/v 0.7 kPa for 4 hours.
suspension.
Assay. Determine by liquid chromatography (2.4.14).
Related substances. Determine by liquid chromatography
NOTE — Prepare the solutions immediately before use.
(2.4.14).
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent
w/v solution of citric acid, 23 volumes of a 13.61 per cent
w/v solution of citric acid, 23 volumes of a 13.61 per cent w/
w/v solution of potassium dihydrogen phosphate, 77 volumes
v solution of potassium dihydrogen phosphate, 77 volumes
of a 17.42 per cent w/v solution of dipotassium hydrogen
of a 17.42 per cent w/v solution of dipotassium hydrogen
phosphate, 640 volumes of water and 250 volumes of
phosphate, 640 volumes of water and 250 volumes of
acetonitrile.
acetonitrile.
Test solution. Dissolve 20.0 mg of the substance under
Test solution. Weigh accurately a quantity of powder
examination in 10.0 ml of acetonitrile and filter. Dilute 5.0 ml of
containing 20 mg of Rifampicin, add 10 ml of acetonitrile,
the filtrate to 100.0 ml with the solvent mixture.
shake and filter. Dilute 5 ml of the filtrate to 50 ml with the
solvent mixture. Reference solution. A solution containing 0.2 per cent w/v of
rifampicin RS in acetonitrile. Dilute 5.0 ml of this solution to
Reference solution. A solution containing 0.02 per cent w/v of
100.0 ml with the solvent mixture.
rifampicin quinone RS in acetonitrile. To 1 ml of the solution,
add 1 ml of the test solution and dilute to 100 ml with the Use the chromatographic system and the system suitability
solvent mixture. parameters described under the test for Related substances.
Chromatographic system Inject the reference solution. The test is not valid unless the
– a stainless steel column 10 cm x 4.6 mm, packed with relative standard deviation for replicate injections is not more
octylsilane bonded to porous silica (5 µm), than 2.0 per cent.
– mobile phase: a mixture of 65 volumes of buffer solution Inject alternately the test solution and the reference solution.
pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per Calculate the content of C43H58N4O12.
cent w/v of citric acid and 2.09 per cent w/v of potassium Storage. Store protected from light, in an atmosphere of
dihydrogen phosphate with water and adjusting the nitrogen.
pH to 6.8 with dilute phosphoric acid, and 35 volumes
of acetonitrile,
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 254 nm, Rifampicin Capsules
– a 20 µl loop injector.
Rifampin Capsules
Inject the reference solution. The test is not valid unless the
resolution between the principal peaks is not less than 4. Rifampicin Capsules contain not less than 92.5 per cent and
not more than 107.5 per cent of the stated amount of rifampicin,
In the chromatogram obtained with the test solution the area C43H58N4O12.
of any peak due to rifampicin quinone is not more than 1.5
times the area of the principal peak in the chromatogram Identification
obtained with the reference solution (1.5 per cent); the area of
A. Shake a quantity of the contents of the capsules containing
any peak, other than the principal peak and the peak
0.15 g of Rifampicin with 5 ml of chloroform, filter and evaporate
corresponding to rifampicin quinone, is not more than the
the filtrate to dryness. The residue complies with the following
area of the peak due to rifampicin in the chromatogram obtained
test.
with the reference solution (1.0 per cent) and the sum of the
areas of any such peaks is not more than 3.5 times the area of Determine by infrared absorption spectrophotometry (2.4.6).
the peak due to rifampicin in the chromatogram obtained with Compare the spectrum with that obtained with rifampicin RS
the reference solution (3.5 per cent ). or with the reference spectrum of rifampicin.

1046
IP 2007 RIFAMPICIN CAPSULES

B. In the Assay, the principal peak in the chromatogram than 4 times the area of the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the obtained with the reference solution, the area of any peak due
chromatogram obtained with the reference solution. to rifampicin N-oxide should not be more than 1.5 times the
area of the principal peak in the chromatogram obtained with
Tests the reference solution and the area of any peak due to
Related substances. Determine by liquid chromatography 3-formylrifamycin SV should not be more than the area of the
(2.4.14). principal peak in the chromatogram obtained with the reference
solution. In the chromatogram obtained with the test solution
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent the area of any unknown peak should not be more than the
w/v solution of citric acid, 23 volumes of a 13.61 per cent area of the principal peak in the chromatogram obtained with
w/v solution of potassium dihydrogen phosphate, 77 volumes the reference solution.
of a 17.42 per cent w/v solution of dipotassium hydrogen
phosphate, 640 volumes of water and 250 volumes of Dissolution (2.5.2).
acetonitrile. Apparatus. No 2
Test solution. Shake a quantity of the contents of the capsules Medium. 900 ml of 0.1 M hydrochloric acid
containing 200 mg of Rifampicin, with 100 ml of acetonitrile Speed and time. 100 rpm and 45 minutes.
and filter. Dilute 5 ml of the filtrate to 50 ml with the solvent Withdraw a suitable volume of the medium and filter promptly
mixture. through a membrane filter disc having an average pore diameter
Reference solution (a). A solution containing 0.02 per cent not greater than 1.0 µm, rejecting the first 1 ml of the filtrate.
w/v of rifampicin RS in acetonitrile. Dilute 1 ml of this solution Dilute the filtrate, if necessary, with the same solvent. Measure
to 100 ml with the solvent mixture. the absorbance (2.4.7) of the resulting solution at the maximum
Reference solution (b). A solution containing 0.01 per cent at about 475 nm. Calculate the content of C43H58N4O12 in the
w/v each of rifampicin RS and rifampicin quinone RS in medium from the absorbance obtained from a solution of
acetonitrile. Dilute 5 ml of this solution to 50 ml with the known concentration of rifampicin RS.
solvent mixture. D. Not less than 75 per cent of the stated amount of
Chromatographic system C43H58N4O12.
– a stainless steel column 10 cm x 4.6 mm, packed with Other tests. Comply with the tests stated under Capsules.
octylsilane bonded to porous silica (5 µm), Assay. Determine by liquid chromatography (2.4.14).
– column. temperature 30º,
– mobile phase: a mixture of 65 volumes of buffer solution Test solution. Shake a quantity of the contents of the capsules
pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric containing about 300.0 mg of Rifampicin, with 200.0 ml of
acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per acetonitrile, filter. Dilute 10.0 ml of the filtrate to 50.0 ml with
cent w/v of citric acid and 2.09 per cent w/v of potassium acetonitrile. Dilute 5.0 ml of this solution to 50.0 ml with the
dihydrogen phosphate with water and adjusting then solvent mixture.
pH to 6.8 with dilute phosphoric acid, and 35 volumes Reference solution (a). A 0.15 per cent w/v solution of
of acetonitrile, rifampicin RS in acetonitrile. Dilute 10.0 ml of this solution to
– flow rate. 1.5 ml per minute, 50.0 ml with acetonitrile. Dilute 5.0 ml of the resulting solution
– spectrophotometer set at 254 nm, to 50.0 ml with the solvent mixture.
– a 20 µl loop injector. Reference solution (b). A solution containing 0.01 per cent
Inject reference solution (b). The test is not valid unless the w/v each of rifampicin RS and rifampicin quinone RS in
resolution between rifampicin and rifampicin quinone is not acetonitrile. Dilute 5ml of this solution to 50 ml with the
less than 4, the tailing factor is not more than 2.0 and the solvent mixture.
column efficiency is not less than 2000 theoretical plates. Use the chromatographic system and system suitability
Inject the test solution and reference solution (a). The relative parameters, as described under the test for Related substances.
retention times are 1.0, 0.55, 1.25 and 2.61 for rifampicin, Inject reference solution (a). The test is not valid unless the
rifampicin quinone, rifampicin N-oxide and 3-formylrifamycin relative standard deviation for replicate injections is not more
SV respectively. The response factors are 1.00, 1.19, 1.03 and than 2.0 per cent.
1.25 for rifampicin, rifampicin quinone, rifampicin N-oxide and
Inject alternately the test solution and reference solution (a).
3-formylrifamycin SV respectively.
Calculate the content of C43H58N4O12 in the capsules.
In the chromatogram obtained with the test solution the area
of any peak due to rifampicin quinone should not be more Storage. Store protected from light and moisture.

1047
RIFAMPICIN ORAL SUSPENSION IP 2007

Rifampicin Oral Suspension – mobile phase: a mixture of 65 volumes of buffer solution


pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
Rifampicin oral suspension contains not less than 90.0 per acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per
cent and not more than 110.0 per cent of the stated amount of cent w/v of citric acid and 2.09 per cent w/v of potassium
rifampicin, C43H58N4O12. dihydrogen phosphate with water and adjusting the
pH to 6.8 with dilute phosphoric acid, and 35 volumes
Identification of acetonitrile,
A. To a quantity containing 0.1 g of Rifampicin add 30 ml of – flow rate. 1.5 ml per minute,
water and shake with two quantities, each of 50 ml, of – spectrophotometer set at 254 nm,
chloroform. Dry the combined extracts with anhydrous sodium – a 20 µl loop injector.
sulphate, filter and evaporate the filtrate to dryness at a Inject reference solution (b). The test is not valid unless the
temperature not exceeding 70°. Wash the residue with 1 ml of resolution between rifampicin and rifampicin quinone is not
ether and dry at 70º. The residue complies with the following less than 4; the tailing factor is not more than 2.0 and the
test. column efficiency is not less than 2000 theoretical plates.
Determine by infrared absorption spectrophotometry (2.4.6). Inject the test solution and reference solution (a). The relative
Compare the spectrum with that obtained with rifampicin RS retention times are 1.0, 0.55, 1.25, 1.51 and 2.61 for rifampicin,
or with the reference spectrum of rifampicin. rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
B. In the Assay, the principal peak in the chromatogram Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
obtained with the test solution corresponds to the peak in the Multiply the areas of each known impurity by their response
chromatogram obtained with the reference solution. factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
for rifampicin, rifampicin quinone, rifampicin N-oxide,
Tests Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
pH (2.4.24). 4.2 to 4.8. In the chromatogram obtained with the test solution the area
of any peak due to rifampicin quinone should not be more
Related substances. Determine by liquid chromatography
than 1.5 times the area of the principal peak in the chromatogram
(2.4.14).
obtained with the reference solution, the area of any peak due
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent to rifampicin N-oxide should not be more than the area of the
w/v solution of citric acid, 23 volumes of a 13.61 per cent principal peak in the chromatogram obtained with the reference
w/v solution of potassium dihydrogen phosphate, 77 volumes solution and the area of any peak due to 3-formylrifamycin SV
of a 17.42 per cent w/v solution of dipotassium hydrogen should not be more than 5.0 times the area of the principal
phosphate, 640 volumes of water and 250 volumes of peak in the chromatogram obtained with the reference solution.
acetonitrile. In the chromatogram obtained with the test solution the area
Test solution. Add 5 ml of water to a quantity of the oral of any unknown peak should not be more than the area of the
suspension containing 20 mg of Rifampicin and extract with principal peak in the chromatogram obtained with the reference
four quantities, each of 10 ml, of dichloromethane, filter the solution.
combined extracts and evaporate to dryness at a temperature Other tests. Comply with the tests stated under Oral
not exceeding 40º. Dissolve the residue in 10 ml of acetonitrile. Suspensions.
Dilute 5 ml of the resulting solution to 50 ml with the solvent
mixture. Assay. Determine by liquid chromatography (2.4.14).

Reference solution (a). A solution containing 0.02 per cent Test solution. Weigh accurately a quantity of the oral
w/v of rifampicin RS in acetonitrile. Dilute 1 ml of this solution suspension containing about 150 mg of rifampicin and dilute
to a 100 ml with the solvent mixture. to 100.0 ml with acetonitrile. Dilute 10.0 ml of this solution to
50.0 ml with acetonitrile. Dilute 10.0 ml of the resulting solution
Reference solution (b). A solution containing 0.01 per cent to 50.0 ml with the solvent mixture.
w/v each of rifampicin RS and rifampicin quinone RS in
acetonitrile. Dilute 5 ml of this solution to 50 ml with the Reference solution (a). A 0.15 per cent w/v solution of
solvent mixture. rifampicin RS in acetonitrile. Dilute 10.0 ml of the solution to
50.0 ml with acetonitrile. Dilute 10.0 ml of the resulting solution
Chromatographic system to 50.0 ml with the solvent mixture.
– a stainless steel column 10 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 µm), Reference solution (b). A solution containing 0.01 per cent
– column temperature 30º, w/v of each of rifampicin RS and rifampicin quinone RS in

1048
IP 2007 RIFAMPICIN TABLETS

acetonitrile. Dilute 5 ml of this solution to 50 ml with the Reference solution (b). A solution containing 0.01 per cent
solvent mixture. w/v of each of rifampicin RS and rifampicin quinone RS in
Use the chromatographic system and the system suitability acetonitrile. Dilute 5 ml of this solution to 50 ml with the
parameters as described under the test for Related substances. solvent mixture.
Chromatographic system
Inject reference solution (a). The test is not valid unless the
– a stainless steel column 10 cm x 4.6 mm, packed with
relative standard deviation for replicate injections is not more
octylsilane bonded to porous silica (5 µm),
than 2.0 per cent.
– column. temperature 30º,
Inject alternately the test solution and reference solution (a). – mobile phase: a mixture of 65 volumes of buffer solution
Determine the weight per ml of the suspension (2.4.29) and pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
calculate the content of C43H58N4O12 weight in volume. acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per
cent w/v of citric acid and 2.09 per cent w/v of potassium
Storage. Store protected from light and moisture. dihydrogen phosphate with water and adjusting the
pH to 6.8 with dilute phosphoric acid, and 35 volumes
of acetonitrile,
Rifampicin Tablets – flow rate. 1.5 ml per minute,
– spectrophotometer set at 254 nm,
Rifampin Tablets – a 20 µl loop injector.
Rifampicin Tablets contain not less than 92.5 per cent and not Inject reference solution (b). The test is not valid unless the
more than 107.5 per cent of the stated amount of rifampicin, resolution between rifampicin and rifampicin quinone is not
C43H58N4O12. less than 4, the tailing factor is not more than 2.0 and the
column efficiency is not less than 2000 theoretical plates.
Identification
Inject the test solution and reference solution (a). The relative
A. Shake a quantity of the powdered tablets containing 0.15 g retention times are 1.0, 0.55, 1.25 and 2.61 for rifampicin,
of Rifampicin with 5 ml of chloroform, filter and evaporate the rifampicin quinone, rifampicin N-oxide, and 3-formylrifamycin
filtrate to dryness. The residue complies with the following SV respectively. The response factors are 1.00, 1.19, 1.03 and
test. 1.25 for rifampicin, rifampicin quinone, rifampicin N-oxide and
Determine by infrared absorption spectrophotometry (2.4.6). 3-formylrifamycin SV respectively.
Compare the spectrum with that obtained with rifampicin RS In the chromatogram obtained with the test solution the area
or with the reference spectrum of rifampicin. of any peak due to rifampicin quinone should not be more
B. In the Assay, the principal peak in the chromatogram than 1.5 times the area of the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the obtained with the reference solution, the area of any peak due
chromatogram obtained with the reference solution. to rifampicin N-oxide should not be more than the area of the
principal peak in the chromatogram obtained with the reference
Tests solution and the area of any peak due to 3-formylrifamycin SV
Related substances. Determine by liquid chromatography should not be more than 5.0 times the area of the principal
(2.4.14). peak in the chromatogram obtained with the reference solution.
In the chromatogram obtained with the test solution the area
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent w/ of any unknown peak should not be more than the area of the
v solution of citric acid, 23 volumes of a 13.61 per cent solution principal peak in the chromatogram obtained with the reference
w/v of potassium dihydrogen phosphate, 77 volumes of a solution.
17.42 per cent w/v solution of dipotassium hydrogen
phosphate, 640 volumes of water and 250 volumes of Dissolution (2.5.2).
acetonitrile. Apparatus. No 2
Test solution. Weigh accurately a quantity of the powdered Medium. 900 ml of 0.1 M hydrochloric acid.
tablets containing 200 mg of Rifampicin, dissolve in 100 ml of Speed and time. 100 rpm and 45 minutes.
acetonitrile, filter. Dilute 5 ml of the filtrate to 50 ml with the Withdraw a suitable volume of the medium and filter promptly
solvent mixture. through a membrane filter disc having an average pore diameter
Reference solution (a). A solution containing 0.02 per cent not greater than 1.0 µm, rejecting the first 1 ml of the filtrate.
w/v of rifampicin RS in acetonitrile. Dilute 1 ml of this solution Dilute the filtrate, if necessary, with the same solvent. Measure
to a 100 ml with the solvent mixture. the absorbance (2.4.7) of the resulting solution at the maximum

1049
RIFAMPICIN AND ISONIAZID TABLETS IP 2007

at about 475 nm. Calculate the content of C43H58N4O12 in the Solvent mixture. A mixture of 10 volumes of a 21.01 per cent
medium from the absorbance obtained from a solution of w/v solution of citric acid, 23 volumes of a 13.61 per cent
known concentration of rifampicin RS. w/v solution of potassium dihydrogen phosphate, 77 volumes
D. Not less than 75 per cent of the stated amount of of a 17.42 per cent w/v solution of dipotassium hydrogen
C43H58N4O12. phosphate, 640 volumes of water and 250 volumes of
acetonitrile.
Other tests. Comply with the tests stated under Tablets.
Test solution. Weigh accurately a quantity of the powdered
Assay. Determine by liquid chromatography (2.4.14). tablets containing 200 mg of Rifampicin in 100 ml of acetonitrile
Test solution. Weigh and powder 20 tablets. Weigh accurately and filter. Dilute 5 ml of the filtrate to 50 ml with the solvent
a quantity of the powder containing about 300.0 mg of mixture.
Rifampicin, dissolve in 200.0 ml of acetonitrile, filter. Dilute Reference solution (a). Dissolve rifampicin RS in acetonitrile
10.0 ml of the filtrate to 50.0 ml with acetonitrile. Dilute 5.0 ml to obtain a solution containing 0.2 mg per ml. Dilute 1 ml of
of this solution to 50.0 ml with the solvent mixture. this solution to 100 ml with the solvent mixture.
Reference solution (a). A 0.15 per cent w/v solution of Reference solution (b). A solution containing 0.01 per cent
rifampicin RS in acetonitrile. Dilute 10.0 ml of this solution to w/v each of rifampicin RS and rifampicin quinone RS in
50.0 ml with acetonitrile. Dilute 5.0 ml of the resulting solution acetonitrile. Dilute 5 ml of this solution to 50 ml with the
to 50.0 ml with the solvent mixture. solvent mixture.
Reference solution (b). A solution containing 0.01 per cent Chromatographic system
w/v each of rifampicin RS and rifampicin quinone RS in – a stainless steel column 10 cm x 4.6 mm, packed with
acetonitrile. Dilute 5.0 ml of this solution to 50.0 ml with the octylsilane bonded to porous silica (5 µm),
solvent mixture. – column temperature 30º,
Use the chromatographic system and system suitability – mobile phase: a mixture of 65 volumes of buffer solution
parameters described under the test for Related substances. pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per
Inject reference solution (a). The test is not valid unless the cent w/v of citric acid and 2.09 per cent w/v of potassium
relative standard deviation for replicate injections is not more dihydrogen phosphate with water and adjusting the
than 2.0 per cent. pH to 6.8 with dilute phosphoric acid, and 35 volumes
Inject alternately the test solution and reference solution (a). of acetonitrile,
– flow rate. 1.5 ml per minute,
Calculate the content of C43H58N4O12 in the tablets.
– spectrophotometer set at 254 nm,
Storage. Store protected from light and moisture. – a 20 µl loop injector.
Inject reference solution (b). The test is not valid unless the
resolution between rifampicin and rifampicin quinone is not
less than 4, the tailing factor is not more than 2.0 and the
Rifampicin and Isoniazid Tablets column efficiency is not less than 2000 theoretical plates.
Rifampin and Isonicotinylhydrazid Tablets Inject alternately the test solution and reference solution (a).
Rifampicin and Isoniazid Tablets contain not less than The relative retention times are 1.0, 0.55, 1.25, 1.51 and 2.61 for
90.0 per cent and not more than 110.0 per cent of the stated rifampicin, rifampicin quinone, rifampicin N-oxide,
amounts of rifampicin, C43H58N4O12 and isoniazid, C6H7N3O. 3-formylrifamycin SV isonicotinyl hydrazone and
3-formylrifamycin SV respectively. Multiply the area of each
Identification known impurity by its response factor. The response factors
are 1.00, 1.19, 1.03, 1.22 and 1.25 for rifampicin, rifampicin
A. In the Assay, the chromatogram obtained with the test quinone, rifampicin N-oxide, isonicotinyl hydrazone and
solution shows peaks that correspond to the peaks due to 3-formylrifamycin SV respectively.
rifampicin RS and isoniazid RS in the chromatogram obtained
In the chromatogram obtained with the test solution the area
with the reference solution.
of any peak due to rifampicin quinone should not be more
than 4 times the area of the principal peak in the chromatogram
Tests
obtained with the reference solution (4 per cent), the area of
Related substances. Determine by liquid chromatography any peak due to rifampicin N-oxide should not be more than
(2.4.14). 1.5 times the area of the principal peak in the chromatogram

1050
IP 2007 RIFAMPICIN AND ISONIAZID TABLETS

obtained with the reference solution (1.5 per cent), the area of Inject alternately the test solution and the reference solution.
any peaks due to 3-formylrifamycin SV and isonicotinyl Calculate the content of C6H7N3O in the dissolution medium.
hydrazone should not be more than 5 times the area of the
principal peak in the chromatogram obtained with the reference D. Not less than 75 per cent of the stated amounts of
solution (5 per cent) and the area of any peak due to C43H58N4O12 and C6H7N3O.
3-formylrifamycin SV should not be more than the area of the Other tests. Comply with the tests stated under Tablets.
principal peak in the chromatogram obtained with the reference
Assay. Determine by liquid chromatography (2.4.14).
solution (1.0 per cent). In the chromatogram obtained with the
test solution the area of any unknown peak should not be Solvent mixture. Dissolve 1.4 g of disodium hydrogen
more than 1.5 times the area of the principal peak in the orthophosphate anhydrous in 1000 ml of water and adjust
chromatogram obtained with the reference solution ( 1.5 per the pH to 6.8 with dilute phosphoric acid.
cent). Test solution. Weigh and powder 20 tablets. Weigh accurately
Dissolution (2.5.2). a quantity of the powder containing about 40 mg of Isoniazid,
dissolve in 100.0 ml of methanol and dilute to 500.0 ml with the
Apparatus. No 2
solvent mixture.
Medium. 900 ml of 0.1 M hydrochloric acid.
Speed and time. 100 rpm and 45 minutes. Reference solution. A solution containing 0.08 per cent w/v of
rifampicin RS and 0.04 per cent w/v of isoniazid RS in
Withdraw a suitable volume of the medium and filter promptly methanol. Dilute 10.0 ml of this solution to 50.0 ml with the
through a membrane filter disc with an average pore diameter solvent mixture.
not greater than 0.8 µm. Reject the first few ml of the filtrate
and dilute a suitable volume of the filtrate with the medium. Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with
Reference stock solution. A solution containing 0.0165 per octadecylsilane bonded to porous silica (5 µm),
cent of rifampicin RS and 0.00825 per cent of isoniazid RS in – column. temperature 30º,
the dissolution medium. – mobile phase: A. a mixture of 96 volumes of buffer
For rifampicin — Measure the absorbance of the sample solution pH 6.8 prepared by dissolving 1.4 g of disodium
solution and the reference stock solution suitably diluted with hydrogen orthophosphate anhydrous in 1000 ml of
the dissolution medium at 475 nm (2.4.7). Calculate the content water and adjusting the pH to 6.8 ± 0.05 with dilute
of C43H58N4O12 in the medium from the absorbance obtained phosphoric acid, and 4 volumes of acetronitrile.
from the reference stock solution. B. a mixture of 45 volumes of the buffer
solution and 55 volumes of acetonitrile,
For isoniazid — Determine by liquid chromatography – flow rate. 1.5 ml per minute,
(2.4.14). – a linear gradient programme using the conditions given
Use the reference stock solution and sample solution suitably below,
diluted with a solution of 0.05 M potassium dihydrogen – spectrophotometer set at 238 nm,
orthophosphate, adjust the pH to 6.2 with 0.1 M sodium – a 20 µl loop injector.
hydroxide to obtain the reference solution and the test Time Mobile phase A Mobile phase B
solution, respectively. (in min.) (per cent v/v) (per cent v/v)
Chromatographic system 0 100 0
– a stainless steel column 30 cm x 3.9 mm, packed with 5 100 0
octadecylsilane bonded to porous silica (5 µm), 6 0 100
– mobile phase: a mixture of 99 volumes of 0.05 M 15 0 100
potassium dihydrogen phosphate and 1 volume of
16 100 0
acetonitrile with the pH adjusted to 4.0 ± 0.05 with 2 per
cent w/v solution of phosphoric acid, 20 100 0
– flow rate. 1 ml per minute, NOTE — Saturate the column with mobile phase B for about
– spectrophotometer set at 254 nm, 1 hour before injection.
– a 20 µl loop injector.
Inject the reference solution. The tailing factor is not more
Inject the reference solution. The test is not valid unless the than 2.0 for rifampicin and isoniazid; the column efficiency for
tailing factor is not more than 2.0, the column efficiency in not the isoniazid peak is not less than 3000 and for rifampicin not
less than 1500 theoretical plates and the relative standard less than 25000 theoretical plates and the relative standard
deviation for replicate injections is not more than 2.0 per cent. deviation for replicate injections is not more than 2.0 per cent.

1051
RIFAMPICIN, ISONIAZID AND ETHAMBUTOL TABLETS IP 2007

Inject alternately the test solution and the reference solution. Chromatographic system
The retention times are about 1.0 for rifampicin and about 0.3 – a stainless steel column 10 cm x 4.6 mm, packed with
for isoniazid. octylsilane bonded to porous silica (5 µm),
Calculate the contents of C43H58N4O12 and C6H7N3O in the – column. temperature 30º,
tablets. – mobile phase: a mixture of 65 volumes of buffer solution
pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
Storage. Store protected from moisture. acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per
cent w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate with water and adjusting the
pH to 6.8 with dilute phosphoric acid, and 35 volumes
Rifampicin, Isoniazid and Ethambutol of acetonitrile,
Tablets – flow rate. 1.5 ml per minute,
– spectrophotometer set at 254 nm,
Rifampin, Isonicotinylhydrazid and Ethambutol
– a 20 µl loop injector.
Hydrochloride Tablets
Inject reference solution (b). The test is not valid unless the
Rifampicin, Isoniazid and Ethambutol Tablets contain not less resolution between rifampicin and rifampicin quinone is not
than 90.0 per cent and not more than 110.0 per cent of the less than 4, the tailing factor is not more than 2.0 and the
stated amounts of rifampicin, C43H58N4O12, isoniazid, C6H7N3O column efficiency is not less than 2000 theoretical plates.
and ethambutol hydrochloride C10H24N2O2.2HCl.
Inject the test solution and reference solution (a). The relative
Identification retention times are 1.0, 0.55, 1.25, 1.51 and 2.61 for rifampicin,
rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
A. In the Assay for rifampicin and isoniazid, the principal Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
peaks in the chromatogram obtained with the test solution Multiply the areas of each known impurity by its response
correspond to the peaks in the chromatogram obtained with factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
the reference solution. for rifampicin, rifampicin quinone, rifampicin N-oxide,
B. In the Assay for ethambutol hydrochloride the principal Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
peak in the chromatogram obtained with the test solution
In the chromatogram obtained with the test solution the area
corresponds to the peak in the chromatogram obtained with
of any peak due to rifampicin quinone should not be more
the reference solution.
than 4 times the area of the principal peak in the chromatogram
Tests obtained with the reference solution (4 per cent), the area of
any peak due to rifampicin N-oxide should not be more than
Related substances. Determine by liquid chromatography 1.5 times the area of the principal peak in the chromatogram
(2.4.14). obtained with the reference solution (1.5 per cent), the area of
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent any peak due to 3-formylrifamycin SV Isonicotinyl hydrazone
w/v solution of citric acid, 23 volumes of a 13.61 per cent should not be more than 5 times the area of the principal peak
w/v solution of potassium dihydrogen phosphate, 77 volumes in the chromatogram obtained with the reference solution
of a 17.42 per cent w/v solution of dipotassium hydrogen ( 5 per cent ) and the area of any peak due to 3-formylrifamycin
phosphate, 640 volumes of water and 250 volumes of SV should not be more than the area of the principal peak in
acetonitrile. the chromatogram obtained with the reference solution (1 per
cent ). In the chromatogram obtained with the test solution
Test solution. Weigh accurately a quantity of the powdered the area of any unknown peak should not be more than 1.5
tablets containing 200 mg of Rifampicin, dissolve in 100 ml of times the area of the principal peak in the chromatogram
acetonitrile and filter. Dilute 5 ml of this solution to 50 ml with obtained with the reference solution (1.5 per cent).
the solvent mixture.
Dissolution (2.5.2).
Reference solution (a). A solution containing 0.02 per cent
w/v of rifampicin RS in acetonitrile. Dilute 1 ml of this solution Apparatus. No 2
to 100 ml with the solvent mixture. Medium. 900 ml of 0.1 M hydrochloric acid.
Speed and time. 100 rpm and 45 minutes.
Reference solution (b). A solution containing 0.01 per cent
w/v each of rifampicin RS and rifampicin quinone RS in Withdraw a suitable volume of the medium and filter promptly
acetonitrile. Dilute 5 ml of this solution to 50 ml with the through a membrane filter disc with an average pore diameter
solvent mixture. not greater than 0.8 µm. Reject the first few ml of the filtrate.

1052
IP 2007 RIFAMPICIN, ISONIAZID AND ETHAMBUTOL TABLETS

Reference stock solution. A solution containing 0.0165 per Other tests. Comply with the tests stated under Tablets.
cent w/v of rifampicin RS, 0.00825 per cent w/v of isoniazid Assay. For rifampicin and isoniazid — Determine by liquid
RS and 0.031 per cent w/v of ethambutol hydrochloride RS in chromatography (2.4.14).
the dissolution medium. Keep this reference stock solution in
the dissolution bath during the test run. Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powdered tablets containing about 40 mg of
For rifampicin — Dilute the filtrate, if necessary, with the Isoniazid dissolve in 100.0 ml of methanol, dilute to 500.0 ml
same solvent. Measure the absorbance (2.4.7) of the resulting with the diluent and mix.
solution at the maximum at about 475 nm. Calculate the content
of C43H58N4O12 in the medium from the absorbance obtained Reference solution. A solution containing 0.08 per cent w/v of
from suitably diluted reference stock solution. rifampicin RS and 0.04 per cent w/v of isoniazid RS in
methanol. Dilute 10.0 ml of this solution to 50.0 ml with the
For isoniazid — Determine by liquid chromatography (2.4.14). diluent.
Test solution. Suitably dilute the filtered medium with 0.05 M Chromatographic system
potassium dihydrogen orthophosphate with the pH adjusted – a stainless steel column 25 cm x 4.6 mm, packed with
to 6.2 with 0.1 M sodium hydroxide. octadecylsilane bonded to porous silica (5 µm),
Reference solution. Suitably dilute the reference stock solution –column. temperature 30°,
with 0.05 M potassium dihydrogen orthophosphate with the – mobile phase: A. a mixture of 96 volumes of buffer
pH adjusted to 6.2 with 0.1 M sodium hydroxide. solution pH 6.8 (diluent) prepared by dissolving 1.4 g of
disodium hydrogen orthophosphate anhydrous in
Chromatographic system 1000 ml of water, and adjusting the pH to 6.8 ± 0.05 with
– a stainless steel column 30 cm x 4.6 mm, packed with dilute phosphoroic acid, and 4 volumes of acetonitrile,
octadecylsilane bonded to porous silica (5 µm), B: a mixture of 45 volumes of the buffer
– mobile phase: a mixture of 99 volumes of 0.05 M solution and 55 volumes of acetonitrile,
potassium dihydrogen phosphate and 1 volume of – flow rate. 1.5 ml per minute,
acetonitrile previously adjusted to pH 4.0 with a 2 per – a linear gradient programme using the conditions given
cent w/v solution of phosphoric acid, below,
– flow rate. 1 ml per minute, – spectrophotometer set at 238 nm,
– spectrophotometer set at 254 nm, – a 20 µl loop injector.
– a 20 µl loop injector.
Time Mobile phase A Mobile phase B
Inject the reference solution. The test is not valid unless the (in min.) (per cent v/v) (per cent v/v)
tailing factor is not more than 2.0, the column efficiency in not 0 100 0
less than 1500 theoretical plates and the relative standard 5 100 0
deviation for replicate injections is not more than 2.0 per cent.
6 0 100
Inject alternately the test solution and the reference solution. 15 0 100
Calculate the content of C6H7N3O in the dissolution medium. 16 100 0
For ethambutol hydrochloride — Determine by liquid 22 100 0
chromatography (2.4.14). NOTE — Saturate the column with mobile phase B for about
Test solution. Suitably dilute the filtered medium with 0.05 M 1 hour.
potassium dihydrogen orthophosphate adjusted to pH 6.2 Inject the reference solution. The test is not valid unless the
with 0.1 M sodium hydroxide. tailing factor is not less than 2.0 for rifampicin and isoniazid,
Reference solution. Suitably dilute the reference stock solution the column efficiency determined from isoniazid peak is not
with 0.05 M potassium dihydrogen orthophosphate with the less than 3000 and that from rifampicin is not less than 25000
pH adjusted to 6.2 with 0.1 M sodium hydroxide. theoretical plates respectively, and the relative standard
deviation for replicate injections is not more than 2.0 per cent.
Use the chromatographic system described under the Assay The relative retention times are about 1.0 for rifampicin and
of ethambutol hydrochloride. about 0.3 for isoniazid.
Calculate the content of C10H24N2O2.2HCl in the dissolution Calculate the contents of C43H58N4O12 and C6H7N3O in the
medium. tablets.
D. Not less than 75 per cent of the stated amounts of For ethambutol hydrochloride — Determine by liquid
C43H58N4O12, C6H7N3O and C10H24N2O2.2HCl. chromatography (2.4.14).

1053
RIFAMPICIN, ISONIAZID AND PYRAZINAMIDE TABLETS IP 2007

Test solution. Weigh accurately a quantity of the powdered phosphate, 640 volumes of water and 250 volumes of
tablets containing about 60 mg of Ethambutol Hydrochloride acetonitrile.
and dissolve in 100.0 ml of the diluent. Test solution. Weigh accurately a quantity of the powdered
Reference solution. A 0.06 per cent w/v solution of ethambutol tablets containing about 200 mg of Rifampicin, dissolve in
hydrochloride RS in the diluent. 100 ml of acetonitrile and filter. Dilute 5 ml of this solution to
50 ml with the solvent mixture.
Chromatographic system
– a stainless steel column 15 cm x 4.6 mm, packed with Reference solution (a). A solution containing 0.02 per cent
nitrile groups chemically bonded to porous silica w/v of rifampicin RS in acetonitrile. Dilute 1ml of this solution
particles 5 (µm) (such as Zorbax SB CN), to a 100 ml with the solvent mixture.
– mobile phase: a mixture of 50 volumes of acetonitrile, Reference solution (b). A solution containing 0.01 per cent
and 50 volumes of a buffer solution pH 7.0 prepared by w/v each of rifampicin RS and rifampicin quinone RS in
dissolving 1 ml of triethylamine in 1000 ml of water and acetonitrile. Dilute 5 ml of this solution to 50 ml with the
adjusting the pH to 7.0 with dilute phosphoric acid, solvent mixture.
– flow rate. 1 ml per minute, Chromatographic system
– spectrophotometer set at 200 nm, – a stainless steel column 10 cm x 4.6 mm, packed with
– a 20 µl loop injector. octylsilane bonded to porous silica (5 µm),
Inject the reference solution. The test is not valid unless the – column. temperature 30º,
relative standard deviation for replicate injections is not more – mobile phase: a mixture of 65 volumes of buffer solution
than 2.0 per cent, the tailing factor is not more than 3.0 and the pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
column efficiency determined from Ethambutol hydrochloride acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per
peak is not less than 1500 theoretical plates. cent w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate with water and adjusting the
Inject alternately the test solution and the reference solution. pH to 6.8 with dilute phosphoric acid, and 35 volumes
Calculate the content of C10H24N2O2.2HCl in the tablets. of acetonitrile,
– flow rate. 1.5 ml per minute,
Storage. Store protected from moisture.
– spectrophotometer set at 254 nm,
– a 20 µl loop injector.
Inject reference solution (b). The test is not valid unless the
Rifampicin, Isoniazid and resolution between rifampicin and rifampicin quinone is not
Pyrazinamide Tablets less than 4, the tailing factor is not more than 2.0 and the
column efficiency is not less than 2000 theoretical plates.
Rifampin, Isonicotinylhydrazid and Pyrazinamide Tablets
Inject the test solution and reference solution (a). The relative
Rifampicin, Isoniazid and Pyrazinamide Tablets contain not retention times are 1.0, 0.55, 1.25, 1.51 and 2.61 for rifampicin,
less than 90.0 per cent and not more than 110.0 per cent of the rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
stated amounts of rifampicin, C43H58N4O12, isoniazid, C6H7N3O Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
and pyrazinamide C5H5N3O. Multiply the area of each known impurity by its response
factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
Identification for rifampicin, rifampicin quinone, rifampicin N-oxide,
Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
In the Assay of rifampicin, isoniazid and pyrazinamide, the
principal peaks in the chromatogram obtained with the test In the chromatogram obtained with the test solution the area
solution correspond to the peaks in the chromatogram of any peak due to rifampicin quinone should not be more
obtained with the reference solution. than 4 times the area of the principal peak in the chromatogram
obtained with the reference solution (4 per cent), the area of
Tests any peak due to rifampicin N-oxide should not be more than
1.5 times the area of the principal peak in the chromatogram
Related substances. Determine by liquid chromatography obtained with the reference solution (1.5 per cent), the area of
(2.4.14). any peak due to 3-formylrifamycin SV Isonicotinyl hydrazone
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent should not be more than 5 times the area of the principal peak
w/v solution of citric acid, 23 volumes of a 13.61 per cent in the chromatogram obtained with the reference solution
w/v solution of potassium dihydrogen phosphate, 77 volumes ( 5 per cent ) and the area of any peak due to 3-formylrifamycin
of 17.42 per cent w/v solution of dipotassium hydrogen SV should not be more than the area of the principal peak in

1054
IP 2007 RIFAMPICIN, ISONIAZID AND PYRAZINAMIDE TABLETS

the chromatogram obtained with the reference solution ( 1 per The test is not valid unless in the chromatogram obtained
cent ). In the chromatogram obtained with the test solution with reference solution (b) the resolution between isonicotinic
the area of any unknown peak should not be more than 1.5 acid and pyrazinamide is not more than 2.5 and between
times the area of the principal peak in the chromatogram pyrazinamide and isoniazid is not more than 4.0.
obtained with the reference solution (1.5 per cent). Calculate the contents of C6H7N3O and C5H5N3O in the medium.
Dissolution (2.5.2). D. Not less than 75 per cent of the stated amounts of
Apparatus. No 2 C43H58N4O12, C6H7N3O and C5H5N3O.
Medium. 900 ml of a solution containing 2 g of sodium chloride
Other tests. Comply with the tests stated under Tablets.
and 7.0 ml of hydrochloric acid in 1000 ml of water, with a pH
of about 1.2. Assay. For rifampicin, isoniazid and pyrazinamide —
Speed and time. 100 rpm and 45 minutes. Determine by liquid chromatography (2.4.14).
Withdraw a suitable volume of the medium and filter through Test solution. Weigh and powder 20 tablets. Weigh accurately
a membrane filter disc with an average pore diameter not greater a quantity of the powdered tablets containing about 40 mg of
than 0.8 µm. Reject the first few ml of the filtrate. Isoniazid, dissolve in 100.0 ml of methanol, dilute to 500 ml
with the solvent mixture and mix.
Reference stock solution. A solution containing 0.0165 per
cent w/v of rifampicin RS, 0.00825 per cent w/v of isoniazid Reference solution. A solution containing 0.08 per cent w/v of
RS and 0.04375 per cent w/v of pyrazinamide RS in the rifampicin RS, 0.04 per cent w/v of isoniazid RS and 0.2 per
dissolution medium and filtered. Keep this reference stock cent w/v of pyrazinamide RS in methanol. Dilute 10.0 ml of
solution in the dissolution bath during the test run. this solution to 50.0 ml with the solvent mixture.
For rifampicin — Dilute the filtrate, if necessary, with the Chromatographic system
same solvent. Measure the absorbance (2.4.7) of the resulting – a stainless steel column 25 cm x 4.6 mm, packed with
solution at the maximum at about 475 nm. Calculate the content octadecylsilane bonded to porous silica (5 µm),
of C43H58N4O12 in the medium from the absorbance obtained – column. temperature 30º,
from suitably diluted reference stock solution. – mobile phase: A. a mixture of 96 volumes of buffer
For isoniazid and pyrazinamide — Determine by liquid solution pH 6.8 (diluent) prepared by dissolving 1.4 g of
chromatography (2.4.14). disodium hydrogen orthophosphate anhydrous in
1000 ml of water, adjusted to pH 6.8 ± 0.05 with dilute
NOTE — Use this solution within one hour from preparation.
phosphoroic acid and 4 volumes of acetonitrile,
Test solution. To 15.0 ml of the filtered medium, add 15 ml of B. a mixture of 45 volumes of the buffer
1 M dibasic potassium phosphate, dilute to 100.0 ml with the solution and 55 volumes of acetonitrile,
mobile phase and mix. – flow rate. 1.5 ml per minute,
Reference solution (a). To 15.0 ml of the reference stock – a linear gradient programme using the conditions given
solution, add 15 ml of 1 M dibasic potassium phosphate, below,
dilute to 100.0 ml with the mobile phase and mix. – spectrophotometer set at 238 nm,
Reference solution (b). To 10.0 ml of a 0.0125 per cent w/v – a 20 µl loop injector.
solution of isonicotinic acid in the dissolution medium, add Time Mobile phase A Mobile phase B
4.0 ml of the reference stock solution and 15 ml of 1 M dibasic (in min.) (per cent v/v) (per cent v/v)
potassium phosphate, dilute to 100.0 ml with the mobile phase 0 100 0
and mix. 5 100 0
Chromatographic system 6 0 100
– a stainless steel column 30 cm x 4.6 mm, packed with 15 0 100
octylsilane bonded to porous silica (5 µm), 16 100 0
– mobile phase: a mixture of 860 volumes of water, 22 100 0
100 volumes of 1 M monobasic potassium phosphate
and 40 volumes of acetonitrile, NOTE — Saturate the column with mobile phase (B) for
– flow rate. 1 ml per minute, about 1 hour.
– spectrophotometer set at 254 nm, Inject the test solution and the reference solution. The test is
– a 50 µl loop injector. not valid unless the tailing factor is not less than 2.0 for
Inject the test solution and reference solutions (a) and (b). rifampicin, isoniazid and pyrazinamide, the column efficiencies
The relative retention times are about 0.7 for isonicotinic acid, determined from Isoniazid, pyrazinamide and that from
1.0 for pyrazinamide and 1.8 for isoniazid. rifampicin are not less than 3000, 5000 and 25000 theoretical

1055
RIFAMPICIN, ISONIAZID, PYRAZINAMIDE AND ETHAMBUTOL TABLETS IP 2007

plates respectively, and the relative standard deviation for acetonitrile. Dilute 5 ml of this solution to 50 ml with the
replicate injections is not more than 2.0 per cent. The relative solvent mixture.
retention times are about 1.8 for rifampicin, about 0.7 for Chromatographic system
isoniazid and about 1.0 for pyrazinamide. – a stainless steel column 10 cm x 4.6 mm, packed with
Inject alternately the test solution and the reference solution. octylsilane bonded to porous silica (5 µm),
– column. temperature 30º,
Calculate the contents of C43H58N4O12, C6H7N3O and C5H5N3O
– mobile phase: a mixture of 65 volumes of buffer solution
in the tablets.
pH 6.8 prepared by mixing 0.1 per cent w/v of phosphoric
Storage. Store protected from moisture. acid, 0.19 per cent w/v of sodium perchlorate, 0.59 per
cent w/v of citric acid and 2.09 per cent w/v of potassium
dihydrogen phosphate with water, adjusting the pH to
Rifampicin, Isoniazid, Pyrazinamide 6.8 with dilute phosphoric acid, and 35 volumes of
and Ethambutol Tablets acetonitrile,
– flow rate. 1.5 ml per minute,
Rifampin, Isonicotinylhydrazid, Pyrazinamide and – spectrophotometer set at 254 nm,
Ethambutol Hydrochloride Tablets – a 20 µl loop injector.
Rifampicin, Isoniazid, Pyrazinamide and Ethambutol Tablets Inject reference solution (b). The test is not valid unless the
contain not less than 90.0 per cent and not more than 110.0 per resolution between rifampicin and rifampicin quinone is not
cent of the stated amounts of rifampicin, C43H58N4O12, isoniazid, less than 4, the tailing factor is not more than 2.0 and the
C 6 H 7 N 3 O, pyrazinamide, C 5 H 5 N 3 O and ethambutol column efficiency is not less than 2000 theoretical plates.
hydrochloride, C10H24N2O2.2HCl. Inject the test solution and reference solution (a). The relative
retention times are 1.0, 0.55, 1.25, 1.51 and 2.61 for rifampicin,
Identification rifampicin quinone, rifampicin N-oxide, 3-formylrifamycin SV
A. In the Assay for rifampicin, isoniazid and pyrazinamide, the Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
principal peaks in the chromatogram obtained with the test Multiply the area of each known impurity by its response
solution correspond to the peaks in the chromatogram factor. The response factors are 1.00, 1.19, 1.03, 1.22 and 1.25
obtained with the reference solution. for rifampicin, rifampicin quinone, rifampicin N-oxide,
Isonicotinyl hydrazone and 3-formylrifamycin SV respectively.
B. In the Assay for ethambutol hydrochloride, the principal
peak in the chromatogram obtained with the test solution In the chromatogram obtained with the test solution the area
corresponds to the peak in the chromatogram obtained with of any peak due to rifampicin quinone should not be more
the reference solution. than 4 times the area of the principal peak in the chromatogram
obtained with the reference solution (4 per cent), the area of
Tests any peak due to rifampicin N-oxide should not be more than
1.5 times the area of the principal peak in the chromatogram
Related substances. Determine by liquid chromatography
obtained with the reference solution (1.5 per cent), the area of
(2.4.14).
any peak due to 3-formylrifamycin SV Isonicotinyl hydrazone
Solvent mixture. A mixture of 10 volumes of a 21.01 per cent should not be more than 5 times the area of the principal peak
w/v solution of citric acid, 23 volumes of a 13.61 per cent in the chromatogram obtained with the reference solution
w/v solution of potassium dihydrogen phosphate, 77 volumes ( 5 per cent ) and the area of any peak due to 3-formylrifamycin
of a 17.42 per cent w/v solution of dipotassium hydrogen SV should not be more than the area of the principal peak in
phosphate, 640 volumes of water and 250 volumes of the chromatogram obtained with the reference solution (1 per
acetonitrile. cent ). In the chromatogram obtained with the test solution
Test solution. Weigh accurately a quantity of powdered tablets the area of any unknown peak should not be more than 1.5
containing 200 mg of Rifampicin, dissolve in 100 ml of times the area of the principal peak in the chromatogram
acetonitrile and filter. Dilute 5 ml of this solution to 50 ml with obtained with the reference solution (1.5 per cent).
the solvent mixture. Dissolution (2.5.2).
Reference solution (a). Dissolve 20 mg of rifampicin RS in Apparatus. No 1
100 ml of acetonitrile. Dilute 1 ml of this solution to a 100 ml
Medium. 900 ml of sodium phosphate buffer pH 6.8 prepared
with the solvent mixture.
by dissolving 7 g of dibasic sodium phosphate anhydrous in
Reference solution (b). A solution containing 0.01 per cent 5000 ml of water and adjusting to pH 6.8 with phosphoric
w/v each of rifampicin RS and rifampicin quinone RS in acid.

1056
IP 2007 RITONAVIR

Speed and time. 100 rpm and 45 minutes. efficiencies for rifampicin, isoniazid and pyrazinamide are not
Withdraw a suitable volume of the medium and filter through less than 3000, 5000 and 25000 theoretical plates respectively.
a membrane filter disc with an average pore diameter not greater Inject alternately the test solution and the reference solution.
than 0.8 µm. Reject the first few ml of the filtrate. Determine the The relative retention time for rifampicin is 1.8, for isoniazid,
amounts of rifampicin, C43H58N4O12, isoniazid, C6H7N3O, 0.7 and for pyrazinamide, 1.0.
pyrazinamide, C5H5N3O and ethambutol hydrochloride, Calculate the contents of C43H58N4O12, C6H7N3O and C5H5N3O
C10H24N2O2.2HCl by using the methods described in the Assay in the tablets.
for rifampicin, isoniazid, pyrazinamide and ethambutol
hydrochloride. For ethambutol hydrochloride — Determine by liquid
chromatography (2.4.14).
D. Not less than 75 per cent of the stated amounts of
C43H58N4O12, C6H7N3O, C5H5N3O and C10H24N2O2.2HCl. Test solution. Weigh accurately a quantity of the powdered
tablets containing about 60 mg of Ethambutol Hydrochloride
Other tests. Comply with the tests stated under Tablets. and dissolve in 100 ml of the solvent mixture.
Assay. For rifampicin, isoniazid and pyrazinamide — Reference solution. A 0.06 per cent w/v solution of ethambutol
Determine by liquid chromatography (2.4.14). hydrochloride RS in the solvent mixture.
Test solution. Weigh and powder 20 tablets. Weigh accurately
Chromatographic system
a quantity of the powdered tablets containing about 40 mg of
– a stainless steel column 15 cm x 4.6 mm, packed with
Isoniazid, dissolve in 100 ml of methanol, dilute to 500 ml with
nitrile groups chemically bonded to porous silica
the solvent mixture, mix and filter.
particles (5 µm),
Reference solution. A solution containing 0.08 per cent w/v of – mobile phase: a mixture of 50 volumes of acetonitrile,
rifampicin RS, 0.04 per cent w/v of isoniazid RS and 0.2 per and 50 volumes of buffer solution pH 7.0 (diluent)
cent w/v of pyrazinamide RS in methanol. Dilute 10.0 ml of prepared by dissolving 1 ml of triethylamine in 1000 ml
this solution to 50.0 ml with the solvent mixture. of water and adjusting the pH to 7.0 with dilute
Chromatographic system phosphoric acid,
– a stainless steel column 25 cm x 4.6 mm, packed with – flow rate. 1 ml per minute,
octadecylsilane bonded to porous silica (5 µm), – spectrophotometer set at 200 nm,
– column. temperature 30º, – a 50 µl loop injector.
– mobile phase: A. a mixture of 96 volumes of buffer Inject the reference solution. The test is not valid unless the
solution pH 6.8 prepared by dissolving 1.4 g of disodium relative standard deviation for replicate injections is not more
hydrogen orthophosphate anhydrous in 1000 ml of than 2.0 per cent, the tailing factor is not more than 3.0 and the
water, adjusting to pH 6.8 ± 0.05 with dilute phosphoroic column efficiency determined from ethambutol hydrochloride
acid and 4 volumes of acetonitrile, peak is not less than 1500 theoretical plates.
B. a mixture of 45 volumes of the buffer
solution and 55 volumes of acetonitrile, Calculate the content of C10H24N2O2.2HCl in the tablets.
– flow rate. 1.5 ml per minute, Storage. Store protected from moisture.
– a linear gradient programme using the conditions given
below,
– spectrophotometer set at 238 nm,
– a 20 µl loop injector.
Ritonavir
Time Solution A Solution B
CH3
(in min.) (per cent v/v) (per cent v/v)
H3C
0 100 0 S
5 100 0 H3C CH3
N
6 0 100 O H O
15 0 100 N
16 100 0 N N N O
22 100 0 CH3 H O OH H
S
Inject the reference solution. The test is not valid unless the N
relative standard deviation for replicate injections is not more
than 2.0 per cent, the tailing factor is not more than 2.0 for
rifampicin, isoniazid and pyrazinamide and the column C37H48N6O5S2 Mol. Wt. 721.0

1057
RITONAVIR CAPSULES IP 2007

Ritonavir is (5S,8S,10S,11S)-10-hydroxy-2-methyl-5-(1- – flow rate. 1 ml per minute,


methylethyl)-1-[2-(1-methylethyl)-4-thiazolyl]-3,6-dioxo-8,11- – spectrophotometer set at 239 nm,
bis(phenylmethyl)-2,4,7,12-tetraazatridecan-13-oic acid 5- – a 20 µl loop injector.
thiazolyl methyl ester. Inject the reference solution. The test is not valid unless the
Ritonavir contains not less than 98.0 per cent and not more column efficiency determined from the ritonavir peak is not
than 102.0 per cent of C37H48N 6O5S2, calculated on the less than 1000 theoretical plates, the tailing factor is not more
anhydrous basis. than 2.0, and the relative standard deviation for replicate
injections is not more than 2.0 per cent.
Description. A white to off-white powder.
Inject alternately the test solution and the reference solution.
Identification
Calculate the content of C37H48N6O5S2.
A. Determine by infrared absorption spectrophotometry (2.4.6). Storage. Store protected from light.
Compare the spectrum with that obtained with ritonavir RS or
with the reference spectrum of ritonavir.
B. In the Assay, the principal peak in the chromatogram Ritonavir Capsules
obtained with the test solution corresponds to the peak due
to ritonavir in the chromatogram obtained with the reference Ritonavir Capsules contain not less than 90.0 per cent and not
solution. more than 110.0 per cent of the stated amount of ritonavir
C37H48N6O5S2.
C. Melting point. 119º to 123º (2.4.21).
Identification
Tests
A. In the test for Assay, the principal spot in the chromatogram
Specific optical rotation (2.4.22). +7.0° to +10.5°, determined obtained with the test solution corresponds to that in the
in a 0.2 per cent w/v solution in methanol. chromatogram obtained with the reference solution.
Related substances. Determine by liquid chromatography B. When examined in the range 200 nm to 350 nm (2.4.7), a
(2.4.14), as described in the Assay and calculate the percentage 0.001 per cent w/v solution in methanol shows absorption
of each impurity peak in the chromatogram obtained with the maxima at the same wavelengths as the reference solution.
test solution. Not more than 0.5 per cent of any individual
impurity and not more than 1.0 per cent of total impurities is Tests
found.
Dissolution (2.5.2).
Heavy metals (2.3.13). 1.0 g complies with the limit test for Apparatus. No 1
heavy metals, Method B (20 ppm).
Medium. 900 ml of 0.1 M hydrochloric acid with 25 mM
Sulphated ash (2.3.18). Not more than 0.1 per cent polyoxyethylene 10 lauryl ether prepared by dissolving
Water (2.3.43). Not more than 0.5 per cent, determined on 2.0 g. 15.65 g of polyoxyethylene 10 lauryl ether in 950 ml of water.
Add 8.5 ml of hydrochloric acid and dilute to 1000 ml with
Assay. Determine by liquid chromatography (2.4.14). water.
Test solution. Dissolve 25.0 mg of the substance under Speed and time. 50 rpm and 90 minutes.
examination in sufficient of a mixture of 45 volumes of Withdraw a suitable volume of the medium and filter.
acetonitrile and 55 volumes of water to produce 100.0 ml.
Determine by liquid chromatography (2.4.14).
Reference solution. A 0.025 per cent w/v solution of ritonavir
RS in a mixture of 45 volumes of acetonitrile and 55 volumes Test solution. Use the filtrate and if necessary, dilute with the
of water. dissolution medium.

Chromatographic system Reference solution. A 0.1 per cent w/v solution of ritonavir
– a stainless steel column 15 cm x 4.6 mm, packed with RS in methanol. Dilute 5 ml of the solution to 50 ml with the
octadecylsilane bonded to porous silica (5 µm), dissolution medium.
– mobile phase: a mixture of 45 volumes of acetonitrile Use the chromatographic system described under Assay.
and 55 volumes of a buffer solution prepared by
Inject the test solution and the reference solution.
dissolving 3.4 g of sodium acetate and 0.94 g of sodium
hexanesulphonate in 1000 ml of water and adjusting D. Not less than 70 per cent of the stated amount of
the pH to 4.0 with hydrochloric acid, C37H48N6O5S2.

1058
IP 2007 RITONAVIR TABLETS

Related substances. Determine by liquid chromatography Assay. Determine by liquid chromatography (2.4.14).
(2.4.14). Test solution. Weigh accurately a quantity of the mixed
Solvent mixture. 40 volumes of 0.03M monobasic potassium contents of 20 capsules containing about 25 mg of Ritonavir,
phosphate buffer and 60 volumes of acetonitrile. disperse in 100.0 ml of methanol and filter.
Test solution. Weigh accurately a quantity of the mixed Reference solution. A 0.025 per cent w/v solution of ritonavir
contents of 20 capsules containing about 25 mg of Ritonavir, RS in methanol.
dissolve in 50 ml of the solvent mixture and filter. Chromatographic system
Reference solution (a). A 0.05 per cent w/v solution of – a stainless steel column 5 cm x 4.6 mm, packed with
ritonavir RS in the solvent mixture. octadecylsilane bonded to porous silica (3.5 µm),
– column temperature 45º,
Reference solution (b). Dilute 1 ml of the solution to 100 ml
– mobile phase: a mixture of 55 volumes of a buffer solution
with the solvent mixture.
prepared by dissolving 3.4 g of sodium acetate
Chromatographic system trihydrate and 0.94 g of sodium 1-hexanesulphonate in
– a stainless steel column 15 cm x 4.6 mm, packed with 1000 ml of water and adjusting the pH to 4.0 with
butyl silane chemically bonded to porous silica (3 µm) hydrochloric acid, and 45 volumes of acetonitrile,
(such as ACE C4), – flow rate. 2.5 ml per minute,
– mobile phase: A. a mixture of 69 volumes of 0.03 M – spectrophotometer set at 240 nm,
monobasic potassium phosphate buffer prepared by – a 10 µl loop injector.
dissolving 4.1 g of monobasic potassium phosphate Inject the reference solution. Run the chromatogram 1.5 times
in 1000 ml of water, 18 volumes of acetonitrile, 8 volumes the retention time (about 3 minutes)of the principal peak The
of tetrahydrofuran and 5 volumes of n-butanol and test is not valid unless the tailing factor is not more than 2.0,
filtering, the column efficiency in not less than 2000 theoretical plates
B. a mixture of 40 volumes of 0.03 M and the relative standard deviation for replicate injections is
monobasic potassium phosphate buffer, 47 volumes of not more than 2.0 per cent.
acetonitrile, 8 volumes of tetrahydrofuran and 5 volumes
of n-butanol, Inject the test solution and the reference solution.
– flow rate. 1 ml per minute, Calculate the content of C37H48N6O5S2 in the capsules.
– a linear gradient programme using the conditions given
below, Storage. Store protected from light in a refrigerator (2º to 8º).
– spectrophotometer set at 240 nm,
– a 50 µl loop injector.
Time mobile phase A mobile phase B Ritonavir Tablets
(in min.) (per cent v/v) (per cent v/v)
0 100 0 Ritonavir Tablets contain not less than 90.0 per cent and not
more than 110.0 per cent of the stated amount of ritonavir,
60 100 0 C37H48N6O5S2.
120 0 100
155 100 0 Identification
Inject the reference solution (a). The test is not valid unless In the Assay, the principal peak in the chromatogram obtained
the tailing factor is not more than 2.0 and the column efficiency with the test solution corresponds to the principal peak in the
in not less than 5000 theoretical plates. chromatogram obtained with the reference solution.
Inject the test solution and reference solution (b). In the
Tests
chromatogram obtained with the test solution, the area of any
secondary peak is not more than 2.5 times the area of the peak Dissolution (2.5.2)
in the chromatogram obtained with the reference solution (b)
Apparatus. No 1
(2.5 per cent) and the sum of areas of all the secondary peaks
is not more than 5 times the area of the peak in the Medium. 900 ml of 0.1 M hydrochloric acid.
chromatogram obtained with the reference solution (b) Speed and time. 100 rpm and 60 minutes.
(5.0 per cent). Withdraw a suitable volume of the medium and filter.
Other tests. Comply with the tests stated under Capsules. Determine by liquid chromatography (2.4.14)

1059
ROSIGLITAZONE MALEATE IP 2007

Test solution. The filtrate obtained as given above. Dilute the Inject reference solution (a). The test is not valid unless the
filtrate if necessary, with the dissolution medium. column efficiency is not less than 2000 theoretical plates and
Reference solution. A 0.1 per cent w/v solution of ritonavir the tailing factor is not more than 2.0.
RS in methanol. Dilute 5 ml of the solution to 50 ml with the Inject the test solution and reference solution (b). In the
dissolution medium. chromatogram obtained with the test solution, the area of any
Use the chromatographic system described under Assay. secondary peak is not more than 2.5 times the area of the peak
in the chromatogram obtained with the reference solution (b)
Inject the reference solution. The test is not valid unless the (2.5 per cent) and the sum of areas of all the secondary peaks
tailing factor not more than 2.0 and the relative standard is not more than 5 times the area of the peak in the
deviation of replicate injections is not more than 2.0 per cent. chromatogram obtained with the reference solution (b)
Inject the test solution and the reference solution. (5.0 per cent).
Calculate the content of C37H48N6O5S2. Other tests. Comply with the tests stated under Tablets.
D. Not less than 75 per cent of the stated amount of Assay. Determine by liquid chromatography (2.4.14)
C37H48N6O5S2. Test solution. Weigh accurately a quantity of the powdered
Related substances. Determine by liquid chromatography tablets containing 25 mg of Ritonavir and disperse in 100.0 ml
(2.4.14). of methanol.
Solvent mixture. A mixture of 40 volumes of 0.03 M monobasic Reference solution. A 0.025 per cent w/v solution of ritonavir
potassium phosphate and 60 volumes of acetonitrile. RS in methanol.
Test solution: Shake a quantity of the powdered tablets Chromatographic system
containing 25 mg of Ritonavir with 50 ml of the solvent mixture. – a stainless steel column 5 cm x 4.6 mm, packed with
phenyl stationary phase bonded to porous silica (3µm),
Reference solution (a). A 0.05 per cent w/v solution of – column temperature 45º,
ritonavir RS in the solvent mixture. – mobile phase: a mixture of 55 volumes of a buffer solution
Reference solution (b). Dilute 1 ml of reference solution (a) to prepared by dissolving 3.4 g of sodium acetate
100 ml with the solvent mixture. trihydrate and 0.94 g of sodium 1-hexane sulphonate
Chromatographic system to 1000 ml with water and adjusting the pH to 4.0 with
– a stainless steel column 15 cm x 4.6 mm, packed with dilute hydrochloric acid, and 45 volumes of
silica gel consisting of porous spherical particles with acetonitrile.
chemically bonded with nitrile group (3µm) (such as – flow rate. 2.5 ml per minute,
YMC C4), – spectrophotometer set at 239 nm,
– mobile phase: A. a mixture of 69 volumes of 0.03 M – a 10 µl loop injector.
monobasic potassium phosphate solution, 18 volumes Inject the reference solution. The test is not valid unless the
of acetonitrile, 8 volumes of tetrahydrofuran and relative standard deviation for replicate injections is not more
5 volumes of n- butanol, than 2.0 per cent.
B. a mixture of 40 volumes of 0.03 M Inject the test solution and the reference solution.
monobasic potassium phosphate solution, 47 volumes
Calculate the content of C37H48N6O5S2 in the tablets.
of acetonitrile, 8 volumes of tetrahydrofuran and 5
volumes of n-butanol, Storage. Store protected from moisture, at a temperature not
– flow rate.1 ml per minute, exceeding 30º.
– a linesr gradient programme using the conditions given
below,
– spectrophotometer set at 240 nm, Rosiglitazone Maleate
– a 100 µl loop injector.
S COOH
Time Mobile phase A Mobile phase B CH3 O ,
(in min.) (per cent v/v) (per cent v/v) N NH
O O COOH
0 100 0
N
60 100 0
120 0 100 C18H19N3O3S,C4H4O4 Mol. Wt. 473.5
121 100 0 Rosiglitazone Maleate is (±)-5-{p-[2-( Methyl-2-pyridylamino)
155 100 0 ethoxy]benzyl}-2,4-thiazolidinedione maleate (1:1).

1060
IP 2007 ROSIGLITAZONE TABLETS

Rosiglitazone Maleate contains not less than 98.0 per cent to 3.0 with dilute phosphoric acid, 25 volumes of
and not more than 102.0 per cent of C18H19N3O3S,C4H4O4, acetonitrile and 10 volumes of methanol,
calculated on the anhydrous basis. – flow rate. 1 ml per minute,
Description. A white to off-white crystalline powder. – spectrophotometer set at 235 nm,
– a 10 µl loop injector.
Identification Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
Determine by infrared absorption spectrophotometry (2.4.6). than 2.0 per cent.
Compare the spectrum with that obtained with rosiglitazone
maleate RS. Inject the test solution and the reference solution.
Calculate the content of C18H19N3O3S,C4H4O4.
Tests
Storage. Store protected from light.
Related substances. Determine by liquid chromatography
(2.4.14).
Test solution. Dissolve 25 mg of the substance under
examination in 50 ml of mobile phase. Rosiglitazone Tablets
Reference solution (a). A 0.05 per cent w/v solution of Rosiglitazone Maleate Tablets
rosiglitazone maleate RS in the mobile phase. Rosiglitazone Tablets contain Rosiglitazone Maleate.
Reference solution (b). Dilute 1 ml of reference solution (a) to Rosiglitazone Tablets contains not less than 90.0 per cent and
100 ml with mobile phase. not more than 110.0 per cent of the stated amount of
Chromatographic system as described under Assay. rosiglitazone, C18H19N3O3S.
Inject reference solution (a). The test is not valid unless the Identification
tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates. In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the peak in the
Inject the test solution and reference solution (b). In the
chromatogram obtained with the reference solution.
chromatogram obtained with the test solution, the area of any
secondary peak is not more than 0.5 times the area of the peak Tests
in the chromatogram obtained with reference solution (b)
(0.5 per cent) and the sum of areas of all the secondary peaks Dissolution (2.5.2).
is not more than the area of the peak in the chromatogram Apparatus. No 1
obtained with the reference solution (b) (1.0 per cent).
Medium. 900 ml of 0.1 M hydrochloric acid.
Heavy metals (2.3.13). 1 g complies with the limit test for heavy
metals, Method B (20 ppm). Speed and time. 75 rpm for 30 minutes.

Sulphated ash (2.3.18). Not more than 0.1 per cent. Withdraw a suitable volume of the medium and filter promptly
through a membrane filter disc having an average pore diameter
Water (2.3.43). Not more than 0.5 per cent, determined on not greater than 1.0 µm, rejecting the first 1 ml of the filtrate.
1.0 g. Dilute the filtrate, if necessary, with the same solvent. Measure
Assay. Determine by liquid chromatography (2.4.14). the absorbance of the resulting solution at the maximum at
about 318 nm (2.4.7). Similarly measure the absorbance of a
Test solution. Dissolve 20 mg of the substance under
standard solution of known concentration of rosiglitazone
examination in 100.0 ml of mobile phase.
maleate RS and calculate the content of C18H19N3O3S.
Reference solution. A 0.020 per cent w/v solution of
D. Not less than 70 per cent of the stated amount of
rosiglitazone maleate RS in mobile phase.
C18H19N3O3S.
Chromatographic system
Related substances. Determine by liquid chromatography
– a stainless steel column 25 cm x 4.6 mm packed with
(2.4.14).
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 65 volumes of buffer solution Test solution. Weigh accurately a quantity of powdered tablets
prepared by dissolving 1.36 g of potassium dihydrogen containing 200 mg of Rosiglitazone, disperse in 100.0 ml of
orthophosphate in 1000 ml of water and adjust the pH mobile phase. Centrifuge and use clear supernatant liquid.

1061
ROSUVASTATIN CALCIUM IP 2007

Reference solution (a). A 0.2 per cent w/v solution of Rosuvastatin Calcium
rosiglitazone maleate RS in the mobile phase.
Reference solution (b). Dilute 1 ml of reference solution (a) to
100 ml with mobile phase. F
Chromatographic system as described under Assay.
Inject reference solution (a). The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in OH OH O
not less than 2000 theoretical plates.
Ca++ N O
Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any H3C CH3
N N
secondary peak is not more than the area of the peak in the
SO2CH3 CH3
chromatogram obtained with reference solution (b) (1.0 per 2
cent) and the sum of areas of all the secondary peaks is not
more than twice the area of the peak in the chromatogram C22H27FN3O6S.Ca Mol. Wt. 1001.1
obtained with the reference solution (b) (2.0 per cent).
Rosuvastatin Calcium is (E)-(3R,5S)-7-{4-(4-fluorophenyl)-6-
Uniformity of content. Comply with the tests stated under isopropyl-2-{methyl(methylsulphonyl amino)]pyrimidin-5-
Tablets. yl}-3,5-dihydroxyhepten-6-oic acid calcium.
Disperse 1 tablet in 0.1 M hydrochloric acid to produce Rosuvastatin Calcium contains not less than 98.0 per cent
0.004 per cent w/v solution. Measure the absorbance of the and not more than 102.0 per cent of C22H27FN3O6S.Ca.
resulting solution at the maximum at about 318 nm (2.4.7). calculated on the dried basis.
Calculate the content of C18H19N3O3S from the absorbance
obtained from same concentration of rosiglitazone maleate Description. An off- white to creamish white crystalline
RS in the same medium. powder.
Other tests. Comply with the tests stated under Capsules. Identification
Assay. Determine by liquid chromatography (2.4.14). A. Determine by infrared absorption spectrophotometry (2.4.6).
Test solution. Weigh and powder 20 tablets. Weigh accurately Compare the spectrum with that obtained with rosuvastatin
a quantity of powdered tablets containing 20 mg of calcium RS.
Rosiglitazone, disperse in 100.0 ml of mobile phase. Centrifuge
B. Dissolve 20 mg in 25 ml of methanol, add 2 drops of methyl
and use clear supernatant liquid.
red indicator neutralise with 6 M ammonium hydroxide. Add
Reference solution. A 0.020 per cent w/v solution of 3 M hydrochloric acid, dropwise until the solution is acidic
rosiglitazone maleate RS in mobile phase. to the indicator. Add ammonium oxalate, a white precipitate
Chromatographic system is obtained.
– a stainless steel column 25 cm x 4.6 mm packed with
octadecysilane bonded to porous silica (5 µm),
Tests
– mobile phase: a mixture of 65 volumes of 0.01 M Related substances. Determine by liquid chromatography
potassium hydrogen phosphate adjusted to pH 3.0 with (2.4.14).
orthophosphoric acid, 25 volumes of acetonitrile and
Test solution. Dissolve 50 mg of the substance under
10 volumes of methanol,
examination in 100 ml of the mobile phase.
– flow rate. 1 ml per minute,
– spectrophotometer set at 235 nm, Reference solution (a). A 0.05 per cent w/v solution of
– a 10 µl loop injector. rosuvastatin calcium RS in the mobile phase.
Inject the reference solution. The test is not valid unless the Reference solution (b). Dilute 1 ml of reference solution (a) to
tailing factor is not more than 2.0, the column efficiency in not 100 ml with mobile phase.
less than 4000 theoretical plates and the relative standard Chromatographic system
deviation for replicate injections is not more than 2.0 per cent. – a stainless steel column 25 cm x 4.6 mm packed with
Inject the test solution and the reference solution. octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 50 volumes of 0.2 per cent
Calculate the content of C18H19N3O3S.
w/v acetic acid in water, 25 volumes of acetonitrile
Storage. Store protected from light and moisture. and 25 volumes of methanol,

1062
IP 2007 ROSUVASTATIN TABLETS

– flow rate. 1 ml per minute, Identification


– spectrophotometer set at 248 nm,
– a 20 µl loop injector. In the Assay, the principal peak in the chromatogram obtained
with the test solution corresponds to the peak in the
Inject reference solution (a). The test is not valid unless the chromatogram obtained with the reference solution.
tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates. Tests
Inject the test solution and reference solution (b). In the Dissolution (2.5.2).
chromatogram obtained with the test solution, the area of any Apparatus. No 1
secondary peak is not more than 0.5 times the area of the peak Medium. 900 ml of phosphate buffer pH 6.8,
in the chromatogram obtained with reference solution (b) (0.5
Speed and time. 50 rpm for 30 minutes.
per cent) and the sum of areas of all the secondary peaks is
not more than twice the area of the peak in the chromatogram Withdraw a suitable volume of the medium and filter.
obtained with the reference solution (b) (2.0 per cent). Determine by liquid chromatography (2.4.14).
Water (2.3.43). Not more than 8.0 per cent, determined on Test solution. The filtrate obtained as given above.
0.3 g.
Reference solution. A 0.05 per cent w/v solution of
Assay. Determine by liquid chromatography (2.4.14). rosuvastatin calcium RS in the mobile phase. Dilute 2 ml of
Test solution. Dissolve 50 mg of the substance under the solution to 100 ml with Dissolution medium.
examination in 100.0 ml of mobile phase. Dilute 5.0 ml of the Chromatographic system as described under Assay.
solution to 25.0 ml with mobile phase, mix and filter.
Calculate the content of C22H27FN3O6S.Ca.
Reference solution. A 0.05 per cent w/v of rosuvastatin D. Not less than 70 per cent of the stated amount of
calcium RS in mobile phase. Dilute 5.0 ml of the solution to C22H27FN3O6S.Ca.
25.0 ml with mobile phase.
Uniformity of content. Comply with the tests stated under
Chromatographic system Tablets.
– a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm), Determine by liquid chromatography (2.4.14), as described
– mobile phase: a mixture of 50 volumes of 0.2 per cent under Assay.
acetic acid in water, 25 volumes of acetonitrile and Test solution. Disperse one tablet in 100 ml of the mobile phase.
25 volumes of methanol, filter and degas. Dilute 5 ml of the solution to 10 ml with mobile phase and
– flow rate. 1 ml per minute, filter.
– spectrophotometer set at 248 nm,
Related substances. Determine by liquid chromatography
– a 20 µl loop injector.
(2.4.14).
Inject the reference solution. The test is not valid unless the
Test solution. Weigh and powder 20 tablets. Weigh accurately
relative standard deviation for replicate injections is not more
a quantity of powdered tablet containing 25 mg of
than 2.0 per cent.
Rosuvastatin, disperse in 50 ml of mobile phase and filter.
Inject the test solution and the reference solution. Reference solution (a). A 0.05 per cent w/v solution of
Calculate the content of C22H27FN3O6S.Ca. rosuvastatin calcium RS in the mobile phase.
Storage. Store protected from light and moisture. Reference solution (b). Dilute 1 ml of reference solution (a) to
100 ml with mobile phase.
Chromatographic system as described under Assay.
Inject reference solution (a). The test is not valid unless the
Rosuvastatin Tablets tailing factor is not more than 2.0 and the column efficiency in
not less than 2000 theoretical plates.
Rosuvastatin Calcium Tablets
Inject the test solution and reference solution (b). In the
Rosuvastatin Tablets contain Rosuvastatin Calcium. chromatogram obtained with the test solution, the area of any
Rosuvastatin Tablets contain not less than 90.0 per cent and secondary peak is not more than 1.5 times the area of the peak
not more than 110.0 per cent of the stated amount of in the chromatogram obtained with reference solution (b) (1.5
rosuvastatin calcium, C22H27FN3O6S.Ca. per cent) and the sum of areas of all the secondary peaks is

1063
ROXITHROMYCIN IP 2007

not more than 3.0 times the area of the peak in the chromatogram Roxithromycin is (3R,4S,5S,6R,7R,9R,11S,12R,13S,14R)-4-
obtained with the reference solution (b) (3.0 per cent). [(2,6-dideoxy-3-C-methyl-3-O-methyl-α-L-
Other tests. Comply with the tests stated under Tablets. ribohexopyranosyl)oxy]-14-ethyl-7,12,13-trihydroxy-10-[(E)-
[(2-methoxyethoxy)methoxy]imino]-3,5,7,9,11,13-hexamethyl-
Assay. Determine by liquid chromatography (2.4.14). 6-[[3,4,6-trideoxy-3-(dimethylamino)-β-D-xylo-
Test solution. Weigh and powder 20 tablets. Weigh accurately hexopyranosyl]oxy]oxacyclotetradecan-2-one;
a quantity of powdered tablet containing 25 mg of erythromycin 9-(E)-[O-[(2-methoxyethoxy)methyl]oxime].
Rosuvastatin, disperse in 100.0 ml of mobile phase. Dilute 5.0 Roxithromycin contains not less than 96.0 per cent and not
ml of the solution to 25.0 ml with mobile phase and filter. more than 102.0 per cent of C41H76N2O15, calculated on the
Reference solution. A 0.025 per cent w/v solution of anhydrous basis.
rosuvastatin calcium RS in mobile phase. Dilute 5.0 ml of the
Description. A white, crystalline powder.
solution to 25.0 ml with mobile phase.
Chromatographic system Identification
– a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm), A. Determine by infrared absorption spectrophotometry (2.4.6).
– column temperature 30°, Compare the spectrum with that obtained with roxithromycin
– mobile phase: a mixture of 585 volumes of a buffer RS.
solution prepared by dissolving 1.54 g of ammonium B. In the Assay, the principal peak in the chromatogram
acetate in 900 ml water, adjust pH to 4.0 with glacial obtained with the test solution corresponds to the peak in the
acetic acid and dilute to 1000 ml with water, 360 volumes chromatogram obtained with reference solution (a).
of acetonitrile and 50 volumes of tetrahydrofurone,
– flow rate. 1.5 ml per minute, Tests
– spectrophotometer set at 248 nm,
Appearance of solution. A 1.0 per cent w/v solution in methanol
– a 20 µl loop injector.
is clear (2.4.1) and colourless (2.4.1).
Inject the reference solution. The test is not valid unless the
column efficiency is not less than 2000 theoretical plates, the Specific optical rotation (2.4.22). – 93.0º to – 96.0º, determined
tailing factor is not more than 2.0 and the relative standard in a 1.0 per cent w/v solution in acetone.
deviation for replicate injections is not more than 2.0 per cent. Related substances. Determine by liquid chromatography
Inject the test solution and the reference solution. (2.4.14).
Calculate the content of C22H27FN3O6S.Ca. Solvent mixture. Mix 30 volumes of acetonitrile and 70
Storage. Store protected from light and moisture. volumes of a 4.8 per cent w/v solution of ammonium
dihydrogen phosphate and adjust the pH to 5.3 with dilute
Labelling. The label states the strength in terms of the sodium hydroxide solution.
equivalent amount of Rosuvastatin.
Test solution. Dissolve 50.0 mg of the substance under
examination in 25.0 ml of the solvent mixture.
Roxithromycin Reference solution (a). A 0.2 per cent w/v solution of
roxithromycin RS in the solvent mixture.
O O Reference solution (b). Dilute 1 ml of reference solution (a) to
N OCH3
CH3 100 ml with the solvent mixture.
H3C
OH CH3 Reference solution (c). Dissolve 2 mg of erythromycin 9-(E)-
HO
OH CH3 N CH3 [O-[[(2-methoxyethoxy)methoxy]methyl]oxime] (impurity
H 3C A) in 10 ml of reference solution (a). Further dilute 1 ml of this
H 3C HO CH3
O solution to 10 ml with reference solution (a).
O O
H 3C OCH3 Reference solution (d). Dilute 1.0 ml of toluene to 100 ml with
O CH3 acetonitrile. Dilute 0.2 ml of this solution to 200 ml with the
O
CH3 OH solvent mixture.
O Chromatographic system
CH3
– a stainless steel column 15 cm x 4.6 mm, packed with
C41H76N2O15 Mol. Wt. 837.0 spherical end-capped octadecylsilyl silica gel for

1064
IP 2007 ROXITHROMYCIN TABLETS

chromatography (5 µm), with a 10 nm pore size and a Sulphated ash (2.3.18). Not more than 0.1 per cent.
carbon loading of about 19 per cent, Water (2.3.43). Not more than 3.0 per cent, determined on
– column temperature 15º, 0.2 g.
– mobile phase: A. a mixture of 26 volumes of acetonitrile
and 74 volumes of a 5.97 per cent w/v solution of Assay. Determine by liquid chromatography (2.4.14).
ammonium dihydrogen phosphate, adjusted to pH 4.3 Solvent mixture. Mix 30 volumes of acetonitrile and 70
with dilute sodium hydroxide solution, volumes of a 4.8 per cent w/v solution of ammonium
B. a mixture of 30 volumes of water and dihydrogen phosphate and adjust the pH to 5.3 with dilute
70 volumes of acetonitrile, sodium hydroxide solution.
– flow rate.1.1 ml per minute, Test solution. Dissolve 50.0 mg of the substance under
– a linear gradient programme using the conditions given examination in 25.0 ml of the solvent mixture.
below,
– spectrophotometer set at 205 nm, Reference solution (a). A 0.2 per cent w/v solution of
roxithromycin RS in the solvent mixture.
– a 20 µl loop injector.
Time Mobile phase A Mobile phase B Reference solution (b). Dissolve 2 mg of erythromycin 9-(E)-
(in min.) (per cent v/v) (per cent v/v) [O-[[(2-methoxyethoxy)methoxy]methyl]oxime] (impurity
A) in 10.0 ml of reference solution (a). Further dilute 1.0 ml of
0 100 0
this solution to 10.0 ml with reference solution (a).
50 100 0
Chromatographic system
80 90 10 – a stainless steel column 15 cm x 4.6 mm, packed with
100 100 0 spherical end-capped octadecylsilyl silica gel for
Inject reference solution (c). Relative retention time between chromatography (5 µm), with a 10 nm pore size and a
roxithromycin and erythromycin 9-(E)-[O-[[(2-methoxyethoxy) carbon loading of about 19 per cent,
methoxy]methyl] oxime] (impurity A) is about 1.15. The test is – column temperature 15º,
not valid unless the peak-to-valley ratio Hp / Hv is not more – mobile phase: mix 307 volumes of acetonitrile and 693
than 2.0, where Hp = height above the baseline of the peak due volumes of a 4.91 per cent w/v solution of ammonium
to impurity A and Hv = height above the baseline of the lowest dihydrogen phosphate adjusted to pH 5.3 with dilute
point of the curve separating this peak from the peak due to sodium hydroxide solution,
roxithromycin. – flow rate.1.5 ml per minute,
– a linear gradient programme using the conditions given
Inject the test solution and reference solutions (b) and (d). In below,
the chromatogram obtained with the test solution the area of – spectrophotometer set at 205 nm,
the impurity A peak obtained immediately after the peak – a 20 µl loop injector.
obtained with roxithromycin is not more than the area of the
principal peak in the chromatogram obtained with reference Inject reference solution (b). The test is not valid unless the
solution (b) (1.0 per cent). The area of any other individual peak-to-valley ratio Hp / Hv is not less than 1.5
impurity peak is not more than 0.5 times the area of the principal Inject alternately the test solution and reference solution (a)
peak in the chromatogram obtained with reference solution
Calculate the content of C14H26N2O15.
(b) (0.5 per cent). The sum of the areas of all the peaks is not
more than 3 times the area of the principal peak in the Storage. Store protected from light and moisture.
chromatogram obtained with reference solution (b) (3.0 per
cent). Ignore any peak with an area 0.05 times the area of the
principal peak in the chromatogram obtained with reference
solution (b) (0.05 per cent). Ignore any peak due to toluene
Roxithromycin Tablets
identified using reference solution (d). Roxithromycin Tablets contain not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
Heavy metals (2.3.13). Dissolve 2.0 g in a mixture of 15 volumes
roxithromycin, C41H76N2O15.
of water and 85 volumes of acetone and dilute to 20 ml with
the same solvent mixture. 12 ml of this solution complies with Identification
the limit test for heavy metals, Method A (10 ppm). Use 1 ml of
lead standard solution (10 ppm lead) prepared by diluting In the Assay, the principal peak in the chromatogram obtained
lead standard solution (100 ppm lead) with a mixture of 15 with the test solution corresponds to the peak in the
volumes of water and 85 volumes of acetone. chromatogram obtained with the reference solution.

1065
ROXITHROMYCIN TABLETS IP 2007

Tests Chromatographic system


– a stainless steel column 15 cm x 4.6 mm, packed with
Dissolution (2.5.2). spherical end-capped octadecylsilyl silica gel (5 µm),
Apprartus No. 1 with a 10 nm pore size and a carbon loading of about 19
Medium. 900 ml of phosphate buffer pH 6.0. per cent,
Speed and time. 50 rpm and 45 minutes. – column temperature. 15º,
– mobile phase: mix 307 volumes of acetonitrile and 693
Withdraw a suitable volume of the medium and filter. volumes of a 4.91 per cent w/v solution of ammonium
Determine by liquid chromatography (2.4.14) as given under dihydrogen phosphate adjusted to pH 5.3 with dilute
the Assay using the following test solution. sodium hydroxide solution,
– flow rate. 1.5 ml per minute,
Test solution. The filtrate obtained as given above.
– spectrophotometer set at 205 nm,
Calculate the content of C41H76N2O15 in the medium. – a 20 µl loop injector.
D. Not less than 70 per cent of the stated amount of Inject reference solution (b). The relative retention time
C41H76N2O15. between roxithromycin and erythromycin -(E)-[O-[[(2-
Other tests. Comply with the tests stated under Tablets. methoxyethoxy)methoxy]methyl]oxime] (impurity A) is about
1.15. The test is not valid unless the peak-to-valley ratio Hp /
Assay. Determine by liquid chromatography (2.4.14). Hv is not more than 1.5, where Hp = height above the baseline
Solvent mixture. Mix 30 volumes of acetonitrile and 70 of the peak due to impurity A and Hv = height above the
volumes of a 4.8 per cent w/v solution of ammonium baseline of the lowest point of the curve separating this peak
dihydrogen phosphate and adjust the pH to 5.3 with dilute from the peak due to roxithromycin.
sodium hydroxide solution. Inject reference solution (a). The test is not valid unless the
relative standard deviation for replicate injections is not more
Test solution. Weigh and powder 20 tablets. Weigh accurately than 2.0 per cent.
a quantity of the powder containing about 0.2 g of
Roxithromycin, add 80 ml of the solvent mixture and mix. Dilute Inject alternately the test solution and reference solution (a).
to 100.0 ml with the solvent mixture and filter. Calculate the content of C41H76N2O15 in the tablets.
Reference solution (a). A 0.2 per cent w/v solution of Storage. Store protected from light and moisture.
roxithromycin RS in the solvent mixture.
Labelling. If the tablets are dispersible, the label states that
Reference solution (b). Dissolve 2 mg of erythromycin 9-(E)- the tablets should be dispersed in water immediately before
[O-[[(2-methoxyethoxy)methoxy]methyl]oxime] (impurity use.
A) in 10 ml of reference solution (a). Further dilute 1 ml of this
solution to 10 ml with reference solution (a).

1066
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

S
Saccharin ....
Saccharin Sodium ....
Salbutamol ....
Salbutamol Inhalation ....
Salbutamol Sulphate ....
Salbutamol Injection ....
Salbutamol Syrup ....
Salbutamol Tablets ....
Salicylic Acid ....
Salmeterol Xinafoate ....
Salmetrol and Fluticasone Propionate Inhalation ....
Salmetrol and Fluticasone Propionate powder for Inhalation ....
Saquinavir ....
Saquinavir Mesylate ....
Saquinavir Mesylate Tablets ....
Sechidazole ....
Sechidazole Tablets ....
Colloidal Silicon Dioxide ....
Silver Nitrate ....
Sodium Acetate ....
Sodium Alginate ....
Sodium Aminosalicylate ....
Sodium Aminosalicylate Tablets ....
Sodium Ascorbate ....
Sodium Aurothiomalate ....
Sodium Aurothiomalate Injection ....
Sodium Benzoate ....
Sodium Bicarbonate ....
Sodium Bicarbonate Injection ....
Sodium Carbonate ....

1055
MONOGRAPHS INDIAN PHARMACOPOEIA 2007

Sodium Chloride .................................................................................................................


Sodium Chloride And Dextrose Injection .............................................................................
Sodium Chloride And Fructose Injection ............................................................................
Compound Sodium Chloride And Dextrose Injection ..........................................................
Sodium Chloride Hypertonic Injection ................................................................................
Sodium Chloride Injection ..................................................................................................
Compound Sodium Chloride Injection ...............................................................................
Compound Sodium Chloride Solution ................................................................................
Sodium Chloride Irrigation Solution ......................................................................................
Sodium Citrate ..................................................................................................................
Sodium Cromoglycate .......................................................................................................
Sodium Cromoglycate Powder for Inhalation ....................................................................
Sodium Diatrizoate .............................................................................................................
Sodium Diatrizoate Injection ..............................................................................................
Sodium Dihydrogen Phosphate Dihydrate ...........................................................................
Sodium Fluoride .................................................................................................................
Sodium Formaldehyde Sulphoxylate ....................................................................................
Sodium Fusidate ...............................................................................................................
Sodium Fusidate Capsules ...................................................................................................
Sodium Hydroxide ..............................................................................................................
Sodium Lactate Injection ....................................................................................................
Compound Sodium Lactate And Dextrose Injection ............................................................
Half Strength Compound Sodium Lactate And Dextrose Injection ....................................
Modified Compound Sodium Lactate And Dextrose Injection ..............................................
Compound Sodium Lactate Injection ..................................................................................
Compound Sodium Lactate Solution For Irrigation ............................................................
Sodium Lauryl Sulphate .....................................................................................................
Sodium Metabisulphite ........................................................................................................
Sodium Methylparaben ......................................................................................................
Sodium Phosphate ...............................................................................................................
Sodium Propylparaben .....................................................................................................
Sodium Salicylate ...............................................................................................................
Sodium Starch Glycollate ....................................................................................................

1056
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

Sodium Stibogluconate ....


Sodium Stibogluconate Injection ....
Sodium Thiosulphate ....
Sodium Thiosulphate Injection ....
Sodium Valproate ....
Sodium Valproate Oral Solution ....
Sodium Valproate Tablets ....
Sorbic Acid ....
Sorbitol ....
Sorbitol Solution (70 Per Cent) (Crystallising) ....
Sorbitol Solution (70 Per Cent) (Non-Crystallising) ....
Spironolactone ....
Spironolactone Tablets ....
Stavudine ....
Stavudine Capsules ....
Stavudine Oral Solution ....
Stavudine And Lamivudine Tablets ....
Stearic Acid ....
Stearyl Alcohol ....
Stilboestrol ....
Stilboestrol Tablets ....
Prepared Storax ....
Streptokinase ....
Streptokinase Injection ....
Streptomycin Sulphate ....
Streptomycin Injection ....
Streptomycin Tablets ....
Succinylcholine Chloride ....
Succinylcholine Injection ....
Sucrose ....
Sulphacetamide Sodium ....
Sulphacetamide Eye Drops ....
Sulphadiazine ....

1057
MONOGRAPHS INDIAN PHARMACOPOEIA 2007

Sulphadiazine Tablets ....


Sulphadimethoxine ....
Sulphadimethoxine Tablets ....
Sulphadimidine ....
Sulphadimidine Sodium ....
Sulphadimidine Injection ....
Sulphadimidine Tablets ....
Sulphadoxine ....
Sulphafurazole ....
Sulphafurazole Tablets ....
Sulphalene ....
Sulphamethizole ....
Sulphamethoxazole ....
Sulphaphenazole ....
Sulphaphenazole Tablets ....
Sulphobromophthalein Sodium ....
Sulphobromophthalein Sodium Injection ....

1058
IP 2007 SACCHARIN

Saccharin Combine the lower layers, dry over anhydrous sodium


sulphate and filter. Wah the filter and the sodium sulphate
O with 10 ml of dichloromethane. Combine the solution and the
O washings and evaporate almost to dryness in a water-bath at
S
NH a temperature not exceeding 40º. With a small quantity of
dichloromethane transfer quantitatively the residue into a
suitable 10-ml tube, evaporate to dryness in a current of
O
nitrogen and dissolve the residue in 1.0 ml of the internal
C7H5NO3S Mol. Wt. 183.2 standard solution.
Saccharin is 1,2-benzisothiazol-3(2H)-one 1,1-dioxide. Blank solution. Evaporate 200 ml of dichloromethane to
dryness in a water-bath at a temperature not exceeding 40º.
Saccharin contains not less than 98.0 per cent and not more
Dissolve the residue in 1 ml of dichloromethane.
than 101.0 per cent of C7H5NO3S, calculated on the dried basis.
Reference solution. Dissolve 20 mg of o-toluenesulphonamide
Description. White crystals or a white, crystalline powder;
and 20 mg of p-toluene sulphonamide in dichloromethane
odourless or with a faint, aromatic odour.
and dilute to 100 ml with the same solvent. Dilute 5 ml of the
Identification solution to 50 ml with dichloromethane. Evaporate 5 ml of the
final solution to dryness in a current of nitrogen. Dissolve the
Test A may be omitted if tests B, C and D are carried out. Tests residue in 1 ml of the internal standard solution.
B, C and D may be omitted if test A is carried out. Chromatographic system
A. Determine by infrared absorption spectrophotometry (2.4.6). – a fused silica column 10 m x 0.53 mm packed with
Compare the spectrum with that obtained with saccharin RS. polymethylphenylsiloxane (film thickness 2 µm),
– temperature:
B. Mix 20 mg with 20 mg of resorcinol, add 0.5 ml of sulphuric
column.180°,
acid and heat over a small flame until a dark green colour is
inlet port and detector. 250°,
produced; allow to cool and add 10 ml of water and an excess
– flame ionization detector,
of 2 M sodium hydroxide; a fluorescent green liquid is
– flow rate. 10 ml per minute of nitrogen (carrier gas),
produced.
– split ratio. 1:2.
C. Dissolve 0.1 g in 5 ml of a 10 per cent w/v solution of
Inject 1 µl of each solution. The order of elution is
sodium hydroxide, evaporate to dryness and gently fuse the
o-toluenesulphonamide, p-toluenesulphonamide, caffeine.
residue over a small flame until ammonia is no longer evolved.
Allow to cool, dissolve in 20 ml of water, make the solution The test is not valid unless the resolution between the peaks
just acidic to litmus paper, filter and add 0.05 ml of ferric due to o-toluenesulphonamide and p-toluenesulphonamide
chloride solution; a violet colour is produced. in the chromatogram obtained with the reference solution is
not less than 1.5 and the chromatogram obtained with the
D. A saturated solution is acidic to litmus paper.
blank solution does not show any peak with the same retention
Tests times as the internal standard, o-toluenesulphonamide and
p-toluenesulphonamide.
Appearance of solution. Dissolve 5.0 g in 20 ml of a 20 per cent
In the chromatogram obtained with the test solution the ratio
w/v solution of sodium acetate and dilute to 25 ml with the
of the area of the peak due to o-toluenesulphonamide to that
same solution; the solution is clear (2.4.1), and colourless
of the internal standard is not greater than the corresponding
(2.4.1).
ratio in the chromatogram obtained with the rference solution
Related substances. Determine by gas chromatography (10 ppm).
(2.4.13).
In the chromatogram obtained with the test solution the ratio
Internal standard solution. Dissolve 25 mg of caffeine in of the area of the peak due to p-toluenesulphonamide to that
dichloromethane and dilute to 100 ml with the same solvent. of the internal standard is not greater than the corresponding
Test solution. Suspend 10.0 g of the substance under ratio in the chromatogram obtained with the reference solution
examination in 20 ml of water and dissolve using about 5 ml of (10 ppm).
strong sodium hydroxide solution. If necessary, adjust the Arsenic (2.3.10). Mix 5.0 g with 3.0 g of anhydrous sodium
pH of the solution to 7.8 with 1 M sodium hydroxide or 1 M carbonate, add 10 ml of bromine solution and mix thoroughly.
hydrochloric acid and dilute to 50 ml with water. Shake the Evaporate to dryness on a water-bath, gently ignite and add
solution with 4 quantities, each of 50 ml, of dichloromethane. to the cooled residue a mixture of 16 ml of brominated

1059
SACCHARIN SODIUM IP 2007

hydrochloric acid AsT and 5 ml of bromine solution. Add A. Determine by infrared absorption spectrophotometry (2.4.6).
40 ml of water and boil gently, adding sufficient bromine Compare the spectrum with that obtained with saccharin
solution from time to time to maintain a slight excess. Filter sodium RS.
and remove the excess of bromine with a sufficient quantity of B. Dissolve 0.1 g in 5 ml of a 10 per cent w/v solution of
stannous chloride solution AsT. The resulting solution sodium hydroxide, evaporate to dryness and gently fuse the
complies with the limit test for arsenic (2 ppm). residue over a small flame until ammonia is no longer evolved.
Heavy metals (2.3.13). 1.2 g complies with the limit test for Allow to cool, dissolve in 20 ml of water, make the solution
heavy metals, Method D (20 ppm). Use lead standard solution just acidic to litmus paper, filter and add 0.05 ml of ferric
(2 ppm Pb). chloride solution; a violet colour is produced.
Readily carbonisable substances. Dissolve 0.2 g in 5 ml of C. Mix 20 mg with 20 mg of resorcinol, add 0.5 ml of sulphuric
sulphuric acid (96 per cent w/w) and maintain at about 50º acid and heat over a small flame until a dark green colour is
for 10 minutes. The solution is not more intensely coloured produced; allow to cool and add 10 ml of water and an excess
than reference solution BYS6 (2.4.1). of 2 M sodium hydroxide; a fluorescent green liquid is
produced.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined D. To 5 ml of a 10 per cent w/v solution add 3 ml of 2 M
on 1.0 g by drying in an oven at 105º for 4 hours. hydrochloric acid; a white precipitate is produced. The
precipitate, after washing with water and drying at 105º melts
Assay. Weigh accurately about 0.5 g, dissolve in 75 ml of hot at 226º to 230º (2.4.21).
water, cool quickly and titrate with 0.1 M sodium hydroxide
E. 0.5 ml of a 10 per cent w/v solution gives reaction B of
using phenolphthalein solution as indicator.
sodium salts (2.3.1).
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01832 g of
C7H5NO3S. Tests
Storage. Store protected from moisture. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Acidity or alkalinity. Dissolve 5.0 g in sufficient carbon
dioxide-free water to produce 50 ml (solution A). To 10 ml of
Saccharin Sodium solution A add 10 ml of 0.005 M sulphuric acid, heat to boiling,
Soluble Saccharin cool and add 0.1 ml of phenolphthalein solution. Not less
than 4.5 ml and not more than 5.5 ml of 0.01 M sodium
O hydroxide is required to change the colour of the solution to
O pink.
S
N Na Related substances. Determine by gas chromatography
(2.4.13).
O Internal standard solution. Dissolve 25 mg of caffeine in
dichloromethane and dilute to 100 ml with the same solvent.
C7H4NNaO3S Mol. Wt. 205.2
Test solution. Suspend 10.0 g of the substance under
Saccharin Sodium is the sodium salt of 1,2-benzisothiazol- examination in 50 ml of water. If necessary, adjust the pH of
3(2H)-3-one 1,1-dioxide. the solution to 7.8 with 1 M sodium hydroxide or 1 M
Saccharin Sodium contains not less than 99.0 per cent and not hydrochloric acid and dilute to 50 ml with water. Shake the
more than 101.0 per cent of C7H4NNaO3S, calculated on the solution with 4 quantities, each of 50 ml, of dichloromethane.
anhydrous basis. Combine the lower layers, dry over anhydrous sodium
sulphate and filter. Wah the filter and the sodium sulphate
Description. A white, crystalline powder or colourless crystals; with 10 ml of dichloromethane. Combine the solution and the
efflorescent in dry air. washings and evaporate almost to dryness in a water-bath at
a temperature not exceeding 40º. With a small quantity of
Identification
dichloromethane transfer quantitatively the residue into a
Test A may be omitted if tests B, C, D and E are carried out. suitable 10-ml tube, evaporate to dryness in a current of
Tests B, C and D may be omitted if tests A and E are carried nitrogen and dissolve the residue in 1.0 ml of the internal
out. standard solution.

1060
IP 2007 SALBUTAMOL

Blank solution. Evaporate 200 ml of dichloromethane to for 10 minutes. The solution is not more intensely coloured
dryness in a water-bath at a temperature not exceeding 40º. than reference solution BS6 (2.4.1).
Dissolve the residue in 1 ml of dichloromethane. Heavy metals (2.3.13). 12 ml of solution A complies with the
Reference solution. Dissolve 20 mg of o-toluenesulphonamide limit test for heavy metals, Method D (20 ppm). Use lead
and 20 mg of p-toluene sulphonamide in dichloromethane standard solution (2 ppm Pb).
and dilute to 100 ml with the same solvent. Dilute 5 ml of this Water (2.3.43). Not more than 15.0 per cent, determined on
solution to 50 ml with dichloromethane. Evaporate 5 ml of the 0.2 g.
final solution to dryness in a current of nitrogen. Dissolve the
residue in 1 ml of the internal standard solution. Assay. Weigh accurately about 0.15 g, dissolve in 50 ml of
anhydrous glacial acetic acid, with slight heating if necessary.
Chromatographic system Titrate with 0.1 M perchloric acid, determining the end-point
– a fused silica column 10 m x 0.53 mm packed with potentiometrically (2.4.25). Carry out a blank titration.
polymethylphenylsiloxane (film thickness 2 µm),
– temperature: 1 ml of 0.1 M perchloric acid is equivalent to 0.02052 g of
column.180°, C7H4NNaO3S.
inlet port and detector. 250° Storage. Store protected from moisture.
flame ionization detector,
– flow rate. 10 ml per minute of nitrogen (carrier gas),
– split ratio. 1:2.
Salbutamol
Inject 1 µl of each solution. The order of elution is
o-toluenesulphonamide, p-toluenesulphonamide, caffeine. Albuterol
The test is not valid unless the resolution between the peaks OH
due to o-toluenesulphonamide and p-toluenesulphonamide H
in the chromatogram obtained with the reference solution is N CH3
not less than 1.5 and the chromatogram obtained with the CH3
H3 C
blank solution does not show any peak with the same retention HO
times as the internal standard, o-toluenesulphonamide and
p-toluenesulphonamide. OH

In the chromatogram obtained with the test solution the ratio C13H21NO3 Mol. Wt. 239.3
of the area of the peak due to o-toluenesulphonamide to that Salbutamol is (RS)-1-(4-hydroxy-3-hydroxymethylphenyl)-2-
of the internal standard is not greater than the corresponding (tert-butylamino)ethanol.
ratio in the chromatogram obtained with the reference solution
(10 ppm). Salbutamol contains not less than 98.0 per cent and not more
than 101.0 per cent of C13H21NO3, calculated on the dried basis.
In the chromatogram obtained with the test solution the ratio
of the area of the peak due to p-toluenesulphonamide to that Description. A white or almost white, crystalline powder.
of the internal standard is not greater than the corresponding Identification
ratio in the chromatogram obtained with the rference solution
(10 ppm). Test A may be omitted if tests B, C and D are carried out. Tests
B, C and D may be omitted if test A is carried out.
Arsenic (2.3.10). Mix 5.0 g with 3 g of anhydrous sodium
carbonate, add 10 ml of bromine solution and mix thoroughly. A. Determine by infrared absorption spectrophotometry (2.4.6),
Evaporate to dryness on a water-bath, gently ignite and add Compare the spectrum with that obtained with salbutamol RS
to the cooled residue a mixture of 16 ml of brominated or with the reference spectrum of salbutamol.
hydrochloric acid AsT and 5 ml of bromine solution. Add B. When examined in the range 230 nm to 360 nm (2.4.7), a
40 ml of water and boil gently, adding sufficient bromine 0.008 per cent w/v solution in 0.1 M hydrochloric acid shows
solution from time to time to maintain a slight excess. Filter an absorption maximum only at about 276 nm; absorbance at
and remove the excess of bromine with a sufficient quantity of about 276 nm, about 0.53 to 0.60.
stannous chloride solution AsT. The resulting solution
complies with the limit test for arsenic (2 ppm). C. In the test for Related substances, the principal spot in the
chromatogram obtained with reference solution (a)
Readily carbonisable substances. Dissolve 0.2 g in 5 ml of corresponds to that in the chromatogram obtained with
sulphuric acid (96 per cent w/w) and maintain at about 50º reference solution (b).

1061
SALBUTAMOL INHALATION IP 2007

D. Dissolve 10 mg in 50 ml of a 2 per cent w/v solution of Sulphated ash (2.3.18). Not more than 0.1 per cent.
borax, add 1 ml of a 3 per cent w/v solution of Loss on drying (2.4.19). Not more than 0.5 per cent, determined
4-aminophenazone, 10 ml of a 2 per cent w/v solution of on 1.0 g by drying in an oven at 105º.
potassium ferricyanide and 10 ml of chloroform, shake and
allow to separate; an orange-red colour is produced in the Assay. Weigh accurately about 0.2 g, dissolve in 30 ml of
chloroform layer. anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
acid, determining the end-point potentiometrically (2.4.25).
Tests Carry out a blank titration.
Appearance of solution. A 2.0 per cent w/v solution in methanol 1 ml of 0.1 M perchloric acid is equivalent to 0.02393 g of
is clear (2.4.1), and not more intensely coloured than reference C13H21NO3.
solution BYS5 (2.4.1). Storage. Store protected from light.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
Mobile phase. A mixture of 50 volumes of ethyl acetate,
30 volumes of 2-propanol, 16 volumes of water and 4 volumes Salbutamol Inhalation
of strong ammonia solution.
Salbutamol Inhalation Aerosol; Albuterol Inhalation
Test solution. Dissolve 0.2 g of the substance under Aerosol
examination in 10 ml of methanol.
Salbutamol Inhalation is a suspension of microfine Salbutamol
Reference solution (a). A 0.01 per cent w/v solution of the or Salbutamol Sulphate in a suitable liquid in a suitable
substance under examination in methanol. pressurised container. It may contain suitable pharmaceutical
Reference solution (b). A 0.01 per cent w/v solution of aids such as surfactants, stabilising agents etc.
salbutamol RS in methanol. Salbutamol Inhalation delivers not less than 80.0 per cent and
Apply to the plate 5 µl of each solution. After development, not more than 120.0 per cent of the stated amount of
dry the plate in air until the odour of the solvent is no longer salbutamol, C13H21NO3, per inhalation, by actuation of the valve.
detectable, place it for a few minutes in an atmosphere saturated
with diethylamine and spray with diazotised sulphanilic acid Identification
solution. Any secondary spot in the chromatogram obtained A. Discharge the container a sufficient number of times into a
with the test solution is not more intense than the spot in the mortar to obtain about 2 mg of salbutamol, grind the residue
chromatogram obtained with reference solution (a). thoroughly with 0.1 g of potassium bromide, add a further 0.2
Boron. To 50 mg add 5 ml of a solution containing 1.3 per cent g of potassium bromide and mix thoroughly.
w/v of anhydrous sodium carbonate and 1.7 per cent w/v of On the resultant dispersion determine by infrared absorption
potassium carbonate, evaporate to dryness on a water-bath spectrophotometry (2.4.6). Compare the spectrum with that
and dry at 120º. Ignite the residue rapidly until the organic obtained with salbutamol RS or with the reference spectrum
matter has been destroyed, allow to cool, add 0.5 ml of water of salbutamol.
and 3.0 ml of a freshly prepared 0.125 per cent w/v solution of
curcumin in glacial acetic acid. Evaporate to dryness and B. In the test for Related substances, the principal spot in the
allow to cool. Add 3 ml of a mixture prepared by adding 5 ml of chromatogram obtained with reference solution (a)
sulphuric acid, slowly and with stirring, to 5 ml of glacial corresponds to that in the chromatogram obtained with
acetic acid. Mix and allow to stand for 30 minutes. Add reference solution (b).
sufficient ethanol (95 per cent) to produce 100.0 ml, filter and
measure the absorbance of the filtrate at 555 nm (2.4.7). Prepare
Tests
a reference solution in the following manner. Dissolve 0.572 g Related substances. Determine by thin-layer chromatography
of boric acid in 1000.0 ml of water. Dilute 1.0 ml to 100.0 ml (2.4.17), coating the plate with silica gel G.
with water. To 2.5 ml of this solution add 5 ml of a solution
containing 1.3 per cent w/v of anhydrous sodium carbonate Mobile phase. A mixture of 50 volumes of ethyl acetate, 30
and 1.7 per cent w/v of potassium carbonate and treat this volumes of 2-propanol, 16 volumes of water and 4 volumes
mixture in the same manner as described above beginning at of strong ammonia solution.
the words “Evaporate to dryness…”. The absorbance of the Test solution. Discharge the inhaler a sufficient number of
solution prepared from the substance under examination is times into a small, dry beaker to obtain 10 mg of Salbutamol
not more than that of the reference solution (50 ppm). and dissolve the residue in 0.5 ml of methanol.

1062
IP 2007 SALBUTAMOL SULPHATE

Reference solution (a). Dilute 1.0 volume of test solution (a) When the active ingredient is Salbutamol Sulphate, the
to 200 volumes with methanol. quantity is stated in terms of the equivalent amount of
Reference solution (b). A 0.010 per cent w/v solution of salbutamol.
salbutamol RS in methanol.
Apply to the plate 5 µl of each solution. After development,
dry the plate in air until the odour of the solvent is no longer Salbutamol Sulphate
detectable, place it for a few minutes in an atmosphere saturated Albuterol Sulphate
with diethylamine and spray with diazotised sulphanilic acid
solution. Any secondary spot in the chromatogram obtained (C13H21NO3)2,H2SO4 Mol. Wt. 576.7
with the test solution is not more intense than the spot in the Salbutamol Sulphate is (RS)-1-(4-hydroxy-3-hydroxy-
chromatogram obtained with reference solution (a). Ignore methylphenyl)-2-(tert-butylamino)ethanol sulphate.
any spot with an Rf value higher than 0.85.
Salbutamol Sulphate contains not less than 98.0 per cent and
Other tests. Complies with the tests stated under Inhalation not more than 101.0 per cent of (C13H21NO3)2, H2SO4, calculated
Preparations (Pressurised metered-dose Preparations). on the dried basis.
Follow the procedure described under Assay wherever the Description. A white or almost white, crystalline powder.
amount of active substance is to be determined in any test.
Identification
Assay. Carry out the test for Content of active ingredient
delivered per actuation stated under Inhalation Preparations Test A may be omitted if tests B, C and D are carried out. Tests
(Pressurised metered-dose Preparations). B and C may be omitted if tests A and D are carried out.
Use 30 ml of ethanol for preparations containing salbutamol A. Determine by infrared absorption spectrophotometry (2.4.6),
and 30 ml of a mixture of equal volumes of ethanol and water Compare the spectrum with that obtained with salbutamol
for preparations containing salbutamol sulphate and dilute sulphate RS or with the reference spectrum of salbutamol
the final solution and washings to 200.0 ml with ethanol. Dilute sulphate.
a suitable volume of this solution with ethanol to produce a
B. When examined in the range 230 nm to 360 nm (2.4.7), a
solution containing 10 µg of salbutamol per ml. To 20 ml of the
0.008 per cent w/v solution in 0.1 M hydrochloric acid shows
solution in a separating funnel add 180 ml of water and in the
an absorption maximum at about 276 nm; absorbance at about
following order, 4 ml of N,N-dimethyl-4-phenylenediamine
276 nm, 0.44 to 0.51.
sulphate solution and 4 ml of a freshly prepared 8 per cent w/
v solution of potassium ferricyanide. Mix, allow to stand for C. In the test for Related substances, the principal spot in the
15 minutes in subdued light and extract with two quantities, chromatogram obtained with reference solution (a)
each of 10 ml, of chloroform. Filter the extracts through a plug corresponds to that in the chromatogram obtained with
of cotton wool, dilute to 25 ml with chloroform and measure reference solution (b).
the absorbance of the resulting solution at 605 nm (2.4.7). D. Dissolve 10 mg in 50 ml of a 2 per cent w/v solution of
Calculate the content of C13H21NO3 in the solution from the borax, add 1 ml of a 3 per cent w/v solution of
absorbance obtained by repeating the operation using a 4-aminophenazone, 10 ml of a 2 per cent w/v solution of
suitable quantity of a 0.001 per cent w/v solution of salbutamol potassium ferricyanide and 10 ml of chloroform, shake and
RS in ethanol. allow to separate; an orange-red colour is produced in the
Calculate the amount of C13H21NO3 delivered per actuation of chloroform layer.
the valve. E. Gives reaction A of sulphates (2.3.1).
Determine the content of active ingredient a second and third
time by repeating the procedure on the middle ten and on the Tests
last ten successive combined actuations of the valve. For Appearance of solution. A 1.0 per cent w/v solution in carbon
each of the three determinations the average content of dioxide-free water is clear (2.4.1), and not more intensely
C13H21NO3 delivered per actuation of the valve meets the coloured than reference solution BYS6 (2.4.1).
requirements.
Acidity or alkalinity. To 10 ml of a 1.0 per cent w/v solution in
Storage. Store protected from light and moisture at a carbon dioxide-free water add 0.15 ml of methyl red solution
temperature not exceeding 30º. and 0.2 ml of 0.01 M sodium hydroxide. The solution is yellow
Labelling. The label states whether the preparation contains and not more than 0.4 ml of 0.01 M hydrochloric acid is
Salbutamol or Salbutamol Sulphate. required to change the colour of the solution to red.

1063
SALBUTAMOL INJECTION IP 2007

Related substances. Determine by thin-layer chromatography Salbutamol Injection


(2.4.17), coating the plate with silica gel G.
Albuterol Sulphate Injection; Salbutamol Sulphate
Mobile phase. A mixture of 50 volumes of ethyl acetate,
30 volumes of 2-propanol, 16 volumes of water and 4 volumes Injection
of strong ammonia solution. Salbutamol Injection is a sterile solution of Salbutamol
Test solution. Dissolve 0.2 g of the substance under Sulphate in Water for Injections containing suitable stabilising
examination in 10 ml of water. agents.
Reference solution (a). A 0.01 per cent w/v solution of the Salbutamol Injection contains not less than 90.0 per cent and
substance under examination in water. not more than 110.0 per cent of the stated amount of
salbutamol, C13H21NO3.
Reference solution (b). A 0.01 per cent w/v of salbutamol
sulphate RS in water.
Identification
Apply to the plate 5 µl of each solution. After development,
dry the plate in air until the odour of the solvent is no longer A. Dilute a volume with sufficient 0.1 M hydrochloric acid to
detectable, place it for a few minutes in an atmosphere saturated produce a solution containing 0.008 per cent w/v of salbutamol.
with diethylamine and spray with diazotised sulphanilic acid When examined in the range 230nm to 360 nm (2.4.7), the
solution. Any secondary spot in the chromatogram obtained resulting solution shows an absorption maximum only at about
with the test solution is not more intense than the spot in the 276 nm.
chromatogram obtained with reference solution (a). B. Determine by thin-layer chromatography (2.4.17), coating
Boron. To 50 mg add 5 ml of a solution containing 1.3 per cent the plate with silica gel G.
w/v of anhydrous sodium carbonate and 1.7 per cent w/v of Mobile phase. A mixture of 50 volumes of ethyl acetate,
potassium carbonate, evaporate to dryness on a water-bath 30 volumes of 2-propanol, 16 volumes of water and 4 volumes
and dry at 120º. Ignite the residue rapidly until the organic of strong ammonia solution.
matter has been destroyed, allow to cool, add 0.5 ml of water
and 3.0 ml of a freshly prepared 0.125 per cent w/v solution of Test solution. Evaporate a suitable volume of the injection to
curcumin in glacial acetic acid. Evaporate to dryness and dryness using a rotary evaporator, wash the residue with four
allow to cool. Add 3 ml of a mixture prepared by adding 5 ml of quantities, each of 5 ml, of ethanol, filter, evaporate the filtrate
sulphuric acid, slowly and with stirring, to 5 ml of glacial to dryness and dissolve the residue in sufficient water to
acetic acid. Mix and allow to stand for 30 minutes. Add produce a solution containing the equivalent of 0.1 per cent
sufficient ethanol (95 per cent) to produce 100.0 ml, filter and w/v of salbutamol.
measure the absorbance of the filtrate at 555 nm (2.4.7). Prepare Reference solution. A 0.12 per cent w/v solution of salbutamol
a reference solution in the following manner. Dissolve 0.572 g sulphate RS in water.
of boric acid in 1000.0 ml of water. Dilute 1.0 ml to 100.0 ml
Apply to the plate 2 µl of each solution. After development,
with water. To 2.5 ml of this solution add 5 ml of a solution
dry the plate in air until the odour of solvent is no longer
containing 1.3 per cent w/v of anhydrous sodium carbonate
detectable, place for a few minutes in an atmosphere saturated
and 1.7 per cent w/v of potassium carbonate and treat this
with diethylamine and spray with diazotised nitroaniline
mixture in the same manner as described above beginning at
solution. The principal spot in the chromatogram obtained
the words “ Evaporate to dryness.....”.The absorbance of the
with the test solution corresponds to that in the chromatogram
solution prepared from the substance under examination is
obtained with the reference solution.
not more than that of the reference solution (50 ppm).
C. Dilute a volume containing 0.5 mg of salbutamol to 50 ml
Sulphated ash (2.3.18). Not more than 0.1 per cent.
with water, add 1 ml of dilute ammonia solution, 1 ml of a 3 per
Loss on drying (2.4.19). Not more than 0.5 per cent, determined cent w/v solution of 4-aminophenazone, 10 ml of a 2 per cent
on 1.0 g by drying in an oven at 105º. w/v solution of potassium ferricyanide and 10 ml of
Assay. Weigh accurately about 0.4 g, dissolve in 5 ml of chloroform. Shake and allow to separate; an orange-red colour
anhydrous formic acid, add 35 ml of anhydrous glacial acetic is produced in the chloroform layer.
acid. Titrate with 0.1 M perchloric acid, determining the end- D. A volume containing 1 mg of salbutamol gives the reactions
point potentiometrically (2.4.25). Carry out a blank titration. of sulphates (2.3.1).
1 ml of 0.1 M perchloric acid is equivalent to 0.05767 g of
(C13H21NO3)2, H2SO4. Tests
Storage. Store protected from light. pH (2.4.24). 3.4 to 5.0.

1064
IP 2007 SALBUTAMOL TABLETS

Other tests. Complies with the tests stated under Parenteral volumetric flask. Discard the ether extracts and dilute the
Preparations (Injections). aqueous solution with sufficient water to produce 250.0 ml.
Assay. Dilute a volume, accurately measured, containing about To 10.0 ml of this solution add sufficient water to produce
0.15 mg of salbutamol with sufficient water to produce 80 ml, 80 ml and add 4 ml of a 5 per cent w/v solution of sodium
add 4 ml of a 5 per cent w/v solution of sodium bicarbonate, bicarbonate, 4 ml of N,N-dimethyl-4-phenylenediamine
4 ml of N,N-dimethyl-4-phenylenediamine sulphate solution sulphate solution and 4 ml of a freshly prepared 8 per cent
and 4 ml of a freshly prepared 8 per cent w/v solution of w/v solution of potassium ferricyanide. Mix, allow to stand
potassium ferricyanide. Mix, allow to stand for 15 minutes, for 15 minutes, protected from light. Extract with two quantities,
protected from light. Extract with two quantities, each of 10 ml, each of 10 ml, of chloroform. Filter the extracts through a plug
of chloroform. Filter the extracts through a plug of cotton of cotton wool and dilute to 25.0 ml with chloroform. Measure
wool and dilute to 25.0 ml with chloroform. Measure the the absorbance of the resulting solution at 605 nm (2.4.7).
absorbance of the resulting solution at 605 nm (2.4.7). Calculate the content of C13H21NO3 from the absorbance
obtained by repeating the operation using 10.0 ml of a
Calculate the content of C13H21NO3 from the absorbance
0.0018 per cent w/v solution of salbutamol sulphate.
obtained by repeating the operation using 10.0 ml of a 0.0018
per cent w/v solution of salbutamol sulphate. Storage. Store protected from light.
Storage. Store protected from light, in single dose containers Labelling. The label states the strength in terms of the
in which the air has been displaced by nitrogen or other equivalent amount of salbutamol in a suitable dose-volume.
suitable inert gas.
Labelling. The label states the strength in terms of the
equivalent amount of salbutamol in a suitable dose-volume. Salbutamol Tablets
Albuterol Sulphate Tablets; Salbutamol Sulphate Tablets
Salbutamol Syrup Salbutamol Tablets contain not less than 90.0 per cent and not
more than 110.0 per cent of the stated amount of salbutamol,
Albuterol Sulphate Syrup; Salbutamol Sulphate Syrup C13H21NO3.
Salbutamol Syrup contains Salbutamol Sulphate equivalent
to not less than 90.0 per cent and not more than 110.0 per cent Identification
of the stated amount of salbutamol, C13H21NO3.
A. Carry out the method described under Related substances
Identification applying separately to the plate 2 µl of each of the following
solutions. For the test solution shake a quantity of the
A. To 5 ml add 50 ml of a 2 per cent w/v solution of borax, 1 ml powdered tablets containing 10 mg of salbutamol with 10 ml
of a 3 per cent w/v solution of 4-aminophenazone, 10 ml of a of methanol (80 per cent) and filter. The reference solution
2 per cent w/v solution of potassium ferricyanide and 10 ml of contains 0.12 per cent w/v of salbutamol sulphate RS. The
chloroform. Shake and allow to separate; an orange-red colour principal spot in the chromatogram obtained with the test
is produced in the chloroform layer. solution corresponds to that in the chromatogram obtained
B. To 5 ml add sufficient 1 M sodium hydroxide to make the with the reference solution.
solution alkaline, add 1 ml of alkaline borate buffer pH 9.2 B. Shake a quantity of the powdered tablets containing 8 mg
and 1 ml of a 0.04 per cent w/v solution of 2,6-dichloroquinone of salbutamol with 50 ml of a 2 per cent w/v solution of borax,
chlorimide in ethanol (95 per cent); a blue colour develops. add 1 ml of a 3 per cent w/v solution of 4-aminophenazone,
10 ml of a 2 per cent w/v solution of potassium ferricyanide
Tests and 10 ml of chloroform. Shake and allow to separate; an
pH (2.4.24). 3.4 to 4.5. orange-red colour is produced in the chloroform layer.
Other tests. Complies with the tests stated under Oral Liquids. C. Shake a quantity of the powdered tablets containing 4 mg
of salbutamol with 10 ml of water and filter; the filtrate gives
Assay. To an accurately measured volume containing about the reactions of sulphates (2.3.1).
4 mg of salbutamol add 25 ml of 0.05 M sulphuric acid and
extract with two quantities, each of 50 ml, of ether. Collect the Tests
aqueous layers into a 250-ml volumetric flask and combine the
ether extracts. Wash the combined ether extracts with 50 ml of Related substances. Determine by thin-layer chromatography
water and add the aqueous layer to the solution in the 250-ml (2.4.17), coating the plate with silica gel G.

1065
SALICYLIC ACID IP 2007

Mobile phase. A mixture of 50 volumes of ethyl acetate, Test solution. Shake 10 tablets or a sufficient number of tablets
30 volumes of 2-propanol, 16 volumes of water and 4 volumes containing 5.0 mg of salbutamol with about 60 ml of water for
of strong ammonia solution. 1 hour, add sufficient water to produce 250.0 ml, mix and
centrifuge 10 ml of the mixture and use the supernatant liquid.
Test solution. Shake a quantity of the powdered tablets
containing 10 mg of salbutamol with 1 ml of water for Reference solution (a). A 0.0024 per cent w/v of salbutamol
15 minutes, centrifuge and use the supernatant liquid. sulphate RS in water..
Reference solution. A 0.006 per cent w/v solution of Reference solution (b). A solution containing 0.0024 per cent
salbutamol sulphate RS in water. w/v of 2-tert-butylamino-1-(4-hydroxy-3-methylphenyl)
Apply to the plate 20 µl of each solution. After development, ethanol sulphate RS and 0.0024 per cent w/v of salbutamol
dry the plate in air until the odour of the solvent is no longer sulphate RS in methanol (10 per cent).
detectable, place it for a few minutes in an atmosphere saturated Follow the chromatographic procedure described under
with diethylamine and spray with diazotised sulphanilic acid Uniformity of content. The test is not valid unless the resolution
solution. Any secondary spot in the chromatogram obtained between the two principal peaks in the chromatogram obtained
with the test solution is not more intense than the spot in the with reference solution (b) is at least 1.5.
chromatogram obtained with the reference solution. Ignore
any pink spot near the line of application. Calculate the content of C13H21NO3 in the tablets.

Uniformity of content. Comply with the test stated under Storage. Store protected from light.
Tablets. Labelling. The label states the strength in terms of the
Determine by liquid chromatography (2.4.14). equivalent amount of salbutamol.

Test solution. Add 50 ml of water to one tablet, shake for


1 hour, add sufficient water to produce 100.0 ml, mix and
centrifuge. Dilute further with water, if necessary, to produce
a solution containing 0.002 per cent w/v of salbutamol. Salicylic Acid
Reference solution (a). A 0.0024 per cent w/v of salbutamol
sulphate RS in water. COOH
OH
Reference solution (b). A 0.048 per cent w/v of 2-tert-
butylamino-1-(4-hydroxy-3-methylphenyl) ethanol sulphate
RS and 0.048 per cent w/v of salbutamol sulphate RS in
methanol (10 per cent). C7H6O3 Mol. Wt. 138.1
Chromatographic system Salicylic Acid is 2-hydroxybenzoic acid.
– a stainless steel column 20 cm x 5 mm, packed with
spherical particles of silica, 5 µm in diameter, the surface Salicylic Acid contains not less than 99.0 per cent and not
of which has been modified with chemically-bonded more than 100.5 per cent of C7H6O3, calculated on the dried
nitrile groups (such as Spherisorb CN), basis.
– mobile phase: a mixture of 65 volumes of water, Description. White or colourless, acicular crystals or a white,
30 volumes of 0.05 M ammonium acetate and 5 volumes crystalline powder.
of 2-propanol, the pH of the mixture being adjusted to
4.5 with glacial acetic acid, Identification
– flow rate. 2 ml per minute,
Test A may be omitted if tests B and C are carried out. Test B
– spectrophotometer set at 276 nm,
may be omitted if tests A and C are carried out.
– a 20 µl loop injector.
A. Determine by infrared absorption spectrophotometry (2.4.6).
The test is not valid unless resolution between two principal
Compare the spectrum with that obtained with salicylic acid
peaks in the chromatogram obtained with reference solution
RS or with the reference spectrum of salicylic acid.
(b) is at least 1.5.
B. Dissolve about 30 mg in 5 ml of 0.05 M sodium hydroxide,
Calculate the content of C13H21NO3 in the tablet.
neutralise if necessary and dilute to 20 ml with water. 1 ml of
Other tests. Comply with the tests stated under Tablets. the solution gives reaction A of salicylates (2.3.1).
Assay. Determine by liquid chromatography (2.4.14). C. Melting point. 158º to 161º (2.4.21).

1066
IP 2007 SALMETEROL XINAFOATE

Tests Salmeterol Xinafoate contains not less than 97.0 per cent and
not more than 102.0 per cent of
Appearance of solution. Dissolve 1.0 g in 10 ml of ethanol
(95 per cent). The resulting solution is clear, and colourless C36H45NO7, calculated on the anhydrous basis.
(2.4.1). Description. A white to off-white powder.
Heavy metals (2.3.13). Dissolve 2.0 g in 15 ml of ethanol
Identification
(95 per cent) and add 5 ml of water. 12 ml of the resulting
solution complies with the limit test for heavy metals, Method Determine by infrared absorption spectrophotometry (2.4.6).
D (20 ppm). Use lead standard solution (100 ppm Pb) diluted Compare the spectrum with that obtained with salmeterol
with a mixture of 3 volumes of ethanol (95 per cent) and xinafoate RS or with the reference spectrum of salmeterol
1 volume of water to contain 2 µg of Pb per ml to prepare the xinafoate.
standard.
Tests
Iron (2.3.14). Boil 12.0 g with 14 ml of dilute ammonia solution
and 35 ml of water. Cool and adjust the pH 5.0 to 6.0 by the Related substances. Determine by liquid chromatography
dropwise addition of dilute ammonia solution or dilute (2.4.14).
sulphuric acid and dilute to 50 ml with water, if necessary. Test solution. Dissolve 50 mg of the substance under
Any pink colour produced is not more intense than that examination in 50 ml of the mobile phase.
obtained by boiling 2.0 g with 1 ml of iron standard solution
(20 ppm Fe), 2 ml of dilute ammonia solution and 45 ml of Reference solution. A 0.002 per cent w/v solution of salmeterol
water, adjusting the pH 5.0 to 6.0 and diluting to 50 ml with xinafoate RS in the mobile phase.
water (2 ppm). Chromatographic system
Chlorides (2.3.12). Dissolve 5.0 g in 50 ml of boiling distilled – a stainless steel column 25 cm x 4.6 mm, packed with
water, cool and filter (solution A). 20 ml of solution A complies octadecylsilane bonded to porous silica (5 µm),
with the limit test for chlorides (125 ppm). – mobile phase: a mixture of 60 volumes of a buffer solution
prepared by dissolving 0.0816 g of potassium
Sulphates (2.3.17). 7.5 ml of solution A complies with the limit dihydrogen phosphate in 1000 ml of water and adjusting
test for sulphates (200 ppm). the pH to 7.0 with triethylamine and 40 volumes of
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined acetonitrile,
on 2.0 g. – flow rate. 1 ml per minute,
– spectrophotometer set at 220 nm,
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
– a 10 µl loop injector.
on 1.0 g by drying over phosphorus pentoxide in a desiccator.
Inject the reference solution. The test is not valid unless the
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of
tailing factor is not more than 2.0 and the column efficiency in
ethanol (95 per cent), add 20 ml of water and titrate with
not less than 4500 theoretical plates.
0.1 M sodium hydroxide, using phenol red solution as
indicator, until a reddish violet colour is obtained. Inject the test solution. Any individual impurity is not more
than 0.5 per cent and the sum of all the impurities found is not
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01381 g of
more than 1.0 per cent.
C7H6O3.
Heavy metals (2.3.13). 1 g complies with limit test for heavy
Storage. Store protected from light.
metals, Method B (20 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Salmeterol Xinafoate Water (2.3.43). Not more than 0.5 per cent, determined on
0.5 g.
OH
OH H COOH Assay. Weigh accurately about 0.2 g and dissolve in 50 ml of
N ,
HO O glacial acetic acid. Titrate with 0.1 M perchloric acid,
HO determining the end-point potentiometrically (2.4.25). Carry
out a blank titration.
C25H37NO4, C11H8O3 Mol. Wt. 603.74
1 ml of 0.1 M perchloric acid is equivalent to 0.06038 g of
Salmeterol Xinafoate is (RS)-4-hydroxy-α′-[[[6-(4-
C36H45NO7.
phenylbutoxy)hexyl]amino]methyl]-1,3-benzenedimethanol
1-hydroxy-2-naphthoate. Storage. Store protected from light.

1067
SALMETEROL AND FLUTICASONE PROPIONATE INHALATION IP 2007

Salmeterol and Fluticasone Propionate – mobile phase: a mixture of 45 volumes of a buffer solution
prepared by dissolving 1.3 g of diammonium hydrogen
Inhalation orthophosphate to 1000 ml of water and adjusting the
Salmeterol and Fluticasone Propionate Inhalation is a pH to 7.0 with orthophosphoric acid, 25 volumes of
suspension of microfine Salmeterol Xinafoate and Fluticasone acetonitrile and 30 volumes of methanol,
Propionate in a suitable liquid filled in a suitable pressurised – flow rate. 2 ml per minute,
container. It may contain suitable pharmaceutical aids such as – spectrophotometer set at 220 nm,
surfactants, stabilizing agents. – inject 200 µl.
Salmeterol and Fluticasone Propionate Inhalation delivers not Inject reference solution(c). The test is not valid unless the
less than 80.0 per cent and not more than 120.0 per cent of the column efficiency for salmeterol and fluticasone propionate
stated amounts of salmeterol, C25H37NO4 and fluticasone peak is not less than 1000 and 2500 theoretical plates
propionate, C25H31F3O5S, per inhalation by actuation of the respectively and the tailing factor is not more than 2.0 for each
valve. peak and the relative standard deviation for replicate injections
for each component is not more than 2.0 per cent.
Identification Inject the test solution and reference solution (c).
In the Assay, the principal peaks in the chromatogram obtained Calculate the contents of C25H37NO4 and C25H31F3O5S in the
with test solution correspond to the peaks in the solution and the contents of C25H37NO4 and C25H31F3O5S
chromatogram obtained with reference solution (c). delivered per actuation of the valve.
Tests Determine the contents of the active ingredients a second
and third time by repeating the procedure on the middle ten
Other tests. Comply with the tests stated under Inhalation and on the last ten successive combined actuations of the
Preparations (Pressurised Metered-dose Preparations). valve. For each of the three determinations the average
Follow the procedure described under Assay with suitable contents of C25H37NO4 and C25H31F3O5S delivered per actuation
dilution of the reference solution wherever the amount of of the valve meet the requirements.
active substance is to be determined in any test. Storage. Store protected from moisture at a temperature not
Assay. Carry out the test for Content of active ingredient exceeding 30º.
delivered per actuation stated under Inhalation Preparations Labelling. The label states the amounts of active ingredients
(Pressurised Metered-dose Preparations). delivered per inhalation.
Determine by liquid chromatography (2.4.14).
Test solution. Prepare using the mobile phase as described
under the test for Content of active ingredient delivered per
Salmeterol and Fluticasone Propionate
actuation stated under Inhalation Preparations (Pressurised Powder for Inhalation
Metered-dose Preparations).
Salmeterol and Fluticasone Propionate Powder for Inhalation
Reference solution (a). A solution containing 0.5 mg of consists of Fluticasone Propionate and Salmeterol Xinafoate
salmeterol per ml prepared by dissolving 10 mg of salmeterol in microfine powder either alone or admixed with Lactose in a
xinafoate RS in 10 ml acetonitrile and adding sufficient of the pre-metered unit for use in a suitable powder inhaler.
mobile phase to produce 20 ml.
Salmeterol and Fluticasone Propionate Powder for Inhalation
Reference solution (b) .A solution containing 0.5 mg of contains not less than 90.0 per cent and not more than 125.0
fluticasone propionate per ml prepared by dissolving 10 mg of per cent of the stated amounts of salmeterol C25H37NO4 and
fluticasone propionate RS in 10 ml acetonitrile and adding fluticasone propionate, C25H31F3O5S per unit dose.
sufficient of the mobile phase to produce 20 ml.
Identification
Reference solution (c). Dilute suitable volumes of reference
solution (a) and reference solution (b) with the mobile phase In the Assay, the principal peaks in the chromatogram obtained
to obtain a solution containing 5 µg of salmeterol and 50 µg with the test solution correspond to the peaks in the
per ml of fluticasone propionate per ml. chromatogram obtained with reference solution (c).
Chromatographic system
Tests
– a stainless steel column 15 cm x 3.9 mm, packed with
octylsilyl silica gel (5 µm), Other tests. Complies with the tests stated under the Inhalation
– column temperature 40º, Preparations (Powders for Inhalation).

1068
IP 2007 SAQUINAVIR

Follow the procedure described under Assay with suitable Saquinavir


dilution of the reference solution wherever the amount of
active substance is to be determined in any test.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. To 10 intact capsules add 10 ml of water, and H CH3
O N
disperse with the aid of ultrasound till the shells get O CH3
disintegrated. Add 60 ml of the mobile phase and mix further H CH3
N
with the aid of ultrasound for 10 minutes with intermittent N N N
shaking. Add sufficient of the mobile phase to produce 100.0 H H
O OH
ml. Dilute suitably with the mobile phase, if required, to get a
O H
final concentration of 5 µg per ml of Salmeterol in the mobile NH2
phase.
Reference solution (a). A solution containing 0.5 mg of
salmeterol per ml prepared by dissolving 10 mg of salmeterol C38H50N6O5 Mol. Wt. 670.8
xinafoate RS in 10 ml acetonitrile and adding sufficient of the Saquinavir is (S)-N-[(αS)-α-{(1R)-2-[(3S,4aS,8aS)-3-(tert-
mobile phase to produce 20 ml. butylcarbamoyl)octahydro-2(1H)-isoquinolyl]-1-
Reference solution (b). A solution containing 0.5 mg of hydroxyethyl}phenethyl]-2-quinaldamidosuccinamide.
fluticasone propionate per ml prepared by dissolving 10 mg of Saquinavir contains not less than 98.5 per cent and not more
fluticasone propionate RS in 10 ml acetonitrile and adding than 101.0 per cent of C38H50N6O5, calculated on the anhydrous
sufficient of the mobile phase to produce 20 ml. basis.
Reference solution (c). Dilute suitable volumes of reference Description. A white crystalline powder.
solution (a) and reference solution (b) with the mobile phase
to obtain a solution containing 5 µg of salmeterol and 50 µg Identification
per ml of fluticasone propionate per ml.
A. Determine by infrared absorption spectrophotometry (2.4.6),
Chromatographic system
Compare the spectrum with that obtained with saquinavir RS
– a stainless steel column 15 cm x 3.9 mm, packed with
or with the reference spectrum of saquinavir.
octylsilyl silica gel (5 mm),
– column temperature 40º, B. In the Assay, the principal peak in the chromatogram
– mobile phase: a mixture of 45 volumes of a buffer solution obtained with the test solution corresponds to the peak due
prepared by dissolving 1.3 g of diammonium hydrogen to saquinavir in the chromatogram obtained with reference
orthophosphate to 1000 ml of water and adjusting the solution (a).
pH to 7.0 with orthophosphoric acid, 25 volumes of C. When examined in the range 220 nm to 350 nm (2.4.7), a
acetonitrile and 30 volumes of methanol, 0.002 per cent w/v solution in methanol shows absorption
– flow rate. 2 ml per minute, maxima at about 239 nm and 290 nm.
– spectrophotometer set at 220 nm,
– inject 200 µl. Tests
Inject reference solution(c). The test is not valid unless the
column efficiency determined from the salmeterol and Specific optical rotation (2.4.22). –50.0º to –55.0º, determined
fluticasone propionate peak is not less than 1000 and 2500 in a 0.5 per cent w/v solution in methanol.
theoretical plates respectively, the tailing factor for each of Related substances. Determine by liquid chromatography
salmeterol and fluticasone propionate peaks is not more than (2.4.14), as described in the Assay using the test solution and
2.0 and the relative standard deviation for replicate injections reference solution (c).
is not more than 2.0 per cent. Inject reference solution (c). Calculate the amount of related
Inject the test solution and reference solution (c). substance by area normalisation method. In the chromatogram
Calculate the contents of C25H37NO4 and C25H31F3O5S per unit. obtained with the test solution, the area of any peak other
than the principal peak is not greater than the area of the
Storage. Store protected from moisture, at temperature not principal peak obtained with reference solution (c) (0.1 per
exceeding 30º. cent) and the sum of the areas of all such peaks is not greater
Labelling. The label states the quantities of active ingredients than 5 times the area of the principal peak in the chromatogram
per pre-metered unit. obtained with reference solution (c) (0.5 per cent).

1069
SAQUINAVIR MESYLATE IP 2007

Heavy metals (2.3.13). 2.0 g complies with the limit tests for Saquinavir Mesylate
heavy metals, Method B (10 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
H CH3
Water (2.3.43). Not more than 2.0 per cent, determined on O N
O CH3
0.5 g. H CH3
N
N N N , CH3SO3H
Assay. Determine by liquid chromatography (2.4.14). H H
O OH
Test solution. Dissolve 25.0 mg of the substance under O H
NH2
examination in 100.0 ml of the mobile phase.
Reference solution (a). A 0.025 per cent w/v solution of C38H50N6O5,CH4O3S Mol. Wt. 767.0
saquinavir RS in the mobile phase.
Saquinavir mesylate is (S)-N-[(αS)-α-{(1R)-2-[(3S,4aS,8aS)-
Reference solution (b). Dissolve suitable quantities of 3-(tert-butylcarbamoyl)octahydro-2(1H)-isoquinolyl]-1-
saquinavir-related compound A RS and saquinavir RS in the hydroxyethyl}phenethyl]-2-quinaldamidosuccinamide
mobile phase to obtain a solution containing 2 µg per ml of methanesulphonate.
saquinavir-related compound A and 0.25 mg per ml of
Saquinavir Mesylate contains not less than 98.5 per cent and
saquinavir.
not more than 101.0 per cent of C38H50N6O5, CH4O3S, calculated
Reference solution (c). A 0.000025 per cent w/v solution of on the anhydrous basis.
saquinavir RS in the mobile phase.
Description. A white or almost white powder.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Identification
octadecylsilane bonded to porous silica (5 µm), A. Determine by infrared absorption spectrophotometry (2.4.6),
– mobile phase: a mixture of 20 volumes of methanol, Compare the spectrum with that obtained with saquinavir
50 volumes of acetonitrile and 30 volumes of a buffer mesylate RS or with the reference spectrum of saquinavir
prepared by dissolving 4 g of sodium dihydrogen mesylate.
phosphate in 1000.0 ml of water to which 1 ml of
diethylamine and 1 g of sodium octane sulphonate has B. In the Assay, the principal peak in the chromatogram
been added, obtained with the test solution corresponds to the peak due
– flow rate. 1 ml per minute, to saquinavir mesylate in the chromatogram obtained with
– spectrophotometer set at 210 nm, reference solution (a).
– a 20 µl loop injector.
Tests
Inject reference solution (b). The relative retention times are
Specific optical rotation (2.4.22). –66.8º to –69.6º, determined
about 0.89 for saquinavir-related compound A and about 1.0
in a 0.5 per cent w/v solution in methanol at 436 nm.
for saquinavir. The test is not valid unless the resolution
between the peaks due to saquinavir-related compound A Related substances. Determine by liquid chromatography
and saquinavir is not less than 1.5, the column efficiency (2.4.14), as described in the Assay using the test solution and
determined from the saquinavir peak is not less than 500 reference solution (c).
theoretical plates and the relative standard deviation for Inject reference solution (c). Calculate the amount of related
replicate injections is not more than 2.0 per cent. substances by area normalisation method. In the
Separately inject the test solution and reference solution (a). chromatogram obtained with the test solution, the area of any
Record the chromatograms up to three times the retention peak other than the principal peak is not greater than the area
time of the principal peak and measure the responses for the of the principal peak in the chromatogram obtained with
principal peak. reference solution (c) (0.1 per cent) and the sum of the areas of
all such peaks is not greater than 5 times the area of the
Calculate the content of C38H50N6O5.
principal peak in the chromatogram obtained with reference
Storage. Store protected from light. solution (c) (0.5 per cent).
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Methanesulphonic acid. 11.9 to 13.1 per cent w/w, calculated
on the anhydrous basis, determined by the following method.

1070
IP 2007 SAQUINAVIR MESYLATE TABLETS

Weigh accurately about 0.1 g of the substance under Saquinavir Mesylate Tablets
examination, dissolve in 50 ml of methanol. Titrate with 0.1 M
sodium hydroxide, determining the end-point Saquinavir Mesylate Tablets contain not less than 90.0 per
potentiometrically (2.4.25). Carry out a blank titration. cent and not more than 110.0 per cent of the stated amount of
saquinavir C38H50N6O5. The tablets may be coated.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.00961 g of
CH3SO3H. NOTE —Perform the tests and assay using low-actinic
glassware.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Water (2.3.43). Not more than 1.0 per cent, determined on 0.5 g. Identification
Assay. Determine by liquid chromatography (2.4.14). A. In the Assay, the principal spot in the chromatogram
Test solution. Dissolve 25.0 mg of the substance under obtained with the test solution corresponds to that in the
examination in 100.0 ml of the mobile phase. chromatogram obtained with the reference solution.
Reference solution (a). A 0.025 per cent w/v solution of B. When examined in the range 200 nm to 400 nm (2.4.7), 1 ml
saquinavir mesylate RS in the mobile phase. of a 0.1 per cent w/v solution in methanol diluted to 100 ml
Reference solution (b). Dissolve suitable quantities of with citrate buffer pH 3.0 (see under Dissolution), shows
saquinavir-related compound A RS and saquinavir mesylate absorption maxima at the same wavelengths as shown by the
RS in the mobile phase to obtain a solution containing 2 µg reference solution.
per ml of saquinavir- related compound A and 0.25 mg per ml
of saquinavir mesylate.
Tests
Reference solution (c). A 0.000025 per cent w/v solution of Dissolution (2.5.2).
saquinavir mesylate RS in the mobile phase. Apparatus. No 1
NOTE — Store the buffer solution protected from light. Make Medium. 900 ml of citrate buffer pH 3.0 prepared by dissolving
adjustments if necessary for system suitability. 5.82 mg of anhydrous dibasic sodium phosphate and 16.7 mg
Chromatographic system of citiric acid monohydrate in 1000 ml of water and adjusting
– a stainless steel column 25 cm x 4.6 mm, packed with the pH to 3.0 with orthophosphoric acid.
octadecylsilane bonded to porous silica (5 µm), Speed and time. 50 rpm and 45 minutes.
– mobile phase: a mixture of 25 volumes of Withdraw a suitable volume of the medium and filter promptly.
tetrahydrofuran, 5 volumes of acetonitrile and Dilute the filtrate, if necessary, with the medium. Measure the
17 volumes of a buffer prepared by mixing 10 ml of absorbance (2.4.7) of the resulting solution at the maximum at
triethylamine with water to make 1000 ml and adjusting about 240 nm. Calculate the content of C38H50N6O5 in the
the pH of the solution to 2.5 with phosphoric acid, medium from the absorbance obtained from a solution of
– flow rate. 1 ml per minute, known concentration of saquinavir mesylate RS.
– spectrophotometer set at 210 nm,
– a 20 µl loop injector. D. Not less than 75 per cent of the stated amount of
C38H50N6O5.
Inject reference solution (b). The relative retention times are
about 0.89 for saquinavir-related compound A and about 1.0 Related substances. Determine by liquid chromatography
for saquinavir mesylate. The test is not valid unless the (2.4.14).
resolution between the peaks due to saquinavir related Test solution. Weigh and powder 20 tablets Weigh accurately
compound A and saquinavir mesylate is not less than 1.5, the a quantity of the powder containing 100 mg of saquinavir in
column efficiency determined from the saquinavir mesylate 100 ml of the mobile phase and filter.
peak is not less than 500 theoretical plates and the relative
Reference solution (a). A 0.1 per cent w/v solution of
standard deviation for replicate injections is not more than
saquinavir mesylate RS in the mobile phase.
2.0 per cent.
Reference solution (b). Dilute 1 ml of reference solution (a) to
Separately inject the test solution and reference solution (a).
100 ml with the mobile phase.
Record the chromatograms upto three times the retention time
of the principal peak and measure the responses for the Chromatographic system
principal peak. – a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm),
Calculate the content of C38H50N6O5, CH4O3S.
– mobile phase: a mixture of 14 volumes of triethylamine
Storage. Store protected from light. phosphate solution prepared by diluting 10 ml of

1071
SECNIDAZOLE IP 2007

triethylamine to 1000 ml with water and adjusting the Inject the reference solution. The test is not valid unless the
pH to 2.5 with orthophosphoric acid, 5 volumes of tailing factor is not more than 2.0, the column efficiency in not
tetrahydrofuran and 1 volume of acetonitrile, less than 2000 theoretical plates and the relative standard
– flow rate. 1 ml per minute, deviation for replicate injections is not more than 2.0 per cent.
– spectrophotometer set at 210 nm, Inject the test solution and the reference solution.
– a 20 µl loop injector.
Calculate the content of C38H50N6O5 in the tablets.
Inject reference solution (a). The test is not valid unless the
column efficiency is not less than 3000 theoretical plates and Storage. Store protected from moisture.
the tailing factor is not more than 2.0. Labelling. The label states the strength in terms of the
Inject the test solution and reference solution (b). Run the equivalent amount of saquinavir.
chromatogram for 5 times the retention time (about 12 minutes)
of the principal peak. In the chromatogram obtained with the
test solution, the area of any secondary peak is not more than
0.2 times the area of the peak in the chromatogram obtained Secnidazole
with the reference solution (b) (0.2 per cent) and the sum of
areas of all the secondary peaks is not more than the area of OH
the peak in the chromatogram obtained with the reference
solution (b) (1.0 per cent). CH3
Uniformity of content. Comply with the test stated under O2N N CH3
Tablets.
N
Determine by liquid chromatography (2.4.14), as described
under Assay.
C7H11N3O3 Mol. Wt. 185.2
Test solution. Disperse one tablet in 500 ml of the mobile phase
and filter. Dilute 5 ml of the filtrate to 20 ml with the mobile Secnidazole is (RS)-1-(2-methyl-5-nitroimidazol-1-yl)propan-
phase. 2-ol.

Calculate the content of C38H50N6O5 in the tablet. Secnidazole contains not less than 98.0 per cent and not more
than 101.0 per cent of C6H9N3O3, calculated on the anhydrous
Other tests. Comply with the tests stated under Tablets. basis.
Water (2.3.43). Not more than 6.0 per cent, determined on Description. A white to yellowish white, crystalline powder.
0.5 g.
Assay. Determine by liquid chromatography (2.4.14). Identification
Test solution. Weigh and powder 20 tablets. Weigh accurately A. When examined in the range 230 nm to 360 nm (2.4.7), a
a quantity of the powder containing 100 mg of saquinavir, 0.001 per cent w/v solution in 0.1 M hydrochloric acid shows
dissolve in 100.0 ml of mobile phase and filter. Dilute 5.0 ml of an absorption maximum at about 277 nm, 0.325 to 0.355.
the filtrate to 20.0 ml with the mobile phase. B. Heat about 10 mg in a water-bath with 10 mg of zinc powder,
Reference solution. A 0.1 per cent w/v solution of saquinavir 1 ml of water and 0.25 ml of 2 M hydrochloric acid for
mesylate RS in the mobile phase. Dilute 5.0 ml of the solution 5 minutes and cool. The solution gives the reaction of primary
to 20.0 ml with the mobile phase. aromatic amines (2.3.1).
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with
Tests
octadecylsilane bonded to porous silica (5 µm), Appearance of solution. A 5 per cent w/v solution in 1 M
– mobile phase: a mixture of 14 volumes of a solution hydrochloric acid is not more opalescent than opalescence
prepared by diluting 10.0 ml of triethylamine to1000 ml standard OS2 (2.4.1) and not more intensely coloured than
with water, adjusting the pH to 2.5 with orthophosphoric reference solution GYS4 (2.4.1).
acid and filtering, 5 volumes of tetrahydrofuran and
Related substances. Determine by liquid chromatography
1 volume of acetonitrile.
(2.4.14).
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 210 nm, Test solution. Dissolve 50 mg of the substance under
– a 20 µl loop injector. examination in 100.0 ml of the mobile phase.

1072
IP 2007 SECNIDAZOLE TABLETS

Reference solution (a). Dilute 1.0 ml of the test solution to Inject the reference solution. The test is not valid unless the
100.0 ml with mobile phase. Dilute 1.0 ml of the solution to tailing factor is not more than 2.0 and the relative standard
10.0 ml with the same solvent. deviation for replicate injections is not more than 2.0 per cent.
Reference solution (b). A solution containing 0.005 per cent Inject the test solution and the reference solution.
w/v each of 2-methyl-5-nitroimidazole RS and secnidazole
Calculate the content of C7H11N3O3.
RS in the mobile phase. Dilute 1.0 ml of the solution to 100.0
ml with the same solvent. Storage. Store protected from light and moisture.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 35 volumes of methanol and Secnidazole Tablets
65 volumes of 0.14 per cent w/v solution of potassium Secnidazole Tablets contain not less than 95.0 per cent and
dihydrogen phosphate, not more than 110.0 per cent of the stated amount of
– flow rate. 1 ml per minute, secnidazole, C7H11N3O3.
– spectrophotometer set at 318 nm,
– a 10 µl loop injector. Identification
Inject reference solution (b). The test is not valid unless the In the Assay, the principal peak in the chromatogram obtained
resolution between 2-methyl-4-nitroimidazole and secnidazole with the test solution corresponds to the peak in the
is not less than 1.5. chromatogram obtained with the reference solution.
Inject the test solution and reference solution (a). In the
chromatogram obtained with the test solution, the area of any Tests
secondary peak is not more than 0.2 times the area of the peak Dissolution (2.5.2).
in the chromatogram obtained with reference solution (a)
(0.2 per cent) and the sum of areas of all the secondary peaks Apparatus. No 1
is not more than 0.5 times the area of the peak in the Medium. 900 ml of 0.1 M hydrochloric acid.
chromatogram obtained with the reference solution (b) (0.5 Speed and time. 100 rpm and 30 minutes.
per cent). Disregard any peak which is 0.1 times the area of the Withdraw a suitable volume of the medium and filter promptly
principal peak in the chromatogram obtained with reference through a membrane filter disc having an average pore diameter
solution (a) (0.01 per cent). not greater than 1.0 µm, rejecting the first 1 ml of the filtrate.
Heavy metals (2.3.13).1.0 g complies with the limit test for Dilute the filtrate, if necessary, with the same solvent. Measure
heavy metals, Method A (20 ppm). the absorbance of the resulting solution at the maximum at
about 277 nm (2.4.7). Calculate the content of C7H11N3O3 in the
Sulphated ash (2.3.18). Not more than 0.1 per cent. medium from the absorbance obtained from a solution of
Water (2.3.43). 4.0 to 5.0 per cent, determined on 0.4 g. known concentration of secnidazole RS in the same medium.
Assay. Determine by liquid chromatography (2.4.14). D. Not less than 80 per cent of the stated amount of C7H11N3O3.
Test solution. Dissolve 25 mg of the substance under Related substances. Determine by liquid chromatography
examination in 25.0 ml of the mobile phase. Dilute 5.0 ml of the (2.4.14).
solution to 100.0 ml with the same solvent. Test solution. Weigh accurately a quantity of powdered tablets
Reference solution. A 0.005 per cent w/v solution of containing 50 mg of Secnidazole, disperse in 100 ml of mobile
secnidazole RS in mobile phase. phase and filter.
Chromatographic system Reference solution (a). A 0.05 per cent w/v solution of
– a stainless steel column 25 cm x 4.6 mm packed with secnidazole RS in the mobile phase.
octadecylsilane bonded to porous silica (5 µm), Reference solution (b). Dilute 1 ml of reference solution (a) to
– mobile phase: a mixture of 35 volumes of methanol and 100 ml with mobile phase.
65 volumes of 0.14 per cent w/v solution of potassium
dihydrogen phosphate, Chromatographic system as described under Assay.
– flow rate. 1 ml per minute, Inject reference solution (a). The test is not valid unless the
– spectrophotometer set at 318 nm, tailing factor is not more than 2.0 and the column efficiency in
– a 20 µl loop injector. not less than 2000 theoretical plates.

1073
COLLOIDAL SILICON DIOXIDE IP 2007

Inject the test solution and reference solution (b). In the Identification
chromatogram obtained with the test solution, the area of any
secondary peak is not more than the area of the peak in the About 20 mg gives the reaction of silicates (2.3.1).
chromatogram obtained with reference solution (b) (1.0 per Tests
cent) and the sum of areas of all the secondary peaks is not
more than 2.0 times the area of the peak in the chromatogram pH (2.4.24). 3.5 to 5.5, determined in a suspension of 1.0 g in
obtained with the reference solution (b) (2.0 per cent). 30 ml of carbon dioxide-free water.
Water (2.3.43). Not more than 6.5 per cent, determined on 0.5 g. Arsenic (2.3.10). To 2.5 g contained in a round-bottomed flask
add 50 ml of 3 M hydrochloric acid and heat under a reflux
Assay. Determine by liquid chromatography (2.4.14).
condenser for 30 minutes. Cool, filter with the aid of suction
Test solution. Weigh and powder 20 tablets. Weigh accurately and transfer the filtrate to a 100-ml volumetric flask. Wash the
a quantity of powdered tablet containing 50 mg of Secnidazole, filter with several portions of hot water and add the washings
disperse in 100.0 ml of mobile phase and filter.. Dilute 5.0 ml of to the volumetric flask. Cool, dilute to volume with water and
the filtrate to 50.0 ml with mobile phase. mix. To 50.0 ml of the solution add 3 ml of hydrochloric acid;
Reference solution. A 0.005 per cent w/v solution of the resulting solution complies with the limit test for arsenic
secnidazole RS in mobile phase. (8 ppm).

Chromatographic system Heavy metals (2.3.13). Suspend 2.5 g in sufficient water to


– a stainless steel column 25 cm x 4.6 mm packed with produce a semi-fluid slurry and dry at 140º. When the dried
octadecylsilane chemically bonded to porous silica substance is white, break up the mass using a glass rod, add
(5 µm), 25 ml of 1 M hydrochloric acid, boil gently for 5 minutes,
– mobile phase: a mixture of 85 volumes of 0.01 M stirring frequently with the glass rod, centrifuge for 20 minutes
potassium dihydrogen orthophosphate and 15 volumes and filter the supernatant liquid through a membrane filter. To
of acetonitrile, the residue in the centrifuge tube add 3 ml of 2 M hydrochloric
– flow rate. 1 ml per minute, acid and 9 ml of water, boil, centrifuge for 20 minutes and filter
– spectrophotometer set at 228 nm, the supernatant liquid through the same membrane filter. Wash
– a 20 µl loop injector. the residue with small quantities of water, combine the filtrates
and washings and dilute to 50.0 ml with water. To 20.0 ml of
Inject the reference solution. The test is not valid unless the the solution add 50 mg of L-ascorbic acid and 1 ml of strong
column efficiency is not less than 2000 theoretical plates, the ammonia solution, neutralise with 2 M ammonia and dilute to
tailing factor is not more than 2.0 and the relative standard 25 ml with water. 12 ml of the solution complies with the limit
deviation of replicate injections is not more than 2.0 per cent. test for heavy metals, Method D (25 ppm). Use lead standard
Inject the test solution and the reference solution. solution (1 ppm Pb) to prepare the standard.
Calculate the content of C7H11N3O3. Chlorides (2.3.12). To 1.0 g add a mixture of 20 ml of 2 M nitric
acid and 30 ml of water, heat on a water-bath for 15 minutes,
Storage. Store protected from light and moisture.
shaking frequently, dilute to 50 ml with water if necessary,
Labelling. The label states the strength of secnidazole. filter and cool. The filtrate complies with the limit test for
chlorides (250 ppm).
Loss on ignition (2.4.20). Not more than 5.0 per cent,
Colloidal Silicon Dioxide determined on 0.2 g by igniting at 900º in a platinum crucible
for 2 hours.
Colloidal Anhydrous Silica
Assay. To the residue obtained in the test for Loss on ignition
SiO2 Mol. Wt. 60.1 add 0.2 ml of sulphuric acid and sufficient ethanol (95 per
Colloidal Silicon Dioxide is a submicroscopic fumed silica cent) to moisten the residue completely, add 6 ml of
prepared by the vapour-phase hydrolysis of a silicon hydrofluoric acid and evaporate to dryness on a hot plate at
compound. 95º to 105º, avoiding loss from sputtering. Wash the sides of
the dish with 6 ml of hydrofluoric acid, evaporate to dryness
Colloidal Silicon Dioxide contains not less than 99.0 per cent in a well-ventilated hood, ignite at 1000º, allow to cool in a
and not more than 100.5 per cent of SiO2, calculated on the desiccator and weigh. The difference between the weight of
ignite basis. the final residue and that of the residue obtained in the test for
Description. A light, fine, white, amorphous powder. It has a Loss on ignition represents the amount of SiO2 in the amount
particle size of about 15 nm. of the substance taken for the test for Loss on ignition.

1074
IP 2007 SODIUM ACETATE

Storage. Store protected from light. Identification


Dissolve 10.0 g in sufficient carbon dioxide-free water to
produce 100.0 ml (solution A). 1 ml of solution A gives reaction
Silver Nitrate B of acetates and reaction A of sodium salts (2.3.1).
AgNO3 Mol. Wt. 169.9
Tests
Silver Nitrate contains not less than 99.0 per cent and not
more than 100.5 per cent of AgNO3. Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Description. Colourless crystals or a white, crystalline powder.
pH (2.4.24). 7.5 to 9.0, determined in a 5.0 per cent w/v solution.
Identification
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 15 ml
A. Gives the reactions of silver salts (2.3.1). of stannated hydrochloric acid AsT. The resulting solution
complies with the limit test for arsenic (2 ppm).
B. 10 mg gives reaction A of nitrates (2.3.1).
Calcium and magnesium. Not more than 50 ppm, calculated
Tests as Ca, determined by the following method. Mix 200 ml of
Appearance of solution. A 4.0 per cent w/v solution (solution water with 10 ml of ammonia buffer pH 10.0, 0.1 g of mordant
A) is clear (2.4.1), and colourless (2.4.1). black 11 mixture and 2 ml of 0.05 M zinc chloride. Add
dropwise 0.02 M disodium edetate until the colour changes
Acidity or alkalinity. To 2 ml of solution A add 0.1 ml of from violet to blue. To this solution add 10 g of the substance
bromocresol green solution; the solution is blue. To 2 ml of under examination, shake to dissolve and titrate with 0.02 M
this solution add 0.1 ml of phenol red solution; the solution is disodium edetate until the blue colour is restored. Not more
yellow. than 0.65 ml of 0.02 M disodium edetate is required.
Aluminium, bismuth, copper and lead. Dissolve 1.0 g in a Heavy metals (2.3.13).12 ml of solution A complies with the
mixture of 4 ml of strong ammonia solution and 6 ml of water; limit test for heavy metals, Method D (10 ppm).
the resulting solution is clear (2.4.1), and colourless (2.4.1).
Iron (2.3.14). 20 ml of solution A complies with the limit test for
Foreign salts. Not more than 0.3 per cent, determined by the iron (20 ppm).
following method. To 30 ml of solution A add 7.5 ml of 2 M
hydrochloric acid, shake vigorously, heat for 5 minutes on a Chlorides (2.3.12). 10 ml of solution A complies with the limit
water-bath, filter and evaporate 20 ml of the filtrate to dryness test for chlorides (250 ppm).
on a water-bath. Dry the residue at 105º and weigh. Sulphates (2.3.17). 15 ml of solution A complies with the limit
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of test for sulphates (225 ppm).
water, add 2 ml of 2 M nitric acid and 2 ml of ferric ammonium Reducing substances. Dissolve 1.0 g in 100 ml of boiling water,
sulphate solution and titrate with 0.1 M ammonium add 5 ml of 1 M sulphuric acid and 0.5 ml of 0.002 M potassium
thiocyanate until a reddish yellow colour is produced. permanganate, mix and boil gently for 5 minutes; the pink
1 ml of 0.1 M ammonium thiocyanate is equivalent to 0.01699 colour is not completely discharged.
g of AgNO3.
Loss on drying (2.4.19). 39.0 to 40.5 per cent, determined on
Storage. Store protected from light and moisture, in non- 0.2 g by drying in an oven at 130º. Place the substance under
metallic containers. examination in the oven while the oven is still cold.
Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of
anhydrous glacial acetic acid, add 5 ml of acetic anhydride,
Sodium Acetate mix and allow to stand for 30 minutes. Titrate with 0.1 M
perchloric acid, using 0.3 ml of 1-naphtholbenzein solution
CH3COONa,3H20 as indicator, until a green colour is produced. Carry out a
C2H3NaO2, 3H2O Mol. Wt. 136.1 blank titration.
Sodium Acetate contains not less than 99.0 per cent and not 1 ml of 0.1 M perchloric acid is equivalent to 0.00820 g of
more than 101.0 per cent of C2H3NaO2, calculated on the C2H3NaO2.
dried basis. Sodium Acetate intended for use in the preparation of dialysis
Description. Colourless crystals or a white, crystalline powder. solutions complies with the following additional requirement.

1075
SODIUM ALGINATE IP 2007

Aluminium. Dissolve 20 g in 100 ml of water and adjust to Tests


pH 6.0 by the addition of about 10 ml of 1 M hydrochloric
acid. Extract with successive quantities of 20, 20 and 10 ml of Heavy metals (2.3.13). 0.5 g complies with the limit test for
a 0.5 per cent w/v solution of 8-hydroxyquinoline in heavy metals, Method B (40 ppm), using nitric acid instead
chloroform and dilute the combined extracts to 50 ml with of sulphuric acid for wetting the sample.
chloroform. Use as the standard solution a mixture of 0.4 ml of Chloride. Not more than 1.0 per cent, determined by the
aluminium standard solution (2 ppm Al), 10 ml of acetate following method. Shake 2.5 g with 50 ml of 2 M nitric acid for
buffer pH 6.0 and 98 ml of water treated in the same manner 1 hour, dilute to 100 ml with 2M nitric acid and filter. To 50 ml of
and as the blank solution a mixture of 10 ml of acetate buffer the filtrate add 10.0 ml of 0.1 M silver nitrate and 5 ml of
pH 6.0 and 100 ml of water treated in the same manner. Measure toluene. Titrate with 0.1 M ammonium thiocyanate using 2 ml
the fluorescence (2.4.5) of the test solution and the standard of ferric ammonium sulphate solution as indicator and shaking
solution, using an excitation wavelength of about 392 nm and vigorously towards the end-point.
emission wavelength of about 518 nm, and setting the
1 ml of 0.1 M silver nitrate is equivalent to 0.00355 g of Cl.
instrument to zero with the blank solution in each case. The
fluorescence of the test solution is not greater than that of the Microbial contamination (2.2.9). 1 g is free from Escherichia
standard solution (0.2 ppm). coli; 10 g is free from salmonellae.
Storage. Store protected from light. Sulphated ash (2.3.18). 30.0 to 36.0 per cent, determined on
0.1 g and calculated on the dried basis.
Labelling. The label states whether or not the material is
intended for use in the manufacture of dialysis solutions. Loss on drying (2.4.19). Not more than 15.0 per cent,
determined on 0.2 g by drying in an oven at 105º for 4 hours.
Storage. Store protected from light.

Sodium Alginate
Sodium Aminosalicylate
Sodium Polymannuronate
Sodium PAS
Sodium Alginate consists mainly of the sodium salt of alginic
acid which is a mixture of polyuronic acids [(C6H8O6)n]
composed of residues of D-mannuronic acid and L-guluronic COONa
acids and is obtained mainly from algae belonging to the order OH
Phaeophyceae. ,2H2O
Description. A cream-coloured to pale yellowish brown
powder; almost odourless. NH2

Identification C7H6NNaO3.2H2O. Mol. Wt. 211.2

A. Dissolve 0.2 g with shaking in 20 ml of water and to 5 ml of Sodium Aminosalicylate is sodium 4-amino-2-
the resulting solution add 1 ml of calcium chloride solution; hydroxybenzoate dihydrate.
a voluminous gelatinous precipitate is produced. Sodium Aminosalicylate contains not less than 99.0 per cent
B. To 10 ml of the solution obtained in test A add 1 ml of 1 M and not more than 101.0 per cent of C7H6NNaO3, calculated on
sulphuric acid; a gelatinous mass is produced. the anhydrous basis.
Description. A white to cream coloured crystalline powder.
C. To 5 mg add 5 ml of water, 1 ml of a freshly prepared 1 per
cent w/v solution of naphthalene-1,3-diol in ethanol (95 per Identification
cent) and 5 ml of hydrochloric acid. Boil for 3 minutes, cool,
add 5 ml of water and shake with 15 ml of di-isopropyl ether. A. Determine by infrared absorption spectrophotometry (2.4.6).
The upper layer exhibits a deeper bluish red colour than the Compare the spectrum with that obtained with sodium
upper layer obtained by repeating the procedure without the aminosalicylate RS or with the reference spectrum of sodium
substance under examination. aminosalicylate.
D. The residue obtained in the test for Sulphated ash gives B. A 5 per cent w/v solution complies with the tests for sodium
reaction A of sodium salts (2.3.1). salts (2.3.1).

1076
IP 2007 SODIUM ALGINATE

Tests excess bromine with a few drops of stannous chloride solution


AsT. The resulting solution complies with the limit test for
pH (2.4.24). 6.5 to 8.5, determined in a 2.0 per cent w/v solution. arsenic (2 ppm).
3-aminophenol. Determine by liquid chromatography (2.4.14). Heavy metals (2.3.13). 0.66 g complies with the limit test for
Test solution. Dissolve 50.0 mg of the substance under heavy metals, Method B (30 ppm).
examination in 100 ml of the mobile phase. Chlorides (2.3.12). Dissolve 1.0 g in 10 ml of water, add 3 ml of
Reference solution (a). Dissolve 25.0 mg of 3-aminophenol acetic acid and filter, dilute the filtrate to 50 ml with water.
RS in 100.0 ml of the mobile phase (solution A). Dilute 5.0 ml of 10 ml of the solution complies with the limit test for chlorides
the solution to 100.0 ml with the mobile phase. Dilute 5.0 ml of (0.25 per cent).
the resulting solution to 50.0 ml with the mobile phase. Sulphates (2.3.17). 0.1 g complies with the limit test for
Reference solution (b). Dissolve 25.0 mg of sodium sulphates (0.15 per cent).
aminosalicylate RS in 100.0 ml of the mobile phase. Dilute Loss on drying (2.4.19). 16.0 to 17.5 per cent, determined on
5.0 ml of this solution and 5 ml of solution A to 100.0 ml with 0.5 g by drying in an oven at 105º.
the mobile phase.
Assay. Determine by liquid chromatography (2.4.14).
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with Test solution. Dissolve 50.0 mg of the substance under
octadecylsilane bonded to porous base deactivated examination in 100.0 ml of the mobile phase. Dilute 5.0 ml of
silica (5 µm) (such as Hypersil BDS), this solution to 100.0 ml with the mobile phase.
– mobile phase: dissolve 6.0 g of disodium hydrogen Reference solution (a). Dissolve 50.0 mg of sodium
orthophosphate and 6.6 g of sodium dihydrogen aminosalicylate RS in 100.0 ml of the mobile phase (solution
orthophosphate dihydrate in 1600 ml with water, add A). Dilute 5.0 ml of this solution to 100.0 ml with the mobile
19 ml of tetra n-butyl ammonium hydroxide (20 per phase.
cent solution) and make the volume to 1700 ml with
water and add 300 ml of methanol, Reference solution (b). Dissolve 25.0 mg of 3-aminophenol
– flow rate. 1.5 ml per minute, RS in 100.0 ml of the mobile phase. Dilute 5.0 ml of this solution
and 5 ml of solution A to 100.0 ml with the mobile phase.
– spectrophotometer set at 280 nm,
– a 20 µl loop injector. Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with
Inject reference solution (a). The relative standard deviation
octadecylsilane bonded to porous silica (5 µm),
for replicate injections is not more than 5.0 per cent
– mobile phase: dissolve 6.0 g of disodium hydrogen
Inject reference solution (b). The test is not valid unless the orthophosphate and 6.6 g of sodium dihydrogen
resolution between m-aminophenol and sodium orthophosphate dihydrate in 1600 ml with water, add
aminosalicylate is at least 5.0. 19 ml of tetra n-butyl ammonium hydroxide (20 per
cent solution) and make the volume to 1700 ml with
Inject the test solution and reference solution (a). In the
water and add 300 ml of methanol,
chromatogram obtained with the test solution, the area of the
– flow rate. 1.5 ml per minute,
peak corresponding to 3-aminophenol is not more than the
– spectrophotometer set at 254 nm,
area of the corresponding peak in the chromatogram obtained
– a 20 µl loop injector.
with reference solution (a) ( 0.25 per cent).
Inject reference solution (a). The tailing factor is not more
Hydrogen Sulphide and Sulphur dioxide. Dissolve 0.5 g in
than 2.0. The column efficiency in not less than 3000 theoretical
5 ml of 1 M sodium hydroxide, add 6 ml of 3 M hydrochloric
plates. The relative standard deviation for replicate injections
acid and stir vigorously. No odour of hydrogen sulphide or
is not more than 5.0 per cent.
sulpher dioxide is perceptible, and not more than a faint odour
of Amyl Alcohol is perceptible. A piece of moistened lead Inject reference solution (b). The test is not valid unless the
acetate paper held over the mixture does not become resolution between 3-aminophenol and sodium
discoloured. aminosalicylate is at least 5.0.
Arsenic (2.3.10). Mix 5.0 g with 10 ml of bromine solution and Inject alternately the test solution and the reference solution.
evaporate to dryness on a water-bath, ignite gently, dissolve
Calculate the content of C7H6NNaO3.
the cooled residue, ignoring any carbon, in 50 ml of water and
14 ml of brominated hydrochloric acid AsT and remove the Storage. Store protected from light and moisture.

1077
SODIUM AMINOSALICYLATE TABLETS IP 2007

Sodium Aminosalicylate Tablets cent aqueous solution) and make the volume to 1700 ml
with water, add 300 ml of methanol, mix well and degas,
Sodium PAS Tablets – flow rate. 1.5 ml per minute,
Sodium aminosalicylate tablet contains not less than 95.0 per – spectrophotometer set at 280 nm,
cent not more than 105.0 per cent of C7H6NNaO3.2H2O. – a 20 µl loop injector.
Inject reference solution (a). The relative standard deviation
Identification for replicate injections is not more than 5.0 per cent.
Digest a quantity of powdered tablets containing about 3.0 g Inject reference solution (c). The resolution between
of sodium aminosalicylate, with 40 ml of water, and filter. Add 3-aminophenol and sodium aminosalicylate is not less than
to the filtrate 15 ml of 1M acetic acid, and allow it to stand 5.0.
until precipitation has occurred. Collect the precipitate on a Inject the test solution and reference solution (a). In the
filter, wash well with water and dry at 105ºC for 30 min. The chromatogram obtained with the test solution, the area of the
residue complies with the following tests. peak corresponding to 3-aminophenol is not more than the
A. Place about 1 g of the residue in a small, round-bottom area of the peak in the chromatogram obtained with reference
flask, and add 10 ml of acetic anhydride. Heat the flask on a solution (a) ( 1.0 per cent).
steam bath for 30 minutes, and add 40 ml of water, mix, filter, Dissolution (2.5.2).
cool, and allow to stand until diacetyl derivative crystallizes,
wash well with water, and dry at 105º for 1 hour. The diacetyl Apparatus. No 2
derivative so obtained melts at 191º to 197º. Medium. 900 ml water
Speed and time.100 rpm for 45 minutes
B. Shake 0.1 g of the residue with 10 ml of water, and filter. To
5 ml of the filtrate add 1 drop of ferric chloride solution. A Withdraw a suitable volume of the medium and filter.
violet colour is produced. Determine by liquid chromatography (2.4.14).
C. In the Assay, the principal peak in the chromatogram Test solution. The filtrate diluted with the mobile phase to
obtained with the test solution corresponds to the peak in the produce a 0.055 per cent w/v solution.
chromatogram obtained with the reference solution.
Reference solution. A 0.055 per cent w/v solution of sodium
Tests aminosalicylate RS in the mobile phase.
Chromatographic system
3-aminophenol. Determine by liquid chromatography (2.4.14).
– a stainless steel column 25 cm x 4.6 mm, packed with
Test solution. Weigh and powder 20 tablets. Weigh a quantity octadecylsilane bonded to porous silica (5 µm),
of the powder containing about 50 mg of Sodium – mobile phase: dissolve 6.0 g of disodium hydrogen
Aminosalicylate and dissolve in 100 ml of the mobile phase. orthophosphate and 6.6 g of sodium dihydrogen
Reference solution (a). A 0.024 per cent w/v solution of orthophosphate dihydrate in 1600 ml with water, add
m-aminophenol RS in the mobile phase. Dilute 5 ml to 100 ml 19 ml of tetra n-butyl ammonium hydroxide (20 per
with the mobile phase. Dilute 5 ml of the resulting solution to cent aqueous solution) and make the volume to 1700 ml
50 ml with the mobile phase. with water, add 300 ml of methanol, mix well and degas,
– flow rate. 1.5 ml per minute,
Reference solution (b). A 0.024 per cent w/v solution of – spectrophotometer set at 254 nm,
sodium aminosalicylate RS in the mobile phase. – a 20 µl loop injector.
Reference solution (c). Mix 5 ml each of reference solution (a) Inject the reference solution. The tailing factor is not more
and reference solution (b) and dilute to 100 ml with the mobile than 2.0 and the relative standard deviation for replicate
phase. injections is not more than 2.0 per cent
Chromatographic system Inject alternatively the test solution and the reference solution.
– a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous base deactivated Calculate the content of C7H6NNaO3.2H2O in the medium.
silica (5 µm) (such as Hypersil BDS), D. Not less than 75 per cent of the stated amount of
– mobile phase: dissolve 6.0 g of disodium hydrogen C7H6NNaO3.2H2O.
orthophosphate and 6.6 g of sodium dihydrogen
Other tests. Comply with the tests stated under Tablets.
orthophosphate dihydrate in 1600 ml with water, add
19 ml of tetra n-butyl ammonium hydroxide (20 per Assay. Determine by liquid chromatography (2.4.14).

1078
IP 2007 SODIUM AUROTHIOMALATE

Test solution. Weigh and powder 20 tablets. Weigh accurately A. Determine by infrared absorption spectrophotometry (2.4.6),
a quantity of the powder containing about 0.5 g of sodium Compare the spectrum with that obtained with sodium
aminosalicylate, add sufficient mobile phase to produce ascorbate RS.
100.0 ml and filter. Dilute 5.0 ml of this solution to 50.0 ml with B. To 4 ml of a 2 per cent w/v solution add 1 ml of 0.1 M
the mobile phase. hydrochloric acid, add a few ml of 2,6-dichlorophenol-
Reference solution. A 0.05 per cent w/v solution of sodium indophenol solution; the solution is decolorised.
aminosalicylate RS in the mobile phase. C. To 4 ml of a 2 per cent w/v solution add 1 ml of 0.1 M
Chromatographic system hydrochloric acid, add 1 drop of a freshly prepared 5 per cent
– a stainless steel column 25 cm x 4.6 mm, packed with w/v solution of sodium nitroprusside and 2 ml of dilute sodium
octadecylsilane bonded to porous silica (5 µm), hydroxide solution. Add 0.6 ml of hydrochloric acid dropwise
– mobile phase: dissolve 6.0 g of disodium hydrogen and stir; the yellow colour turns blue.
orthophosphate and 6.6 g of sodium dihydrogen D. A 2 per cent w/v solution gives the reactions of sodium
orthophosphate dihydrate in 1600 ml with water, add salts (2.3.1).
19 ml of tetra n-butyl ammonium hydroxide (20 per
cent aqueous solution) and make the volume to 1700 ml Tests
with water, add 300 ml of methanol, mix well and degas,
Appearance of solution. A 5.0 per cent w/v solution in carbon
– flow rate. 1.5 ml per minute,
dioxide-free water is clear (2.4.1) and not more intensely
– spectrophotometer set at 254 nm,
coloured than reference solution BYS7 (2.4.1).
– a 20 µl loop injector.
pH (2.4.24). 7.0 to 8.0, determined in a 10.0 per cent w/v solution.
Inject the reference solution. The tailing factor is not more
than 2.0 and the relative standard deviation for replicate Specific optical rotation (2.4.22). +103º to +108º, determined
injections is not more than 2.0 per cent in a 10.0 per cent w/v solution.

Inject alternately the test solution and the reference solution. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Calculate the content of C7H6NNaO3. 2H2O in the tablets.
Loss on drying (2.4.19). Not more than 0.25 per cent,
Storage. Store protected from moisture. determined on 1.0 g by drying over phosphorus pentoxide at
a pressure not exceeding 0.7 kPa for 24 hours.
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
Sodium Ascorbate 100 ml of carbon dioxide-free water and 25 ml of 1 M sulphuric
acid. Titrate immediately with 0.05 M iodine, using 1 ml of
starch solution as indicator, until a persistent violet-blue
CH2OH
colour is obtained.
H COH
O 1 ml of 0.05 M iodine is equivalent to 0.009905 g of C6H7NaO6.
O
Storage. Store protected from light.

NaO OH

C6H7NaO6 Mol. Wt. 198.1


Sodium Aurothiomalate
Sodium Ascorbate is L-ascorbic acid, monosodium salt. Sodium Aurothiomalate consists mainly of the disodium salt
of (aurothio) succinic acid.
Sodium Ascorbate contains not less than 99.0 per cent and
Sodium Aurothiomalate contains not less than 44.5 per cent
not more than 101.0 per cent of C6H7NaO6, calculated on the
and not more than 46.0 per cent of Au and not less than
dried basis.
10.8 per cent and not more than 11.5 per cent of Na, both
Description. White or faintly yellow crystals or a crystalline calculated on the dried basis.
powder; odourless or almost odourless. It darkens gradually
Description. A fine, pale yellow powder; odour, slight;
on exposure to light.
hygroscopic.
Identification Identification
Test A may be omitted if tests B, C and D are carried out. Test A. Ignite 0.1 g and dissolve a portion of the residue by warming
B and C may be omitted if tests A and D are carried out. with 2 ml of a mixture of 3 volumes of hydrochloric acid and

1079
SODIUM AUROTHIOMALATE INJECTION IP 2007

1 volume of nitric acid and dilute to 20 ml with water (solution Identification


A). Reserve the remainder of the residue for use in test D. To
0.2 ml add 20 ml of water, boil, pour the boiling solution into A. To a volume equivalent to 20 mg of Sodium Aurothiomalate
5 ml of stannous chloride solution and mix; a purple colour is add 2 ml of hydrogen peroxide solution (100 vol) and 1 ml of
produced. 5 M sodium hydroxide and boil for 30 seconds; a colloidal
precipitate is produced which appears bluish green by
B. To 2 ml of solution A add 2 ml of hydrogen peroxide solution transmitted light.
(20 vol) and 1 ml of 5 M sodium hydroxide; a precipitate is
produced which appears brownish black by reflected light B. To a volume equivalent to 10 mg of Sodium Aurothiomalate
and bluish green by transmitted light. add 0.1 ml of potassium cyanide solution and 0.1 ml of a 1 per
cent w/v solution of sodium nitroprusside; a deep magenta
C. Solution A gives a black precipitate with hydrogen sulphide colour is produced.
which is insoluble in 2 M hydrochloric acid but soluble in
ammonium polysulphide solution. Tests
D. Extract a portion of the residue obtained in test A with 10 ml
Appearance of solution. Dilute, if necessary, with water to
of 2 M hydrochloric acid. The solution, after neutralisation if
give a solution containing 1.0 per cent w/v of Sodium
necessary, gives the reactions of sodium salts and the reaction
Aurothiomalate. The colour of the solution is not more intense
of sulphates (2.3.1).
than that of a 0.02 per cent w/v solution of potassium
Tests ferricyanide.
Other tests. Complies with the tests stated under Parenteral
pH (2.4.24). 6.0 to 7.0, determined in a 10.0 per cent w/v solution.
Preparations (Injections).
Stability. Dissolve 1.0 g in 10 ml of water, filter, seal in an
Assay. To an accurately measured volume containing about
ampoule, heat at 100º for 1 hour, cool and add sufficient water
0.1 g of Sodium Aurothiomalate add 0.4 g of potassium bromide
to produce 100 ml. The solution remains bright and is not
and 5 ml of nitric acid. Slowly evaporate the solution to
more intensely coloured than a 0.01 per cent w/v solution of
dryness and continue heating until fumes cease to be evolved.
potassium ferricyanide.
Allow to cool, add 50 ml of water, warm, filter, wash the residue
Loss on drying (2.4.19). Not more than 2.0 per cent, determined of gold, Au, with hot water, dry and ignite for 3 hours at a
on 1.0 g by drying over phosphorus pentoxide at a pressure temperature not lower than 600º.
not exceeding 0.7 kPa for 24 hours.
Storage. Store protected from light.
Assay. For Au — Weigh accurately about 0.2 g, heat with
10 ml of sulphuric acid and continue to boil gently until a
clear, pale yellow liquid is produced. Cool, add about 1 ml of
nitric acid dropwise and boil again for 1 hour. Cool, dilute Sodium Benzoate
with 70 ml of water, boil for 5 minutes, filter, wash the residue
of gold Au, with hot water, dry and ignite for 3 hours at a COONa
temperature not lower than 600º.
For Na — Evaporate to dryness the filtrate and washings
obtained in the Assay for Au, moisten with sulphuric acid
and ignite for 3 hours at 600º.
1 g of residue is equivalent to 0.3237 g of Na. C7H5NaO2 Mol. Wt. 144.1
Storage. Store protected from light. Sodium Benzoate contains not less than 99.0 per cent and
not more than 100.5 per cent of C7H5NaO2, calculated on the
dried basis.
Description. A white, crystalline or granular powder or flakes;
Sodium Aurothiomalate Injection odourless or with a faint odour; hygroscopic.
Sodium Aurothiomalate Injection is a sterile solution of Sodium
Aurothiomalate in Water for Injection. Identification
Sodium Aurothiomalate Injection contains gold, Au, equivalent A. To a 10 per cent w/v solution add ferric chloride test
to not less than 42.3 per cent and not more than 48.3 per cent solution; a buff coloured precipitate is formed. Add dilute
of the stated amount of sodium aurothiomalate. hydrochloric acid; a white crystalline precipitate is produced.

1080
IP 2007 SODIUM BICARBONATE

B. Gives the reactions of sodium salts and reactions B and C Description. A white, crystalline powder or small, opaque,
of benzoates (2.3.1). monoclinic crystals. It gradually forms sodium carbonate on
heating in the dry state or in solution.
Tests
Identification
Appearance of solution. A 10.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely A. To 5 ml of a 5.0 per cent w/v solution in carbon dioxide-
coloured than reference solution YS6 (2.4.1). free water (solution A) add 0.1 ml of phenolphthalein solution;
a pale pink colour is produced. On heating, a gas is evolved
Acidity or alkalinity. To 20 ml of a 5.0 per cent w/v solution in and the solution becomes red.
carbon dioxide-free water add 0.2 ml of phenolphthalein
solution. Not more than 0.2 ml of 0.1 M hydrochloric acid or B. Gives reaction A of bicarbonates (2.3.1).
0.2 ml of 0.1 M sodium hydroxide is required to change the C. Solution A gives the reactions of sodium salts (2.3.1).
colour of the solution.
Tests
Arsenic (2.3.10). Mix 5.0 g with 10 ml of bromine solution and
evaporate to dryness on a water-bath. Ignite gently, dissolve Appearance of solution. Solution A is clear (2.4.1), and
the cooled residue, ignoring any carbon, in 50 ml of water and colourless (2.4.1).
14 ml of brominated hydrochloric acid AsT, and remove the
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water, add 15 ml of
excess of bromine with 2 ml of stannous Chloride solution
brominated hydrochloric acid AsT and remove the excess of
AsT. The resulting solution complies with the limit test for
bromine with a few drops of stannous chloride solution AsT.
arsenic (2 ppm).
The resulting solution complies with the limit test for arsenic
Heavy metals (2.3.13). Dissolve 2.0 g in 45 ml of water and 5 ml (2 ppm).
of hydrochloric acid. 25 ml of the solution complies with the
Calcium. A 2.0 per cent w/v solution, when boiled for 5 minutes,
limit test for heavy metals, Method B (20 ppm).
is clear.
Chlorinated compounds. Dissolve 0.33 g in 5 ml of 0.5 M Heavy metals (2.3.13). Mix 4.0 g with 5 ml of water and 18 ml of
sodium carbonate, evaporate to dryness and heat the residue dilute hydrochloric acid, heat to boiling and maintain that
until completely charred, keeping the temperature below 400º. temperature for 1 minute. Add 0.05 ml of phenolphthalein
Extract the residue with a mixture of 10 ml of water and 12 ml of solution and sufficient 5 M ammonia dropwise to give the
dilute nitric acid and filter; the filtrate complies with the limit solution a faint pink colour, cool and dilute to 25 ml with water.
test for chlorides (2.3.12). The solution complies with the limit test for heavy metals,
Loss on drying (2.4.19). Not more than 2.0 per cent, determined Method A (5 ppm).
on 1.0 g by drying in an oven at 105º. Iron (2.3.14). Dissolve 2.0 g in 20 ml of water and 4 ml of
Assay. Weigh accurately about 0.25 g, dissolve in 20 ml of hydrochloric acid and dilute to 40 ml with water; the resulting
anhydrous glacial acetic acid, warming to 50º if necessary, solution complies with the limit test for iron (20 ppm).
cool. Titrate with 0.1 M perchloric acid, using 0.05 ml of Carbonate. pH of solution A, when freshly prepared, not more
1-naphtholbenzein solution as indicator. Carry out a blank than 8.6.
titration.
Chlorides (2.3.12). 1.25 g dissolved in 15 ml of water and 2 ml
1 ml of 0.1 M perchloric acid is equivalent to 0.01441 g of of nitric acid complies with the limit test for chlorides
C7H5NaO2. (200 ppm).
Storage. Store protected from light. Sulphates (2.3.17). Suspend 1.0 g in 10 ml of distilled water,
neutralise with hydrochloric acid and dilute to 15 ml with
distilled water. The resulting solution complies with the limit
test for sulphates (150 ppm).
Sodium Bicarbonate Assay. Weigh accurately about 1.5 g, dissolve in 50 ml of
carbon dioxide-free water and titrate with 1 M hydrochloric
Sodium Hydrogen Carbonate acid using 0.2 ml of methyl orange solution as indicator.
NaHCO3 Mol. Wt. 84.0 1 ml of 1 M hydrochloric acid is equivalent to 0.08401 g of
NaHCO3.
Sodium Bicarbonate contains not less than 99.0 per cent and
not more than 101.0 per cent of NaHCO3. Storage. Store protected from moisture.

1081
SODIUM BICARBONATE INJECTION IP 2007

Sodium Bicarbonate Injection Description. Anhydrous — A white or almost white, slightly


granular powder, hygroscopic.
Sodium Bicarbonate Intravenous Infusion
Monohydrate — A white, crystalline powder or colourless
Sodium Bicarbonate Injection is a sterile solution of Sodium crystals.
Bicarbonate in Water for Injections.
Identification
Sodium Bicarbonate Injection contains not less than 94.0 per
cent and not more than 106.0 per cent of the stated amount of A. Dissolve 1 g in water and dilute to 10 ml with water; the
sodium bicarbonate, NaHCO3. solution is strongly alkaline.
Description. A clear, colourless solution. B. The solution prepared for test A gives reaction A of
carbonates and reactions of sodium salts (2.3.1)
Identification
Tests
A. The residue on evaporation, when moistened with
hydrochloric acid and introduced on a platinum wire into a Appearance of solution. Dissolve 2.0 g in 10 ml of water. The
flame, imparts a yellow colour to the flame. resulting solution is clear and not more intensely coloured
B. Gives reaction A of sodium salts and the reactions of than reference solution YS6, (2.4.1).
bicarbonates (2.3.1). Alkali hydroxides and bicarbonates. Dissolve 0.4 g in 20 ml of
water, add 20 ml of barium chloride solution and filter. To
Tests 10 ml of the filtrate add 0.1 ml of phenolphthalein solution.
The solution does not become red. Heat the remainder of the
pH (2.4.24). 7.0 to 8.5.
filtrate to boiling for 2 minutes. The solution remains clear.
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
Heavy metals (2.3.13). Dissolve 2.0 g in portions in a mixture of
per ml.
5 ml of hydrochloric acid and 25 ml of water. Heat the solution
Other tests. Complies with the tests stated under Parenteral to boiling and cool. Add dilute sodium hydroxide solution
Preparations (Injections). until the solution is neutral. Dilute to 50 ml with water (solution
Assay. Titrate a volume containing 1.5 g of Sodium Bicarbonate A). 12 ml of the resulting solution complies with the limit test
with 1 M hydrochloric acid using methyl orange solution as for heavy metals, Method A (50 ppm). Use lead standard
indicator. solution (2 ppm Pb) for the standard.

1 ml of 1 M hydrochloric acid is equivalent to 0.08401 g of Iron (2.3.14). Dilute 5 ml of solution A to 10 ml with water. The
NaHCO3. solution complies with the limit test for iron (50 ppm).

Storage. Store in single dose containers. Chlorides (2.3.12). Dissolve 0.4 g in water, add 4 ml of dilute
nitric acid and dilute to 15 ml with water. The solution complies
Labelling. The label states (1) the strength as the percentage with the limit test for chlorides (125 ppm).
w/v of Sodium Bicarbonate; (2) the approximate
concentrations, in millimoles per litre, of the sodium ions and Sulphates (2.3.17). 15 ml of solution A complies with the limit
the bicarbonate ions; (3) that an injection containing visible test for sulphates, (250 ppm).
particles in the solution should not be used. Loss on drying (2.4.19). Not more than 1.0 per cent (for
anhydrous form) or between 12.0 per cent and 15.0 per cent
(for monohydrate form), determined on 2.0 g by drying in an
oven at 300º.
Sodium Carbonate Assay. Weigh accurately about 1.0 g, dissolve in 25 ml of
Na2CO3 Mol. Wt. 106.0 (anhydrous) water and titrate with 1 M hydrochloric acid using methyl
orange solution as indicator.
Na2CO3, H2O Mol.Wt.124.0 (monohydrate)
1 ml of 1 M hydrochloric acid is equivalent to 0.05299 g of
Sodium Carbonate is anhydrous or contains one molecule
Na2CO3.
of water of hydration.
Storage. Store in tightly-closed, non-metallic containers.
Sodium Carbonate contains not less than 99.5 per cent and
not more than 100.5 per cent of Na2CO3, calculated on the Labelling. The label states whether the material is anhydrous
dried basis. or monohydrate.

1082
IP 2007 SODIUM CHLORIDE

Sodium Chloride Heavy metals (2.3.13). 4.0 g in 2 ml of dilute acetic acid and
sufficient water to produce 25 ml. The solution complies with
NaCl Mol. Wt. 58.4 the limit test for heavy metals, Method A (5 ppm).
Sodium Chloride contains not less than 99.0 per cent and not Iodide. Moisten 5 g by adding dropwise, a solution freshly
more than 100.5 per cent of NaCl, calculated on the dried basis. prepared by mixing 25 ml of iodide-free starch solution 2 ml of
Description. White or colourless crystals or a white crystalline 0.5 M sulphuric acid, 0.15 ml of sodium nitrite solution and
powder. 25 ml of water and examine the mixture in daylight; the
substance shows no blue colour after 5 minutes.
Identification Iron (2.3.14). 2.0 g dissolved in 20 ml of water complies with
A. Gives the reactions of chlorides (2.3.1). the limit test for iron (20 ppm).
Sulphates (2.3.17). 2.5 ml of solution A diluted to 15 ml with
B. A 20 per cent w/v solution in carbon dioxide-free water
water complies with the limit test for sulphates (300 ppm).
prepared from distilled water (solution A) gives the reactions
of sodium salts (2.3.1). Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105º for 3 hours.
Tests
Assay. Weigh accurately about 0.1 g and dissolve in 50 ml of
Appearance of solution. Solution A is clear (2.4.1), and water in a glass-stoppered flask. Add 50.0 ml of 0.1 M silver
colourless (2.4.1). nitrate, 5 ml of 2 M nitric acid and 2 ml of dibutyl phthalate,
shake well and titrate with 0.1 M ammonium thiocyanate using
Acidity or alkalinity. To 20 ml of solution A add 0.1 ml of
2 ml of ferric ammonium sulphate solution as indicator, until
bromothymol blue solution; not more than 0.5 ml of 0.01 M
the colour becomes reddish yellow.
hydrochloric acid or of 0.01 M sodium hydroxide is required
to change the colour of the solution. 1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl.
Arsenic (2.3.10). Dissolve 10.0 g in 50 ml of water and 12 ml of Sodium Chloride intended for use in the manufacture of
stannated hydrochloric acid AsT. The resulting solution parenteral preparations or in the manufacture of dialysis
complies with the limit test for arsenic (1 ppm). solutions complies with the following additional requirement.
Barium. Dissolve 2 g in 10 ml of water, and add 2 ml of dilute Potassium. Not more than 0.1 per cent, determined by flame
sulphuric acid; no turbidity is produced within 2 hours. photometry (2.4.4), using a 1.0 per cent w/v solution and
measuring at 768 nm. Use suitable dilutions in water of
Bromide. To 0.5 ml of solution A add 4.0 ml of water, 2.0 ml of potassium solution FP for the standard solution.
phenol red reagent and 1.0 ml of 0.01 per cent w/v solution of
chloramine T and mix immediately. After exactly 2 minutes, Sodium Chloride intended for use in the preparation of dialysis
add 0.15 ml of 0.1 M sodium thiosulphate, mix and dilute to solutions complies with the following additional requirement.
10.0 ml with water. The absorbance of the solution measured Aluminium. Not more than 0.2 ppm, determined by the
at about 590 nm (2.4.7), using water as the blank, is not more following method. Dissolve 20 g in 100 ml of water and add
than that of the standard solution prepared at the same time 10 ml of acetate buffer pH 6.0. Extract the resulting solution
and in the same manner, using 5.0 ml of a 0.0003 per cent w/v with successive quantities of 20, 20 and 10 ml of a 0.5 per cent
solution of potassium bromide (100 ppm). w/v solution of 8-hydroxyquinoline in chloroform and dilute
Calcium and magnesium. Not more than 50 ppm, calculated the combined extracts to 50 ml with chloroform. Use as the
as Ca, determined by the following method. Dissolve 20.0 g in blank solution a mixture of 10 ml of acetate buffer pH 6.0 and
200 ml of water, and add 0.1 ml of hydrochloric acid, 5 ml of 100 ml of water treated in the same manner and as the standard
strong ammonia-ammonium chloride solution, 5 drops of solution a mixture of 2 ml of aluminium standard solution
eriochrome black T solution and titrate with 0.005 M disodium (2 ppm Al), 10 ml of acetate buffer pH 6.0 and 90 ml of water
edetate to a blue end-point. treated in the same manner. Measure the fluorescence of the
test solution and of the standard solution (2.4.5), using an
1 ml of 0.005 M disodium edetate is equivalent to 0.0002004 g excitation wavelength of 392 nm and a secondary filter with a
of Ca. transmission band centered at 518 nm, or a monochromator
Ferrocyanide. Dissolve 2.0 g in 6 ml of water and add 0.5 ml of set to transmit at this wavelength, and setting the instrument
a mixture of 5 ml of a 1 per cent w/v solution of ferric ammonium to zero with the blank solution in each case. The fluorescence
sulphate in a 0.25 per cent w/v solution of sulphuric acid, and of the test solution is not greater than that of the standard
95 ml of a 1 per cent w/v solution of ferrous sulphate; no blue solution.
colour is produced within 10 minutes. Storage. Store protected from light.

1083
SODIUM CHLORIDE AND DEXTROSE INJECTION IP 2007

Labelling. The label states whether or not the material is multiplied by 0.9477 represents the weight, in g, of dextrose,
suitable for use in the manufacture of parenteral preparations C6H12O6, in the volume of the injection taken for assay.
or for the preparation of dialysis solutions. Storage. Store in single dose containers. On keeping, small
solid particles may separate from a glass container.
Labelling. The label states (1) the strength as the percentages
w/v of Sodium Chloride and Dextrose; (2) that a solution
Sodium Chloride and Dextrose containing visible particles must not be used.
Injection When the preparation is intended for intravenous infusion,
Sodium Chloride and Dextrose Intravenous Infusion; the label states the approximate concentrations, in millimoles
Sodium Chloride and Glucose Injection; Sodium Chloride per litre, of the sodium and chloride ions and the number of
and Glucose Intravenous Infusion grams per litre of dextrose, C6H12O6.

Sodium Chloride and Dextrose Injection is a sterile solution of


Sodium Chloride and Dextrose in Water for Injections.
Sodium Chloride and Dextrose Injection contains not less than
Sodium Chloride and Fructose
95.0 per cent and not more than 105.0 per cent of the stated Injection
amounts of sodium chloride, NaCl, and dextrose, C6H12O6.
Sodium Chloride and Fructose Intravenous Infusion;
Description. A clear, colourless or faintly straw-coloured Sodium Chloride and Fructose Infusion; Fructose and
solution. Sodium Chloride Injection.
Identification Sodium Chloride and Fructose Injection is a sterile solution of
Sodium Chloride and Fructose in Water for Injections.
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution;
Sodium Chloride and Fructose Injection contains not less than
the solution remains blue and clear. Heat to boiling, a copious
95.0 per cent and not more than 105.0 per cent of the stated
red precipitate is formed.
amounts of sodium chloride, NaCl, and fructose, C6H12O6. It
B. Gives reaction B of sodium salts and reaction A of chlorides contains no antimicrobial agent.
(2.3.1).
Description. A clear, colourless or faintly straw-coloured
solution.
Tests
pH (2.4.24). 3.5 to 6.5. Identification
5-Hydroxymethylfurfural and related substances. Dilute a A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution;
volume containing 1.0 g of dextrose, C6H12O6, to 500 ml with the solution remains blue and clear. Heat to boiling, a copious
water and measure the absorbance of the resulting solution red precipitate is formed.
at the maximum at about 284 nm (2.4.7); absorbance at about B. The solution prepared in the Assay is laevo-rotatory.
284 nm, not more than 0.25.
C. Gives reaction B of sodium salts and reaction A of chlorides
Bacterial endotoxins (2.2.3). Not more than 10 Endotoxin Units (2.3.1).
per g of dextrose.
Other tests. Complies with the tests stated under Parenteral Tests
Preparations (Intravenous Infusions).
pH (2.4.24). 3.0 to 6.0.
Assay. For sodium chloride — Titrate an accurately measured
5-Hydroxymethylfurfural and related substances. Dilute a
volume containing 0.1 g of Sodium Chloride with 0.1 M silver
volume containing 1.0 g of Fructose to 500.0 ml with water
nitrate using potassium chromate solution as indicator.
and measure the absorbance of the resulting solution at the
1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl. maximum at about 284 nm (2.4.7); absorbance at about 284 nm,
For dextrose — To an accurately measured volume containing not more than 0.50.
2 to 5 g of anhydrous dextrose, C6H12O6, add 0.2 ml of 5 M Heavy metals (2.3.13). Evaporate a volume containing 4.0 g of
ammonia and sufficient water to produce 100.0 ml. Mix well, Fructose to 10 ml and add 2 ml of dilute acetic acid and
allow to stand for 30 minutes and measure the optical rotation sufficient water to produce 25 ml. The solution complies with
in a 2-dm tube (2.4.22). The observed rotation in degrees the limit test for heavy metals, Method A (5 ppm).

1084
IP 2007 COMPOUND SODIUM CHLORIDE AND DEXTROSE INJECTION

Other tests. Complies with the tests stated under Parenteral Identification
Preparations (Injections).
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution;
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit the solution remains blue and clear. Heat to boiling, a copious
per ml. red precipitate is formed.
Assay. For sodium chloride — Titrate an accurately measured B. Gives reaction B of sodium salts and reaction A of chlorides
volume containing about 0.1 g of Sodium Chloride with 0.1 M (2.3.1).
silver nitrate using potassium chromate solution as indicator.
C. After evaporation to one half of its original volume, the
1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl. solution gives reaction A of potassium salts and reaction B of
calcium salts (2.3.1).
For fructose — To an accurately measured volume containing
about 5.0 g of Fructose, add 0.2 ml of 6 M ammonia and Tests
sufficient water to produce 100.0 ml. Mix well, allow to stand
for 30 minutes and measure the optical rotation in a 2-dm tube pH (2.4.24). 3.5 to 6.5.
(2.4.22). The observed rotation in degrees multiplied by 0.5427 5-Hydroxymethylfurfural and related substances. Dilute a
represents the weight, in g, of fructose, C6H12O6, in the volume volume containing 1.0 g of Dextrose to 500.0 ml with water
taken for assay. and measure the absorbance of the resulting solution at the
Storage. Store in single dose containers. On keeping, small maximum at about 284 nm; absorbance at about 284 nm, not
solid particles may separate from glass containers. more than 0.25 (2.4.7).

Labelling. The label states (1) the strength as the percentages Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
w/v of Sodium Chloride and Fructose; (2) when the preparation per ml.
is intended for intravenous infusion, the approximate Other tests. Complies with the tests stated under Parenteral
concentrations, in millimoles per litre, of the sodium and Preparations (Injections).
chloride ions and the number of grams per litre of fructose; (3)
Assay. For sodium chloride — Dilute suitably with water and
that a solution containing visible particles should not be used.
determine by Method A for flame photometry (2.4.4), or by
Method A for atomic absorption spectrophotometry (2.4.2),
measuring at 589 nm and using sodium solution FP or sodium
solution AAS respectively, suitably diluted with water for the
Compound Sodium Chloride and standard solutions.
Dextrose Injection 1 g of Na is equivalent to 2.542 g of NaCl.
Compound Sodium Chloride and Dextrose Intravenous For potassium chloride — Dilute suitably with water and
Infusion determine by Method A for flame photometry (2.4.4), or by
Method A for atomic absorption spectrophotometry (2.4.2),
Compound Sodium Chloride and Dextrose Injection is a sterile measuring at 767 nm and using potassium solution FP or
solution containing 0.86 per cent w/v of Sodium Chloride, potassium solution AAS respectively, suitably diluted with
0.03 per cent w/v of Potassium Chloride, 0.033 per cent w/v of water for the standard solutions.
Calcium Chloride and 5 per cent w/v of Dextrose in Water for
Injections. 1 g of K is equivalent to 1.907 g of KCl.

Compound Sodium Chloride and Dextrose Injection contains For calcium chloride — To 50.0 ml add 5.0 ml of 0.01 M
not less than 0.82 per cent and not more than 0.90 per cent magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and
w/v of sodium chloride, NaCl, not less than 0.0285 per cent titrate with 0.01 M disodium edetate using eriochrome black
and not more than 0.0315 per cent w/v of potassium chloride, T mixture as indicator. From the volume of 0.01 M disodium
KCl, not less than 0.030 per cent and not more than 0.036 per edetate required subtract the volume of 0.01 M magnesium
cent w/v of calcium chloride, CaCl2,2H2O, and not less than sulphate added.
0.523 per cent and not more than 0.580 per cent w/v of chloride, 1 ml of the remainder of 0.01 M disodium edetate is equivalent
Cl, and not less than 4.75 per cent and not more than 5.25 per to 0.00147 g of CaCl2, 2H2O.
cent w/v of dextrose, C6H12O6. It contains no antimicrobial
For total chloride — To 20.0 ml add 30 ml of water, 50.0 ml of
agent.
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the
Description. A clear, colourless or faintly straw-coloured precipitate with water slightly acidified with nitric acid and
solution. titrate the excess of silver nitrate with 0.1 M ammonium

1085
SODIUM CHLORIDE HYPERTONIC INJECTION IP 2007

thiocyanate using ferric ammonium sulphate solution as Other tests. Complies with the tests stated under Parenteral
indicator until a reddish yellow colour is produced. Carry out Preparations (Intravenous Infusions).
a blank titration. Assay. To an accurately measured volume containing about
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total 0.16 g of Sodium Chloride in a glass-stoppered flask add 50 ml
Chloride, calculated as Cl. of water and 50.0 ml of 0.1 M silver nitrate, 5 ml of 2 M nitric
acid and 2 ml of dibutyl phthalate. Shake well and titrate with
For dextrose — To an accurately measured volume containing
0.1 M ammonium thiocyanate using 2 ml of ferric ammonium
2 g to 5 g of Dextrose, add 0.2 ml of 5 M ammonia and sufficient
sulphate solution as indicator, until the colour becomes reddish
water to produce 100.0 ml. Mix well, allow to stand for
yellow.
30 minutes and measure the optical rotation in a 2-dm tube
(2.4.22). The observed rotation in degrees multiplied by 0.9477 1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl.
represents the weight, in g, of dextrose, C6H12O6, in the volume Storage. Store in single dose containers of glass or plastic.
taken for assay. On keeping, small solid particles may separate from a glass
Storage. Store in single dose containers of glass or plastic at container.
a temperature not exceeding 30º. On keeping, small solid Labelling. The label states (1) the strength as the percentage
particles may separate from the solution in glass containers. w/v of Sodium Chloride; (2) that a solution containing visible
Labelling. The label states (1) the strength as the percentages solid particles must not be used.
w/v of Sodium Chloride, Potassium Chloride, Calcium Chloride When the preparation is intended for intravenous infusion,
and Dextrose; (2) that the injection contains, in millimoles per the label states that the injection contains approximately
litre, the following approximate amounts of the ions. sodium, 270 millimoles each of sodium and chloride ions per litre.
147.5, potassium, 4, calcium, 4.5, and Chloride, 156; (3) the
total osmolar concentration in mOsmol per litre; (4) that the
injection should not be used if it contains visible particles.
Sodium Chloride Injection
Sodium Chloride Intravenous Infusion
Sodium Chloride Hypertonic Injection Sodium Chloride Injection is a sterile 0.9 per cent w/v solution
of Sodium Chloride in Water for Injections. It contains no
Hypertonic Saline
antimicrobial agent.
Sodium Chloride Hypertonic Injection is a sterile 1.6 per cent
Sodium Chloride Injection contains not less than 0.85 per cent
w/v solution of Sodium Chloride in Water for Injections. It
w/v and not more than 0.95 per cent w/v of sodium Chloride,
contains no antimicrobial agent.
NaCl.
Sodium Chloride Hypertonic Injection contains not less than
Description. A clear, colourless solution.
1.52 per cent w/v and not more than 1.68 per cent w/v of
sodium chloride, NaCl. Identification
Description. A clear, colourless solution.
Gives the reactions of sodium salts and reaction A of chlorides
Identification (2.3.1).
Gives the reactions of sodium salts and reaction A of chlorides
Tests
(2.3.1).
pH (2.4.24). 4.5 to 7.0.
Tests
Heavy metals (2.3.13). Evaporate 67 ml to about 20 ml, add 2 ml
pH (2.4.24). 5.0 to 7.5. of dilute acetic acid and dilute to 25 ml with water. The solution
Heavy metals (2.3.13). Evaporate 40 ml to about 20 ml, add 2 ml complies with the limit test for heavy metals, Method A
of dilute acetic acid and dilute to 25 ml with water. The solution ( 0.3 ppm).
complies with the limit test for heavy metals, Method A Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
(0.5 ppm). per ml.
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit Other tests. Complies with the tests stated under Parenteral
per ml. Preparations (Intravenous Infusions).

1086
IP 2007 COMPOUND SODIUM CHLORIDE SOLUTION

Assay. To an accurately measured volume containing about Other tests. Complies with the tests stated under Parenteral
0.16 g of Sodium Chloride in a glass-stoppered flask add 50 ml Preparations (Intravenous Infusions).
of water and 50.0 ml of 0.1 M silver nitrate, 5 ml of 2 M nitric Assay. For sodium chloride — Dilute appropriately with water
acid and 2 ml of dibutyl phthalate. Shake well and titrate with and determine by Method A for flame photometry (2.4.4),
0.1 M ammonium thiocyanate using 2 ml of ferric ammonium measuring at 589 nm or by Method A for atomic absorption
sulphate solution as indicator, until the colour becomes reddish spectrophotometry (2.4.2), using sodium solution FP, suitably
yellow. diluted with water for the standard solutions.
1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl.
1 g of Na is equivalent to 2.54 g of NaCl.
Storage. Store in single dose containers of glass or plastic.
For potassium Chloride — Dilute appropriately with water
On keeping, small solid particles may separate from a glass
and determine by Method A for flame photometry (2.4.4),
container.
measuring at 767 nm or by Method A for atomic absorption
Labelling. The label states (1) the strength as the percentage spectrophotometry (2.4.2), using potassium solution FP,
w/v of Sodium Chloride; (2) that a solution containing visible suitably diluted with water for the standard solutions.
solid particles must not be used.
1 g of K is equivalent to 1.007 g of KCl.
When the preparation is intended for intravenous infusion,
For Calcim Chloride — To 50.0 ml add 5.0 ml of 0.01 M
the label states that the injection contains approximately
magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and
150 millimoles each of sodium and chloride ions per litre.
titrate with 0.01M disodium edetate using mordant black II
mixture as indicator. From the volume of 0.01 M disodium
edetate required subtract the volume of 0.01M magnesium
Compound Sodium Chloride Injection sulphate added.
Ringer’s Injection 1 ml of the remainder of 0.01 M disodium edetate is equivalent
Compound Sodium Chloride Injection is a sterile solution to 0.00147 g of CaCl2, 2H2O.
containing 0.86 per cent w/v of Sodium Chloride, 0.03 per cent Storage. Store in single dose containers of glass or plastic.
w/v of Potassium Chloride and 0.033 per cent w/v of Calcium On keeping, small solid particles may separate from a glass
Chloride in Water for Injections. It contains no antimicrobial container.
agent.
Labelling. The label states (1) the strength as the percentages
Compound Sodium Chloride Injection contains not less than w/v of Sodium Chloride, Potassium Chloride and Calcium
0.82 per cent w/v and not more than 0.90 per cent w/v of Chloride; (2) that a solution containing visible solid particles
sodium chloride, NaCl, not less than 0.0285 per cent w/v and must not be used.
not more than 0.0315 per cent w/v of potassium chloride, KCl,
and not less than 0.030 per cent w/v and not more than When the preparation is intended for intravenous infusion,
0.036 per cent w/v of calcium chloride, CaCl2, 2H2O. the label states that the Injection contains in millimoles per
litre, the following approximate amounts of ions; sodium, 147.5;
Description. A clear, colourless solution. potassium, 4; calcium, 2.25 and chloride, 156.
Identification
Gives reaction B of sodium salts and reaction A of chlorides
(2.3.1). When concentrated to one half of its original volume,
Compound Sodium Chloride Solution
it gives reaction A of potassium salts and reaction B of calcium Ringer’s Solution
salts (2.3.1).
Compound Sodium Chloride Solution is a solution containing
Tests 0.86 per cent w/v of Sodium Chloride, 0.03 per cent w/v of
Potassium Chloride and 0.033 per cent w/v of Calcium Chloride
pH (2.4.24). 5.0 to 7.5. in Purified Water.The solution may be clarified by filtration.
Heavy metals (2.3.13). Evaporate 67 ml to about 20 ml, add 2 ml Compound Sodium Chloride Solution contains not less than
of dilute acetic acid and dilute to 25 ml with water. The solution 0.82 per cent w/v and not more than 0.90 per cent of sodium
complies with the limit test for heavy metals, Method A chloride, NaCl, not less than 0.025 per cent and not more than
( 0.3 ppm). 0.035 per cent w/v of potassium chloride, KCl, and not less
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin than 0.030 per cent and not more than 0.036 per cent w/v of
Unit per ml. calcium chloride, CaCl2, 2H2O.

1087
SODIUM CHLORIDE IRRIGATION SOLUTION IP 2007

Description. A clear, colourless solution. Sodium Chloride Irrigation Solution contains not less than
0.85 per cent w/v and not more than 0.95 per cent w/v of
Identification sodium Chloride, NaCl. It contains no antimicrobial agent.
Gives reaction B of sodium salts and reaction A of chlorides Description. A clear, colourless solution.
(2.3.1). When concentrated to one half of its original volume,
it gives reaction A of potassium salts and reaction B of calcium Identification
salts (2.3.1).
Gives the reactions of sodium salts and reaction A of chlorides
Tests (2.3.1).

pH (2.4.24). 5.0 to 7.5. Tests


Heavy metals (2.3.13). Evaporate 67 ml to about 20 ml, add 2 ml pH (2.4.24). 4.5 to 7.0.
of dilute acetic acid and dilute to 25 ml with water. The solution
complies with the limit test for heavy metals, Method A Heavy metals (2.3.13). Evaporate 67 ml to about 20 ml, add 2 ml
( 0.3 ppm). of dilute acetic acid and dilute to 25 ml with water. The solution
complies with limit test for heavy metals, Method A ( 0.3 ppm).
Assay. For sodium chloride — Dilute appropriately with water
and determine by Method A for flame photometry (2.4.4), Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
measuring at 589 nm or by Method A for atomic absorption per ml.
spectrophotometry (2.4.2), using sodium solution FP, suitably Other tests. Complies with the tests stated under Parenteral
diluted with water for the standard solutions. Preparations (Injections).
1 g of Na is equivalent to 2.54 g of NaCl. Assay. Titrate an accurately measured volume containing
For potassium chloride — Dilute appropriately with water about 0.135 g of Sodium Chloride with 0.1 M silver nitrate
and determine by Method A for flame photometry (2.4.4), using potassium chromate solution as indicator.
measuring at 767 nm or by Method A for atomic absorption 1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl.
spectrophotometry (2.4.2), using potassium solution FP,
Storage. Store in single dose containers. The container may
suitably diluted with water for the standard solutions.
be designed to empty rapidly and may contain a volume of
1 g of K is equivalent to 1.007 g of KCl. more than 1 litre.
For calcium Chloride — To 50.0 ml add 5.0 ml of 0.01 M Labelling. The label states (1) the strength as the percentage
magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and w/v of Sodium Chloride; (2) that the solution should not be
titrate with 0.01 M disodium edetate using mordant black II used if it contains visible particles; (3) ‘For Irrigation only’
mixture as indicator. From the volume of 0.01 M disodium and ‘Not for Injection’; (4) that once the container is opened,
edetate required subtract the volume of 0.01M magnesium the unused portion should be discarded.
sulphate added.
1 ml of the remainder of 0.01 M disodium edetate is equivalent
to 0.00147 g of CaCl2, 2H2O.
Sodium Citrate
Sterility (2.2.11). Complies with the test for sterility.
Trisodium Citrate
Storage. Store protected from light.
Labelling. The label states the strength as the percentages HO COONa
,2H2O
w/v of Sodium Chloride, Potassium Chloride and Calcium NaOOC COONa
Chloride. If the contents of the container are sterile, the label
states (1) Sterile Compound Sodium Chloride Solution; (2) C6H5Na3O7,2H2O Mol. Wt. 294.1
that the solution is not intended for injection. Sodium Citrate is trisodium 2-hydroxypropane-1,2,3-
tricarboxylate dihydrate.
Sodium Citrate contains not less than 99.0 per cent and not
Sodium Chloride Irrigation Solution more than 101.0 per cent of C6H5Na3O7, calculated on the
anhydrous basis.
Sodium Chloride Irrigation Solution is a sterile solution
containing 0.9 per cent w/v of Sodium Chloride in Water for Description. White, granular crystals or a white, crystalline
Injections. powder; odourless; slightly deliquescent in moist air.

1088
IP 2007 SODIUM CROMOGLYCATE

Identification to cool. Titrate with 0.1 M perchloric acid, using 0.25 ml of


1-naphtholbenzein solution as indicator. Carry out a blank
A. 10.0 per cent w/v solution in carbon dioxide-free water titration.
(solution A) gives the reactions of sodium salts and reaction
A of citrates (2.3.1). 1 ml of 0.1 M perchloric acid is equivalent to 0.008602 g of
C6H5Na3O7.
Tests Storage. Store protected from light.
Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Acidity or alkalinity. Titrate 20 ml of solution A with 0.05 M Sodium Cromoglycate
sulphuric acid or 0.1 M sodium hydroxide using thymol blue Sodium Cromoglicate
solution as indicator; not more than 0.5 ml of 0.05 M sulphuric
acid or 0.1 M sodium hydroxide is required. NaOOC O O COONa
OH
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 15 ml
of stannated hydrochloric acid AsT. The resulting solution O O O O
complies with the limit test for arsenic (2 ppm).
Heavy metals (2.3.13). Dissolve 2.0 g in 10 ml of water, 5 ml of
dilute hydrochloric acid and sufficient water to produce C23H14Na2O11 Mol. Wt. 512.3
25 ml. The solution complies with the limit test for heavy metals, Sodium Cromoglycate is disodium 4,4′-dioxo-5,5′-(2-
Method A ( 10 ppm). hydroxytrimethylenedioxy)di(chromene-2-carboxylate).
Chlorides (2.3.12). 25 ml of solution A complies with the limit Sodium Cromoglycate contains not less than 98.0 per cent
test for chlorides (100 ppm). and not more than 101.0 per cent of C23H14Na2O11, calculated
Oxalate. Dissolve 0.5 g in 4 ml of water, add 3 ml of hydrochloric on the dried basis.
acid and 1 g of zinc, in granules, and heat on a water-bath for
Description. A white, crystalline powder; odourless;
1 minute. Allow to stand for 2 minutes, decant the liquid into a
hygroscopic.
test-tube containing 0.25 ml of a 1 per cent w/v solution of
phenylhydrazine hydrochloride and heat to boiling. Cool Identification
rapidly, transfer to a graduated cylinder and add an equal
volume of hydrochloric acid and 0.25 ml of potassium Test A may be omitted if tests B, C and D are carried out. Tests
ferrocyanide solution. Shake and allow to stand for 30 minutes. B and C may be omitted if tests A and D are carried out.
Any pink colour produced is not more intense than that A. Determine by infrared absorption spectrophotometry (2.4.6).
obtained by treating at the same time and in the same manner Compare the spectrum with that obtained with sodium
4 ml of a 0.005 per cent w/v solution of oxalic acid (300 ppm, cromoglycate RS or with the reference spectrum of sodium
calculated as anhydrous oxalic acid). cromoglycate.
Sulphates (2.3.17). To 10 ml of solution A add 2 ml of 7 M B. When examined in the range 230 nm to 360 nm (2.4.7), a
hydrochloric acid and dilute to 15 ml with distilled water; the 0.001 per cent w/v solution in phosphate buffer pH 7.4 shows
resulting solution complies with the limit test for sulphates absorption maxima at about 239 nm and 327 nm. The ratio of
(150 ppm). the absorbance at 327 nm to that at about 239 nm, 0.25 to 0.30.
Tartrates. To a solution of 1 g in 2 ml of water in a test-tube, C. Dissolve about 5 mg in 0.5 ml of methanol, add 3 ml of a
add 1 ml of a 10 per cent w/v solution of potassium acetate solution in methanol containing 0.5 per cent w/v of
and 1 ml of 6 M acetic acid. Scratch the walls of the test-tube 4-aminophenazone and 1 per cent v/v of hydrochloric acid
with a glass rod; no crystalline precipitate is formed. and allow to stand for 5 minutes; an intense yellow colour is
Readily carbonisable substances. Heat 0.2 g, in powder, with produced.
10 ml of sulphuric acid (96 per cent w/w) in a water-bath at D. Gives reaction A of sodium salts (2.3.1).
90º ± 1º for 1 hour and cool rapidly. The solution is not more
intensely coloured than reference solution YS2 or GYS2 (2.4.1). Tests
Water (2.3.43). 11.0 to 13.0 per cent, determined on 0.3 g and Appearance of solution. A 2.0 per cent w/v solution in carbon
after stirring for 15 minutes before titrating. dioxide-free water is not more opalescent than opalescence
Assay. Weigh accurately about 0.15 g and dissolve in 20 ml of standard OS2 (2.4.1), and not more intensely coloured than
anhydrous glacial acetic acid, warming to about 50º. Allow reference solution BYS5 (2.4.1).

1089
SODIUM CROMOGLYCATE POWDER FOR INHALATION IP 2007

Acidity or alkalinity. To 10 ml of a 2.0 per cent w/v solution in appropriately treated or Sodium Cromoglycate admixed with
carbon dioxide-free water add 0.1 ml of phenolphthalein an approximately equal amount of Lactose. The contents of
solution; the solution is colourless. Add 0.2 ml of 0.01 M the capsules are in powder of a suitable fineness.
sodium hydroxide; the solution is pink. Add 0.4 ml of 0.01 M Sodium Cromoglycate Powder for Inhalation contain not less
hydrochloric acid; the solution is colourless. Add 0.25 ml of than 100.0 per cent and not more than 120.0 per cent of the
methyl red solution; the solution is red. stated amount of sodium cromoglycate, C23H14Na2O11.
Related substances. Determine by thin-layer chromatography
Description. A white powder; hygroscopic.
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 50 volumes of ethyl acetate, Identification
50 volumes of toluene and 5 volumes of glacial acetic acid.
A. Dissolve a suitable quantity in sufficient phosphate buffer
Test solution. A 2per cent w/v solution of the substance under
pH 7.4 to produce a solution containing 0.001 per cent w/v of
examination in a mixture of 6 volumes of water, 4 volumes of
Sodium Cromoglycate.
tetrahydrofuran and 1 volume of acetone.
When examined in the range 230 nm to 360 nm, (2.4.7), the
Reference solution. A 0.01 per cent w/v of 1,3-bis(2-acetyl-3-
resulting solution shows absorption maxima at about 239 nm
hydroxyphenoxy)-2-propanol RS in chloroform.
and 327 nm.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. B. To a quantity containing 0.1 g of Sodium Cromoglycate
Any secondary spot in the chromatogram obtained with the add 2 ml of water and 2 ml of 1.25 M sodium hydroxide and
test solution is not more intense than the spot in the boil for 1 minute; a yellow colour is produced. Add 0.5 ml of
chromatogram obtained with the reference solution. The diazobenzenesulphonic acid solution; a blood-red colour is
principal spot does not move from the line of application. produced.

Heavy metals (2.3.13). 2.0 g complies with the limit test for C. Gives reaction A of sodium salts (2.3.1).
heavy metals, Method A (10 ppm). D. For capsules containing Lactose, the contents on heating
Oxalate. Dissolve 0.1 g in 20 ml of water, add 5.0 ml of iron with potassium cupri-tartrate solution give a copious red
salicylate solution and sufficient water to produce 50 ml. precipitate.
Measure the absorbance of the resulting solution at the
maximum at about 480 nm (2.4.7). The absorbance is not less Tests
than that obtained by repeating the operation using 0.35 mg Related substances. Determine by thin-layer chromatography
of oxalic acid in place of the substance under examination. (2.4.17), coating the plate with silica gel GF254.
Loss on drying (2.4.19). Not more than 10.0 per cent,
Mobile phase. A mixture of 50 volumes of ethyl acetate,
determined on 0.5 g by drying over phosphorus pentoxide at
50 volumes of toluene and 5 volumes of glacial acetic acid.
105º at a pressure of 0.3 kPa to 0.6 kPa.
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of Test solution. Dissolve a quantity containing 0.1 g of Sodium
25 ml of ethane-1,2-diol and 5 ml of 2-propanol with the aid Cromoglycate in sufficient of a mixture of 6 volumes of water,
of heat and cool, add 30 ml of dioxan. Titrate with 0.1 M 4 volumes of tetrahydrofuran that has been freed from
perchloric acid, determining the end-point potentiometrically stabiliser by passage through a column of suitable alumina
(2.4.25). Carry out a blank titration. and 1 volume of acetone to produce 5 ml and filter.
1 ml of 0.1 M perchloric acid is equivalent to 0.02562 g of Reference solution. Dilute 1 volume of the test solution to
C23H14Na2O11. 200 volumes with the same solvent.
Storage. Store protected from moisture. Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the
Sodium Cromoglycate Powder for test solution is not more intense than the spot in the
Inhalation chromatogram obtained with the reference solution. The
principal spot does not move from the line of application.
Sodium Cromoglicate Powder for Inhalation; Sodium
Uniformity of weight. Weigh one capsule. Open without loss
Cromoglycate Insufflation of shell material, remove the contents and weigh all parts of
Sodium Cromoglycate Powder for Inhalation consist of hard the shell; the difference between the weights represents the
gelatin capsules containing either Sodium Cromoglycate weight of the contents. Repeat the operation with a further 19

1090
IP 2007 SODIUM DIATRIZOATE

capsules and calculate the average weight of the contents of Identification


the 20 capsules.
Tests A and D may be omitted if tests B, C, E and F are carried
For preparations containing no Lactose the weight of the out. Tests B, C and F may be omitted if tests A, D and E are
contents of each capsule does not deviate from the average carried out.
weight by more than 25 per cent. For preparations containing
Lactose, the weight of the contents of each capsule does not A. Determine by infrared absorption spectrophotometry (2.4.6).
deviate from the average weight by more than 15 per cent Compare the spectrum with that obtained with sodium
except that, for two capsules, the weight of the contents may diatrizoate RS or with the reference spectrum of sodium
deviate by more than 25 per cent. diatrizoate.

Loss on drying (2.4.19). 9.0 to 18.0 per cent for capsules B. Determine by thin-layer chromatography (2.4.17), coating
containing no Lactose and 5.5 to 10.0 per cent for capsules the plate with silica gel GF254.
containing Lactose, determined on 0.5 g by drying in an oven Mobile phase. A mixture of 20 volumes of chloroform,
at 105º at a pressure not exceeding 0.7 kPa. 10 volumes of methanol and 2 volumes of strong ammonia
Assay. Weigh accurately a quantity of the mixed contents of solution.
20 capsules containing about 0.1 g of Sodium Cromoglycate, Test solution. Dissolve 0.1 g of the substance under
dissolve in sufficient phosphate buffer pH 7.4 to produce examination in 100 ml of 0.08 per cent w/v solution of sodium
200.0 ml. Dilute 5.0 ml to 100.0 ml with phosphate buffer pH 7.4 hydroxide in methanol.
and measure the absorbance of the resulting solution at 327
nm (2.4.7). Reference solution. A 0.1 per cent w/v of sodium diatrizoate
RS in 0.08 per cent w/v solution of sodium hydroxide in
Calculate the content of C23H14Na2O11 in a capsule of average methanol.
content weight taking 164 as the specific absorbance at
327 nm. Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
Storage. Store protected from light and moisture at a The principal spot in the chromatogram obtained with the test
temperature not exceeding 30º. solution corresponds to that in the chromatogram obtained
Labelling. The label states (1) the strength in terms of the with the reference solution.
equivalent amount of Sodium Cromoglycate; (2) that the C. Heat 0.5 g in a crucible; violet vapours of iodine are evolved.
capsules are intended for use in an inhaler and are not to be
swallowed; (3) where applicable, that the capsules contain D. To 20 mg add 5 ml of 1 M sodium hydroxide and boil gently
Lactose. under a reflux condenser for 10 minutes. Cool, add 5 ml of 2 M
hydrochloric acid and cool in ice for 5 minutes. Add 4 ml of a
1 per cent w/v solution of sodium nitrite, cool in ice for
5 minutes, add 0.3 g of sulphamic acid, shake gently until
Sodium Diatrizoate effervescence ceases and add 2 ml of a 0.4 per cent w/v solution
of N-(1-naphthyl) ethylenediamine dihydrochloride; an
Diatrizoate Sodium
orange-red colour is produced.
COONa E. Heat 0.5 g with 1 ml of sulphuric acid on a water-bath until
I I a pale violet solution is produced, add 2 ml of ethanol (95 per
O O cent) and heat again; odour of ethyl acetate is produced.

H3C N N CH3 F. Gives the reactions of sodium salts (2.3.1).


H I H
Tests
C11H8I3N2NaO4 Mol. Wt. 635.9 pH (2.4.24). 7.5 to 9.5, determined in a 50 per cent w/v solution.
Sodium Diatrizoate is sodium 3,5-diacetamido-2,4,6- Free amine. Place 1.0 g in a 50-ml glass-stoppered volumetric
triiodobenzoate. flask, add 5 ml of water, 10 ml of 0.1 M sodium hydroxide and
25 ml of dimethyl sulphoxide. Stopper the flask, mix the
Sodium Diatrizoate contains not less than 98.0 per cent and
contents gently and cool in ice, protected from light. After
not more than 101.0 per cent of C11H8I3N2NaO4, calculated on
5 minutes, slowly add 2 ml of hydrochloric acid, mix and allow
the anhydrous basis.
to stand for 5 minutes. Add 2 ml of a 2 per cent w/v solution of
Description. A white powder; odourless or almost odourless. sodium nitrite, mix and allow to stand for 5 minutes. Add 1 ml

1091
SODIUM DIATRIZOATE INJECTION IP 2007

of an 8 per cent w/v solution of sulphamic acid, mix and allow amounts of suitable buffers, stabilisers and antimicrobial agents
to stand for 5 minutes. Add 2 ml of a 0.1 per cent w/v solution but the preparation intended for intravenous administration
of N-(1-naphthyl)ethylenediamine dihydrochloride in a 70 contains no antimicrobial preservative.
per cent w/v solution of 1,2-propanediol and mix. Remove Sodium Diatrizoate Injection contains not less than 95.0 per
the flask from the ice and allow to stand in water at 25º ± 2º for cent and not more than 105.0 per cent of the stated amount of
10 minutes, with occasional shaking. Add sufficient dimethyl sodium diatrizoate, C11H8I3N2NaO4.
sulphoxide to produce 50 ml and mix. Within 5 minutes, measure
the absorbance of the resulting solution at the maximum at Identification
about 470 nm (2.4.7), using as the blank a solution prepared
by treating 5 ml of water in the same manner; absorbance, not Evaporate a volume of the injection containing 1 g of Sodium
more than 0.40, calculated on the anhydrous basis. Diatrizoate to dryness. The residue complies with following
tests.
Free iodine and iodide. Dissolve 2.0 g in 24 ml of water taken
in a 50-ml glass-stoppered centrifuge tube. Add 5 ml of toluene A. Heat 0.5 g of residue in a crucible; violet vapours of iodine
and 5 ml of 1 M sulphuric acid, shake well and centrifuge; no are evolved.
red colour appears in the toluene layer. To the mixture add 1 ml B. To 20 mg of the residue add 5 ml of 1 M sodium hydroxide
of a 2 per cent w/v solution of sodium nitrite, shake and and boil gently under a reflux condenser for 10 minutes. Cool,
centrifuge; any red colour in the toluene layer is not more add 5 ml of 2 M hydrochloric acid and cool in ice for 5 minutes.
intense than that produced in a solution prepared in the same Add 4 ml of a 1 per cent w/v solution of sodium nitrite, cool in
manner using 2.0 ml of a 0.025 per cent w/v solution of ice for 5 minutes, add 0.3 g of sulphamic acid, shake gently
potassium iodide and 22 ml of water (220 ppm). until effervescence ceases and add 2 ml of a 0.4 per cent w/v
Heavy metals. Dissolve 1.0 g in 20.0 ml of water and 5 ml of solution of N-(1-naphthyl) ethylenediamine dihydrochloride;
1 M sodium hydroxide, transfer the solution to a 50-ml Nessler an orange-red colour is produced.
cylinder, dilute with water to 40 ml and mix. Add 10 ml of C. Heat 0.5 g of residue with 1 ml of sulphuric acid on a water-
sodium sulphide solution, shake and allow to stand for bath until a pale violet solution is produced, add 2 ml of ethanol
5 minutes (20 ppm), the colour of the solution when viewed (95 per cent) and heat again; odour of ethyl acetate is
downward over a white surface is not more intense than that produced.
produced by treating 2.0 ml of lead standard solution
(10 ppm Pb) in the same manner in place of the substance Tests
under examination.
pH (2.4.24). 6.6 to 7.6.
Water (2.3.43). 4.0 to 7.0 per cent, determined on 0.4 g.
Free amine. To a volume containing 1.0 g of Sodium Diatrizoate
Assay. Weigh accurately about 0.4 g in a glass-stoppered in a 50-ml glass-stoppered volumetric flask, add 5 ml of water,
conical flask, add 12 ml of 5 M sodium hydroxide and 1 g of 10 ml of 0.1 M sodium hydroxide and 25 ml of dimethyl
zinc powder and boil under a reflux condenser for 30 minutes. sulphoxide. Stopper the flask, mix the contents gently and
Cool, rinse the condenser with 30 ml of water, filter through cool in ice, protected from light. After 5 minutes, slowly add
cotton and wash the flask and filter with two quantities, each 2 ml of hydrochloric acid, mix and allow to stand for 5 minutes.
of 20 ml, of water. To the combined filtrate and washings add Add 2 ml of a 2 per cent w/v solution of sodium nitrite, mix
80 ml of hydrochloric acid, cool and titrate with 0.05 M and allow to stand for 5 minutes. Add 1 ml of an 8 per cent
potassium iodate until the dark brown colour becomes pale w/v solution of sulphamic acid, mix and allow to stand for
brown. Add 5 ml of chloroform and continue the titration, 5 minutes. Add 2 ml of a 0.1 per cent w/v solution of N-(1-
shaking well after each addition, until the chloroform becomes naphthyl)ethylenediamine dihydrochloride in a 70 per cent
colourless. w/v solution of 1,2-propanediol and mix. Remove the flask
1 ml of 0.05 M potassium iodate is equivalent to 0.02120 g of from the ice and allow to stand in water at 25º ± 2º for
C11H8I3N2NaO4. 10 minutes, with occasional shaking. Add sufficient dimethyl
sulphoxide to produce 50 ml and mix. Within 5 minutes, measure
Storage. Store protected from light.
the absorbance of the resulting solution at the maximum at
about 470 nm (2.4.7), using as the blank a solution prepared
Sodium Diatrizoate Injection by treating 5 ml of water in the same manner; absorbance, not
more than 0.30, calculated on the anhydrous basis.
Diatrizoate Sodium Injection Free iodine and iodide. To a volume containing 2.0 g of Sodium
Sodium Diatrizoate Injection is a sterile solution of Sodium Diatrizoate in 24 ml of water taken in a 50-ml glass-stoppered
Diatrizoate in Water for Injections. It may contain small centrifuge tube add 5 ml of toluene and 5 ml of 1 M sulphuric

1092
IP 2007 SODIUM DIHYDROGEN PHOSPHATE DIHYDRATE

acid, shake well and centrifuge; no red colour appears in the C. Solution A gives the reactions of phosphates (2.3.1).
toluene layer. To the mixture add 1 ml of a 2 per cent w/v
solution of sodium nitrite, shake and centrifuge; any red colour Tests
in the toluene layer is not more intense than that produced in
a solution prepared in the same manner using 2.0 ml of a Appearance of solution. Solution A is clear (2.4.1), and
0.025 per cent w/v solution of potassium iodide and 22 ml of colourless (2.4.1).
water (220 ppm). pH (2.4.24). 4.2 to 4.5, determined in a mixture of 5 ml of solution
Pyrogens (2.2.8). Complies with test for pyrogens, using per A and 5 ml of carbon dioxide-free water.
kg of the rabbit’s weight a volume containing 2.5 g of Sodium Arsenic (2.3.10). Dissolve 0.5 g in 50 ml of water and add 10 ml
Diatrizoate. of stannated hydrochloric acid AsT. The resulting solution
Other tests. Complies with the tests stated under Parenteral complies with the limit test for arsenic (2 ppm).
Preparations (Injections). Heavy metals (2.3.13).12 ml of solution A complies with the
Assay. Weigh accurately about 0.5 g in a glass-stoppered limit test for heavy metals, Method D (10 ppm). Use lead
conical flask, add 12 ml of 5 M sodium hydroxide and 1 g of standard solution (1 ppm Pb) to prepare the standard.
zinc powder and boil under a reflux condenser for 30 minutes. Iron (2.3.14). 20 ml of solution A complies with the limit test for
Cool, rinse the condenser with 30 ml of water, filter through iron (20 ppm).
cotton and wash the flask and filter with two quantities, each
of 20 ml, of water. To the combined filtrate and washings add Chlorides (2.3.12). 10 ml of solution A diluted to 20 ml with
80 ml of hydrochloric acid, cool and titrate with 0.05 M water complies with the limit test for chlorides (250 ppm)
potassium iodate until the dark brown colour becomes pale Sulphates (2.3.17). To 5 ml of solution A add 0.5 ml of
brown. Add 5 ml of chloroform and continue the titration, hydrochloric acid and dilute to 15 ml with distilled water; the
shaking well after each addition, until the chloroform becomes solution complies with the limit test for sulphates (300 ppm).
colourless.
Reducing substances. To 5 ml of solution A add 0.25 ml of
1 ml of 0.05 M potassium iodate is equivalent to 0.02120 g of 0.02 M potassium permanganate and 5 ml of 1 M sulphuric
C11H8I3N2NaO4. acid and heat in a water-bath for 5 minutes; the pink colour is
Storage. Store protected from light. not completely discharged.

Labelling. The label states (1) the concentration of the active Disodium phosphate. Dilute 10 ml of solution A to 50 ml with
ingredient; (2) whether the contents are intended for water and titrate with 0.05 M sulphuric acid using
intravenous injection and, if so, that the unused portion bromocresol green solution as indicator; not more than 1 ml
remaining in the container after use must be discarded. of 0.05 M sulphuric acid is required.
Loss on drying (2.4.19). 21.5 to 24.0 per cent, determined on
0.25 g by drying in an oven at 130º.
Assay. Weigh accurately about 2.5 g, dissolve in 40 ml of
Sodium Dihydrogen Phosphate water and titrate with carbonate-free 1 M sodium hydroxide,
Dihydrate determining the end-point potentiometrically (2.4.25).
Sodium Acid Phosphate 1 ml of 1 M sodium hydroxide is equivalent to 0.120 g of
NaH2PO4,2H2O Mol. Wt. 156.0 NaH2PO4.

Sodium Dihydrogen Phosphate Dihydrate contains not less Storage. Store protected from moisture.
than 98.0 per cent and not more than 100.5 per cent of
NaH2PO4, calculated on the dried basis.
Description. Colourless crystals or a white powder; odourless.
Sodium Fluoride
Identification
NaF Mol. Wt. 41.9
A. Dissolve 10.0 g in sufficient carbon dioxide-free water to
produce 100 ml (solution A). Solution A is faintly acid. Sodium Fluoride contains not less than 98.5 per cent and not
more than 100.5 per cent of NaF, calculated on the dried basis.
B. Solution A neutralised with a 10 per cent w/v solution of
potassium hydroxide gives reaction A of sodium salts (2.3.1). Description. A white powder or colourless crystals.

1093
SODIUM FORMALDEHYDE SULPHOXYLATE IP 2007

Identification Sodium Formaldehyde Sulphoxylate


A. Dissolve 2.5 g in sufficient carbon dioxide-free water
without heating to produce 100 ml (solution A). To 2 ml of O
solution A add 0.5 ml of calcium chloride solution; a gelatinous S OH ,2H2 O
Na O
white precipitate is produced which dissolves on adding 5 ml
of ferric chloride solution.
CH3NaO3S,2H2O Mol Wt. 154.1
B. Add about 4 mg to a mixture of 0.1 ml of alizarin red S
Sodium Formaldehyde Sulphoxylate is monosodium
solution and 0.1 ml of zirconyl nitrate solution and mix; the
hydroxymethane sulphinate dihydrate. It may contain a
colour changes to yellow.
suitable stabilising agent such as sodium carbonate.
C. Gives reaction A of sodium salts (2.3.1).
Sodium Formaldehyde Sulphoxylate contains an amount of
Tests CH3NaO3S equivalent to not less than 45.0 per cent and
not more than 55.0 per cent of SO2, calculated on the dried
Appearance of solution. Solution A is clear (2.4.1), and basis.
colourless (2.4.1).
Description. White crystals or hard white masses; odour,
Acidity or alkalinity. Dissolve 2.5 g of potassium nitrate in characteristic and garlic-like.
40 ml of solution A, dilute to 50 ml with carbon dioxide-free
water, cool to 0º and add 0.2 ml of dilute phenolphthalein Identification
solution. If the solution is colourless, not more than 1.0 ml of
0.1 M sodium hydroxide is required to produce a red colour A. Dissolve about 4 g in 10 ml of water in a test-tube and add
that persists for not less than 15 seconds. If the solution is 1 ml of ammoniacal silver nitrate solution; metallic silver is
red, not more than 0.25 ml of 0.1 M hydrochloric acid is produced either as a finely divided grey precipitate or as a
required to change the colour of the solution. Reserve the bright metallic mirror on the inner surface of the tube.
neutralised solution for the test for Fluorosilicate. B. Add about 50 mg to a solution of 40 mg of salicylic acid in
Chlorides (2.3.12). 40 ml of solution A complies with the limit 5 ml of sulphuric acid and warm very gently; a permanent
test for chlorides (250 ppm). deep red colour develops.
Fluorosilicate. Heat to boiling the solution reserved in the Tests
test for Acidity or alkalinity and titrate while hot with 0.1 M
sodium hydroxide until a red colour is produced. Not more Appearance of solution. A 5.0 per cent w/v solution in carbon
than 1.5 ml of 0.1 M sodium hydroxide is required. dioxide-free water is clear (2.4.1), and colourless (2.4.1).
Sulphates (2.3.17). Dissolve 0.25 g in 10 ml of a saturated Alkalinity. Dissolve 1.0 g in 50 ml of water and add 0.15 ml of
solution of boric acid in distilled water and add 5 ml of dilute phenolphthalein solution; not more than 3.5 ml of 0.05
distilled water and 0.6 ml of 7 M hydrochloric acid. The M sulphuric acid is required to change the colour of the
solution complies with the limit test for sulphates (200 ppm). solution.
Prepare the standard by mixing together 0.6 ml of 7 M pH (2.4.24). 9.5 to 10.5, determined in a 2.0 per cent w/v solution
hydrochloric acid, 5 ml of sulphate standard solution in carbon dioxide-free water.
(10 ppm SO4) and 10 ml of a saturated solution of boric acid
in distilled water. Iron (2.3.14). Ignite 1.0 g, initially at a low temperature until
thoroughly charred and finally at about 600º, preferably in a
Loss on drying (2.4.19). Not more than 0.5 per cent, determined muffle furnace, until all the carbon has been burnt off. Cool,
on 1.0 g by drying in an oven at 130º for 3 hours. dissolve the residue in 2 ml of hydrochloric acid and dilute to
Assay. Weigh accurately about 80 mg, add a mixture of 5 ml of 50 ml with water. Add about 50 mg of ammonium persulphate
acetic anhydride and 20 ml of anhydrous glacial acetic acid and 5 ml of ammonium thiocyanate solution, mix and transfer
and heat to dissolve. Cool, add 20 ml of dioxan. Titrate with to a Nessler cylinder. The red colour of the solution is not
0.1 M perchloric acid, using crystal violet solution as more intense than that of 1.0 ml of iron standard solution
indicator, until a green colour is produced. Carry out a blank (10 ppm) treated in the same manner.
titration. Sulphides. Dissolve 6 g in 14 ml of water in a test-tube and
1 ml of 0.1 M perchloric acid is equivalent to 0.004199 g of wet a strip of lead acetate paper in the clear solution; no
NaF. discolouration is evident within 5 minutes.
Storage. Store protected from moisture. Sodium sulphite. Not more than 5.0 per cent, calculated as

1094
IP 2007 SODIUM FUSIDATE

Na2SO3, determined by the following method. Transfer 4.0 ml vigorously for 1 minute, allow to separate and filter the lower
of the solution obtained in the Assay to a flask, add 2 ml of layer through absorbent cotton covered with anhydrous
formaldehyde solution and titrate with 0.1 M iodine that is sodium sulphate. Repeat the extraction with two quantities,
used for the Assay, adding starch solution towards the end each of 5 ml, of chloroform, evaporate the combined extracts
of the titration as indicator. at a pressure of 2 kPa, dry the residue over phosphorus
Calculate the percentage of Na2SO3 from the expression pentoxide at a pressure not exceeding 0.7 kPa for 2 hours and
78.775(V2 – V1)/W, dissolve in 1 ml of chloroform IR.
On the residue determine by infrared absorption
where V1 and V2 are the volumes, in ml, of 0.1 M iodine
spectrophotometry (2.4.6). Compare the spectrum with that
consumed in this test and in the Assay respectively and W is
obtained with fusidic acid RS or with the reference spectrum
the weight, in g, of the substance under examination taken for
of fusidic acid.
the Assay.
Loss on drying (2.4.19). Not more than 27.0 per cent, B. In the test for Related substances, the principal spot in the
determined on 0.5 g by drying in an oven at 105º for 3 hours. chromatogram obtained with the test solution (b) corresponds
to that in the chromatogram obtained with reference solution
Assay. Weigh accurately about 1.0 g, dissolve in 25 ml of (a).
water, add sufficient water to produce 50.0 ml and mix. To 4.0
ml of this solution add 100 ml of water and titrate with 0.1 M C. Ignite 1 g. The residue gives the reactions of sodium salts
iodine using 3 ml of starch solution, added towards the end (2.3.1).
of the titration, as indicator.
Tests
1 ml of 0.1 M iodine is equivalent to 0.001602 g of SO2.
Storage. Store protected from light and moisture. Appearance of solution. A 15.0 per cent w/v solution in carbon
dioxide-free water is not more intensely coloured than
reference solution BS6 (2.4.1).

Sodium Fusidate pH (2.4.24). 7.5 to 9.0, determined in a 1.25 per cent w/v solution.
Specific optical rotation (2.4.22). +5.0º to +8.0º, determined at
H3C CH3 20º by dissolving 1.5 g in 25 ml of water, adding 0.1 ml of 5 M
ammonia and diluting to 50 ml with water.
COONa Related substances. Determine by thin-layer chromatography
H
HO (2.4.17), coating the plate with silica gel G.
H3C CH 3 O Mobile phase. A mixture of 80 volumes of chloroform,
CH3 10 volumes of glacial acetic acid, 10 volumes of cyclohexane
H CH3 O and 2.5 volumes of methanol.
HO
H Test solution (a). Dissolve 0.2 g of the substance under
H3C examination in 10 ml of ethanol.

C31H47NaO6 Mol. Wt. 538.7 Test solution (b). Dissolve 0.2 g of the substance under
examination in 100 ml of ethanol.
Sodium Fusidate is sodium (17Z)-16β-acetoxy-3α,11α-
dihydroxyfusida-17(20),24-dien-21-oate, produced by the Reference solution (a). A 0.24 per cent w/v solution of
growth of certain strains of Fusidium coccineum or by any diethanolamine fusidate RS in ethanol.
other means. Reference solution (b). A 0.04 per cent w/v solution of
Sodium Fusidate contains not less than 97.5 per cent and not diethanolamine fusidate RS in ethanol.
more than 101.0 per cent of C31H47NaO6, calculated on the Reference solution (c). A 0.04 per cent w/v solution of
anhydrous basis. 3-ketofusidic acid RS in ethanol.
Description. A white or almost white, crystalline powder; Apply to the plate 5 µl of each solution. After development,
slightly hygroscopic. dry the plate at 110º for 10 minutes, spray with ethanolic
sulphuric acid (10 per cent), dry at 110º for 10 minutes and
Identification
examine in ultraviolet light at 365 nm. Any red secondary spot
A.Dissolve 0.1 g in 5 ml of water, add 5 ml of chloroform and in the chromatogram obtained with test solution (a) is not
0.1 ml of a 10 per cent w/w solution of phosphoric acid, shake more intense than the spot in the chromatogram obtained

1095
SODIUM FUSIDATE CAPSULES IP 2007

with reference solution (b). Any yellow secondary spot in the Test solution (a). Extract a quantity of the contents of the
chromatogram obtained with test solution (a) is not more capsules containing 50 mg of Sodium Fusidate with 5 ml of
intense than the spot in the chromatogram obtained with ethanol (95 per cent), centrifuge and use the supernatant
reference solution (c). liquid.
Water (2.3.43). Not more than 2.0 per cent, determined on Test solution (b). Dissolve 0.2 g of the substance under
0.5 g. examination in 100 ml of ethanol.
Assay. Weigh accurately about 0.2 g and dissolve in a mixture Reference solution (a). A 0.24 per cent w/v solution of
of 15 ml of water and 20 ml of ethanol (95 per cent). Titrate diethanolamine fusidate RS in ethanol.
with 0.1 M hydrochloric acid to pH 4.1, stirring continuously,
determining the end-point potentiometrically (2.4.25). Reference solution (b). A 0.04 per cent w/v solution of
diethanolamine fusidate RS in ethanol.
1 ml of 0.1 M hydrochloric acid is equivalent to 0.05387 g of
C31H47NaO6. Reference solution (c). A 0.04 per cent w/v solution of 3-
ketofusidic acid RS in ethanol.
Storage. Store protected from light and moisture.
Apply to the plate 5 µl of each solution. After development,
dry the plate at 110º for 10 minutes, spray with ethanolic
sulphuric acid (10 per cent), dry at 110º for 10 minutes and
Sodium Fusidate Capsules examine in ultraviolet light (365 nm). Any red secondary spot
in the chromatogram obtained with the test solution (a) is not
Sodium Fusidate Capsules contain not less than 92.5 per cent more intense than the spot in the chromatogram obtained
and not more than 107.5 per cent of the stated amount of with reference solution (b). Any yellow secondary spot in the
sodium fusidate, C31H47NaO6. chromatogram obtained with the test solution (a) is not more
intense than the spot in the chromatogram obtained with
Ident ification
reference solution (c).
A.Dissolve a quantity of the contents of the capsules
Other tests. Complies with the tests stated under Capsules.
containing 0.1 g of Sodium Fusidate in 5 ml of water, add 5 ml
of chloroform and 0.1 ml of a 10 per cent w/w solution of Assay. Determine by liquid chromatography (2.4.14)
phosphoric acid, shake vigorously for 1 minute, allow to
Test solution. Dissolve a quantity of the mixed contents of 20
separate and filter the lower layer through absorbent cotton
capsules containing about 25 mg of Sodium Fusidate in 20.0
covered with anhydrous sodium sulphate. Repeat the
ml of the mobile phase.
extraction with two quantities, each of 5 ml, of chloroform,
evaporate the combined extracts at a pressure of 2 kPa, dry Reference solution. A 0.15 per cent w/v solution of
the residue over phosphorus pentoxide at a pressure not diethanolamine fusidate RS in the mobile phase.
exceeding 0.7 kPa for 2 hours and dissolve in 1 ml of chloroform
Chromatographic system
IR.
– a stainless steel column 20 cm x 4.6 mm, packed with
Determine by infrared absorption spectrophotometry (2.4.6). octadecylsilane bonded to porous silica (5 µm), (such
Compare the spectrum with that obtained with fusidic acid RS as Lichrosorb RP 18),
or with the reference spectrum of fusidic acid. – mobile phase: a mixture of 60 volumes of acetonitrile,
B. In the test for Related substances, the principal spot in the 30 volumes of a 1 per cent v/v solution of glacial acetic
chromatogram obtained with test solution (b) corresponds to acid and 10 volumes of methanol,
that in the chromatogram obtained with reference solution (a). – flow rate. 1.5 ml per minute,
C. Ignite a portion of the contents of the capsules. The residue – spectrophotometer set at 235 nm,
gives the reactions of sodium salts (2.3.1). – a 20 µl loop injector.
Inject alternately the test solution and the reference solution.
Tests
The column efficiency, determined using the principal peak in
Related substances. Determine by thin-layer chromatography the chromatogram obtained with the reference solution, should
(2.4.17), coating the plate with silica gel G. be not less than 14,000 theoretical plates per metre.
Mobile phase. A mixture of 80 volumes of chloroform, Calculate the content of C31H47NaO6 in the capsules.
10 volumes of glacial acetic acid, 10 volumes of cyclohexane
and 2.5 volumes of methanol. Storage. Store protected from light and moisture.

1096
IP 2007 SODIUM LACTATE INJECTION

Sodium Hydroxide solution and titrate with 1 M hydrochloric acid. Add 0.3 ml of
methyl orange solution and continue the titration with 1 M
Caustic Soda hydrochloric acid.
NaOH Mol. Wt. 40.0 1 ml of 1 M hydrochloric acid used in the second part of the
Sodium Hydroxide contains not less than 97.0 per cent and titration is equivalent to 0.0530 g of Na2CO3.
not more than 100.5 per cent of total alkali, calculated as NaOH. 1 ml of 1 M hydrochloric acid used in the combined titrations
Description. White, crystalline masses supplied as sticks, is equivalent to 0.0400 g of total alkali, calculated as NaOH.
pellets or slabs; deliquescent. Readily absorbs carbon dioxide. Storage. Store protected from moisture, in non-metallic
CAUTION — Great care should be exercised in handling containers.
Sodium Hydroxide as it rapidly destroys tissues.

Identification Sodium Lactate Injection


A. Carefully dissolve 5.0 g in 12 ml of distilled water, add 17 Sodium Lactate Intravenous Infusion
ml of 7 M hydrochloric acid, adjust the pH to 7.0 with 1 M
hydrochloric acid and add sufficient distilled water to Sodium Lactate Injection is a sterile solution containing
produce 50 ml (solution A). 2 ml of solution A gives reaction A 1.85 per cent w/v of sodium lactate in Water for Injections. It is
of sodium salts (2.3.1). prepared from Lactic Acid with the aid of Sodium Hydroxide
and sufficient Dilute Hydrochloric Acid to adjust the pH of
B. pH of a 0.01 per cent w/v solution, not less than 11.0 (2.4.24). the solution.
Tests Sodium Lactate Injection contains not less than 1.75 per cent
and not more than 1.95 per cent w/v of sodium lactate,
Appearance of solution. A 10.0 per cent w/v solution is clear
C3H5NaO3.
(2.4.1), and colourless (2.4.1).
Arsenic (2.3.10). Dissolve 2.5 g in 50 ml of water, add 16 ml of Description. A clear, colourless solution.
brominated hydrochloric acid AsT, and remove the excess of Identification
bromine with a few drops of stannous chloride solution AsT.
The resulting solution complies with the limit test for arsenic A. When warmed with potassium permanganate, gives
(4 ppm). acetaldehyde, recognisable by its odour.
Heavy metals (2.3.13). Dissolve 1.0 g in 5 ml of water and 10 ml B. The residue on evaporation, when moistened with
of 3 M hydrochloric acid, heat to boiling, cool and dilute to hydrochloric acid and introduced on a platinum wire into a
25 ml with water. The solution complies with the limit test for flame, imparts a yellow colour to the flame.
heavy metals, Method A ( 20 ppm). C. Carry out reaction C of calcium salts (2.3.1); no white
Iron (2.3.14). 20 ml of solution A complies with the limit test for precipitate is produced.
iron (20 ppm).
Tests
Carbonates. Not more than 2.0 per cent, calculated as Na2CO3,
determined in the Assay. pH (2.4.24). 5.0 to 7.0.
Chlorides (2.3.12). Dissolve 2.0 g in 5 ml of water, acidify with Bacterial endotoxins (2.2.3). Not more than 2.0 Endotoxin Units
about 8 ml of nitric acid and dilute to 20 ml with water. The per millimole.
solution, without the addition of dilute nitric acid, complies Other tests. Complies with the tests stated under Parenteral
with the limit test for chlorides (125 ppm). Preparations (Intravenous Infusions).
Sulphates (2.3.17). Dissolve 2.0 g in 6 ml of distilled water, Assay. Measure accurately 10 ml, evaporate to dryness in a
adjust the pH to 7 with hydrochloric acid and dilute to 20 ml platinum dish and ignite very gently until completely
with distilled water. The resulting solution complies with the carbonised. Boil the residue with 25.0 ml of 0.05 M sulphuric
limit test for sulphates (75 ppm). acid, filter and wash thoroughly with hot water. Titrate the
Potassium. Acidify 2.5 ml of solution A with acetic acid and excess of acid in the combined filtrate and washings with
add 0.15 ml of sodium cobaltinitrite solution; no precipitate 0.1 M sodium hydroxide using methyl orange solution as
is formed. indicator.
Assay. Weigh accurately about 2.0 g, dissolve in about 80 ml 1 ml of 0.05 M sulphuric acid is equivalent to 0.01121 g of
of carbon dioxide-free water, add 0.3 ml of phenolphthalein C3H5NaO3.

1097
COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION IP 2007

Storage. Store in single dose containers of glass or plastic. D. Gives reaction C of calcium salts and reaction A of chlorides
On keeping, small solid particles may separate from the solution (2.3.1).
in glass containers.
Tests
Labelling. The label states (1) that the Injection is one-sixth
molar and contains, in one litre, approximately 167 millimoles pH (2.4.24). 4.0 to 6.5.
each of sodium ions and of bicarbonate ions (as lactate); (2)
5-Hydroxymethylfurfural and Related substances. Dilute a
that the injection should not be used if the solution contains
volume containing 1.0 g of Dextrose to 500.0 ml with water
visible solid particles.
and measure the absorbance of the resulting solution at the
maximum at about 284 nm; absorbance at about 284 nm, not
more than 0.25 (2.4.7).
Heavy metals (2.3.13). Evaporate a volume containing 4 g of
Compound Sodium Lactate and dextrose to 10 ml and add 2 ml of dilute acetic acid and
Dextrose Injection sufficient water to produce 25 ml. The solution complies with
the limit test for heavy metals, Method A ( 5 ppm).
Compound Sodium Lactate with Dextrose Intravenous
Infusion; Ringer-Lactate Solution with Dextrose for Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
Injection; Hartmann’s Solution with Dextrose for per ml.
Injection. Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Compound Sodium Lactate and Dextrose Injection is a sterile
solution containing 0.24 per cent v/v of Lactic Acid (equivalent Assay. For sodium — Dilute suitably with water and determine
to 0.32 per cent w/v of sodium lactate) with 0.6 per cent w/v of by Method A for flame photometry (2.4.4), or by Method A for
Sodium Chloride, 0.04 per cent w/v of Potassium Chloride, atomic absorption spectrophotometry (2.4.2), measuring at
0.027 per cent w/v of Calcium Chloride and 5 per cent w/v of 589 nm and using sodium solution FP or sodium solution
Dextrose in Water for Injections. AAS respectively, suitably diluted with water for the standard
solutions.
Compound Sodium Lactate and Dextrose Injection contains
not less than 0.27 per cent and not more than 0.32 per cent For potassium — Dilute suitably with water and determine
w/v of sodium, Na, not less than 0.019 per cent and not more by Method A for flame photometry (2.4.4), or by Method A for
than 0.022 per cent w/v of potassium, K, not less than 0.37 per atomic absorption spectrophotometry (2.4.2), measuring at
cent and not more than 0.42 per cent w/v of total chloride, Cl, 767 nm and using potassium solution FP or potassium
not less than 0.025 per cent and not more than 0.029 per cent solution AAS respectively, suitably diluted with water for the
w/v of calcium chloride, CaCl2,2H2O, and not less than 0.23 standard solutions.
per cent and not more than 0.28 per cent w/v of lactate, For total chlorides — To 20.0 ml add 30 ml of water, 50.0 ml of
calculated as C3H6O3, and not less than 4.50 per cent and not 0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the
more than 5.25 per cent w/v of dextrose, C6H12O6. It contains precipitate with water slightly acidified with nitric acid and
no antimicrobial agent. titrate the excess of silver nitrate with 0.1 M ammonium
Description. A clear, colourless or faintly straw-coloured thiocyanate using ferric ammonium sulphate solution as
solution. indicator until a reddish yellow colour is produced. Carry out
a blank titration.
Identification 1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution; chloride, calculated as Cl.
the solution remains blue and clear. Heat to boiling, a copious For calcium chloride — To 50.0 ml add 5.0 ml of 0.01 M
red precipitate is formed. magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and
B. When warmed with potassium permanganate gives titrate with 0.01 M disodium edetate using eriochrome black
acetaldehyde, recognisable by its odour. T mixture as indicator. From the volume of 0.01 M disodium
edetate required subtract the volume of 0.01 M magnesium
C. The residue on evaporation, when moistened with sulphate added.
hydrochloric acid and introduced on a platinum wire into a
flame imparts a yellow colour to the flame. When viewed 1 ml of the remainder of 0.01 M disodium edetate is equivalent
through a suitable blue glass, the flame is tinged reddish to 0.00147 g of CaCl2, 2H2O.
purple. For lactate — Determine by liquid chromatography (2.4.14).

1098
IP 2007 HALF STRENGTH COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION

Test solution. The preparation under examination. than 0.011 per cent w/v of potassium, K, not less than
Reference solution. A 0.28 per cent w/v solution of lithium 0.185 per cent and not more than 0.210 per cent w/v of total
lactate RS in the mobile phase. chloride, Cl, not less than 0.0125 per cent and not more than
0.0145 per cent w/v of calcium chloride, CaCl2,2H2O, and not
Chromatographic system less than 0.115 per cent and not more than 0.140 per cent w/v
– a stainless steel column 20 cm x 4.6 mm, packed with of lactate, calculated as C3H6O3, and not less than 4.5 per cent
octadecylsilane bonded to porous silica (5 µm), and not more than 5.25 per cent w/v of dextrose, C6H12O6. It
– mobile phase: a mixture of 90 volumes of water and contains no antimicrobial agent.
10 volumes of a 2 per cent v/v solution of octylamine in
Description. A clear, colourless or faintly straw-coloured
acetonitrile, the pH of which is adjusted to 7.0 with a
solution.
10 per cent v/v solution of phosphoric acid,
– flow rate. 2 ml per minute,
– spectrophotometer set at 210 nm,
Identification
– a 10 µl loop injector. A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution;
Inject separately the test solution and the reference solution the solution remains blue and clear. Heat to boiling, a copious
and measure the responses for the major peak. red precipitate is formed.
Calculate the content of C3H6O3, in the injection. B. When warmed with potassium permanganate gives
acetaldehyde, recognisable by its odour.
For dextrose — To an accurately measured volume containing
2 g to 5 g of Dextrose, add 0.2 ml of 5 M ammonia and sufficient C. The residue on evaporation, when moistened with
water to produce 100.0 ml. Mix well, allow to stand for hydrochloric acid and introduced on a platinum wire into a
30 minutes and measure the optical rotation in a 2-dm tube flame imparts a yellow colour to the flame. When viewed
(2.4.22). The observed rotation in degrees multiplied by through a suitable blue glass, the flame is tinged reddish
0.9477 represents the weight, in g, of dextrose, C6H12O6, in the purple.
volume taken for assay. D. Gives reaction C of calcium salts and reaction A of chlorides
Storage. Store in single dose containers of glass or plastic at (2.3.1).
a temperature not exceeding 30º. On keeping, small particles
may separate from the solution in glass containers. Tests
Labelling. The label states (1) that the injection contains, in pH (2.4.24). 4.0 to 6.5.
millimoles per litre, the following approximate amounts of the
5-Hydroxymethylfurfural and Related substances. Dilute a
ions. sodium, 131, potassium, 5, calcium, 2; bicarbonate
volume containing 1.0 g of Dextrose to 500.0 ml with water
(as lactate), 29 and Chloride, 111; (2) the total osmolar
and measure the absorbance of the resulting solution at the
concentration in mOsmol per litre; (3) that the injection should
maximum at about 284 nm; absorbance at about 284 nm, not
not be used if it contains visible particles.
more than 0.25 (2.4.7).
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
Half Strength Compound Sodium per ml.
Lactate and Dextrose Injection Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Half Strength Compound Sodium Lactate with Dextrose
Assay. For sodium — Dilute suitably with water and determine
Intravenous Infusion; Half Strength Ringer-Lactate
by Method A for flame photometry (2.4.4), or by Method A for
Solution with Dextrose Injection atomic absorption spectrophotometry (2.4.2), measuring at
Half Strength Compound Sodium Lactate and Dextrose 589 nm and using sodium solution FP or sodium solution
Injection is a sterile solution containing 0.12 per cent v/v of AAS respectively, suitably diluted with water for the standard
Lactic Acid (equivalent to 0.16 per cent w/v of sodium lactate) solutions.
with 0.3 per cent w/v of Sodium Chloride, 0.02 per cent w/v of For potassium — Dilute suitably with water and determine
Potassium Chloride, 0.0135 per cent w/v of Calcium Chloride by Method A for flame photometry (2.4.4), or by Method A for
and 5 per cent w/v of Dextrose in Water for Injections. atomic absorption spectrophotometry (2.4.2), measuring at
Compound Sodium Lactate and Dextrose Injection contains 767 nm and using potassium solution FP or potassium
not less than 0.135 per cent and not more than 0.16 per cent solution AAS respectively, suitably diluted with water for the
w/v of sodium, Na, not less than 0.0095 per cent and not more standard solutions.

1099
MODIFIED COMPOUND SODIUM LACTATE AND DEXTROSE INJECTION IP 2007

For total chlorides — To 20.0 ml add 30 ml of water, 50.0 ml of concentration in mOsmol per litre; (3) that the injection should
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the not be used if it contains visible particles.
precipitate with water slightly acidified with nitric acid and
titrate the excess of silver nitrate with 0.1 M ammonium
thiocyanate using ferric ammonium sulphate solution as
indicator until a reddish yellow colour is produced. Carry out Modified Compound Sodium Lactate
a blank titration. and Dextrose Injection
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total Modified Compound Sodium Lactate with Dextrose
chloride, calculated as Cl. Intravenous Infusion; Modified Lactated Ringer’s and
For calcium chloride — To 50.0 ml add 5.0 ml of 0.01 M Dextrose Injection.
magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and
Modified Compound Sodium Lactate and Dextrose Injection
titrate with 0.01 M disodium edetate using eriochrome black
is a sterile solution containing 0.048 per cent v/v of Lactic
T mixture as indicator. From the volume of 0.01 M disodium
Acid (equivalent to 0.064 per cent w/v of sodium lactate) with
edetate required subtract the volume of 0.01 M magnesium
0.12 per cent w/v of Sodium Chloride, 0.008 per cent w/v of
sulphate added.
Potassium Chloride, 0.0054 per cent w/v of Calcium Chloride
1 ml of the remainder of 0.01 M disodium edetate is equivalent and 5 per cent w/v of Dextrose in Water for Injections.
to 0.00147 g of CaCl2, 2H2O.
Modified Compound Sodium Lactate and Dextrose Injection
For lactate — Determine by liquid chromatography (2.4.14). contains not less than 0.054 per cent and not more than
Test solution. The preparation under examination. 0.064 per cent w/v of sodium, Na, not less than 0.0038 per cent
and not more than 0.0044 per cent w/v of potassium, K, not
Reference solution. A 0.28 per cent w/v solution of lithium less than 0.074 per cent and not more than 0.084 per cent w/v
lactate RS in the mobile phase. of total chloride, Cl, not less than 0.005 per cent and not more
Chromatographic system than 0.0058 per cent w/v of calcium chloride, CaCl2,2H2O, and
– a stainless steel column 20 cm x 4.6 mm, packed with not less than 0.046 per cent and not more than 0.056 per cent
octadecylsilane bonded to porous silica (5 µm), w/v of lactate, calculated as C3H6O3, and not less than 4.5 per
– mobile phase: a mixture of 90 volumes of water and cent and not more than 5.25 per cent w/v of dextrose, C6H12O6.
10 volumes of a 2 per cent v/v solution of octylamine in It contains no antimicrobial agent.
acetonitrile, the pH of which is adjusted to 7.0 with a Description. A clear, colourless or faintly straw-coloured
10 per cent v/v solution of phosphoric acid, solution.
– flow rate. 2 ml per minute,
– spectrophotometer set at 210 nm, Identification
– a 10 µl loop injector.
A. To 1 ml add 0.05 ml of potassium cupri-tartrate solution;
Inject separately the test solution and the reference solution the solution remains blue and clear. Heat to boiling, a copious
and measure the responses for the major peak. red precipitate is formed.
Calculate the content of C3H6O3 in the injection. B. When warmed with potassium permanganate gives
For dextrose — To an accurately measured volume containing acetaldehyde, recognisable by its odour.
2 g to 5 g of Dextrose, add 0.2 ml of 5 M ammonia and sufficient
C. The residue on evaporation, when moistened with
water to produce 100.0 ml. Mix well, allow to stand for
hydrochloric acid and introduced on a platinum wire into a
30 minutes and measure the optical rotation in a 2-dm tube
flame imparts a yellow colour to the flame. When viewed
(2.4.22). The observed rotation in degrees multiplied by 0.9477
through a suitable blue glass, the flame is tinged reddish
represents the weight, in g, of dextrose, C6H12O6, in the volume
purple.
taken for assay.
Storage. Store in single dose containers of glass or plastic at D. Gives reaction C of calcium salts and reaction A of chlorides
a temperature not exceeding 30º. On keeping, small particles (2.3.1).
may separate from the solution in glass containers.
Tests
Labelling. The label states (1) that the injection contains, in
pH (2.4.24). 4.0 to 6.5.
millimoles per litre, the following approximate amounts of the
ions. sodium, 65.5, potassium, 2.5, calcium, 1, bicarbonate 5-Hydroxymethylfurfural and Related substances. Dilute a
(as lactate), 14.5, and chloride, 55.5; (2) the total osmolar volume containing 1.0 g of Dextrose to 500.0 ml with water

1100
IP 2007 COMPOUND SODIUM LACTATE INJECTION

and measure the absorbance of the resulting solution at the – flow rate. 2 ml per minute,
maximum at about 284 nm; absorbance at about 284 nm (2.4.7), – spectrophotometer set at 210 nm,
not more than 0.25. – a 10 µl loop injector.
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit Inject separately the test solution and the reference solution
per ml. and measure the responses for the major peak.
Other tests. Complies with the tests stated under Parenteral Calculate the content of C3H6O3 in the injection.
Preparations (Injections). For dextrose — To an accurately measured volume containing
Assay. For sodium — Dilute suitably with water and determine 2 g to 5 g of Dextrose, add 0.2 ml of 5 M ammonia and sufficient
by Method A for flame photometry (2.4.4), or by Method A for water to produce 100.0 ml. Mix well, allow to stand for
atomic absorption spectrophotometry (2.4.2), measuring at 30 minutes and determine the optical rotation in a 2-dm tube
589 nm and using sodium solution FP or sodium solution (2.4.22). The observed rotation in degrees multiplied by
AAS respectively, suitably diluted with water for the standard 0.9477 represents the weight, in g, of dextrose, C6H12O6, in the
solutions. volume taken for assay.
For potassium — Dilute suitably with water and determine Storage. Store in single dose containers of glass or plastic at
by Method A for flame photometry (2.4.4), or by Method A for a temperature not exceeding 30º. On keeping, small particles
atomic absorption spectrophotometry (2.4.2), measuring at may separate from the solution in glass containers.
767 nm and using potassium solution FP or potassium Labelling. The label states (1) that the injection contains, in
solution AAS respectively, suitably diluted with water for the millimoles per litre, the following approximate amounts of the
standard solutions. ions. sodium, 26.2, potassium, 1, calcium, 0.4, bicarbonate (as
For total chlorides — To 20.0 ml add 30 ml of water, 50.0 ml of lactate), 5.8, and chloride, 22.2; (2) the total osmolar
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the concentration in mOsmol per litre; (3) that the injection should
precipitate with water slightly acidified with nitric acid and not be used if it contains visible particles.
titrate the excess of silver nitrate with 0.1 M ammonium
thiocyanate using ferric ammonium sulphate solution as
indicator until a reddish yellow colour is produced. Carry out Compound Sodium Lactate Injection
a blank titration.
Compound Sodium Lactate Intravenous Infusion; Ringer-
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total
chloride, calculated as Cl.
Lactate Solution for Injection; Hartmann’s Solution for
Injection
For calcium chloride — Evaporate 100.0 ml on a water-bath
to 50 ml, add to this solution 5.0 ml of 0.01 M magnesium Compound Sodium Lactate Injection is a sterile solution
sulphate and 5 ml of ammonia buffer pH 10.9 and titrate with containing 0.24 per cent v/v of Lactic Acid (equivalent to
0.01 M disodium edetate using eriochrome black T mixture 0.32 per cent w/v of sodium lactate) with 0.6 per cent w/v of
as indicator. From the volume of 0.01 M disodium edetate Sodium Chloride, 0.04 per cent w/v of Potassium Chloride and
required subtract the volume of 0.01 M magnesium sulphate 0.027 per cent w/v of Calcium Chloride in Water for Injections.
added. Compound Sodium Lactate Injection contains not less than
1 ml of the remainder of 0.01 M disodium edetate is equivalent 0.27 per cent and not more than 0.32 per cent w/v of sodium,
to 0.00147 g of CaCl2, 2H2O. Na, not less than 0.019 per cent and not more than 0.022 per
cent w/v of potassium, K, not less than 0.37 per cent and not
For lactate — Determine by liquid chromatography (2.4.14). more than 0.42 per cent w/v of total chloride, Cl, not less than
Test solution. The preparation under examination. 0.025 per cent and not more than 0.029 per cent w/v of calcium
chloride, CaCl2,2H2O, and not less than 0.23 per cent and not
Reference solution. A 0.28 per cent w/v solution of lithium
more than 0.28 per cent w/v of lactate, calculated as C3H6O3.
lactate RS in the mobile phase.
Description. A clear, colourless solution.
Chromatographic system
– a stainless steel column 20 cm x 4.6 mm, packed with Identification
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 90 volumes of water and A. When warmed with potassium permanganate gives
10 volumes of a 2 per cent v/v solution of octylamine in acetaldehyde, recognisable by its odour.
acetonitrile, the pH of which is adjusted to 7.0 with a B. The residue on evaporation, when moistened with
10 per cent v/v solution of phosphoric acid, hydrochloric acid and introduced on a platinum wire into a

1101
COMPOUND SODIUM LACTATE SOLUTION FOR IRRIGATION IP 2007

flame, imparts a yellow colour to the flame. When viewed On keeping, small solid particles may separate from the solution
through a suitable blue glass, the flame is tinged with reddish in glass containers.
purple. Labelling. The label states (1) that the injection contains, in
C. Gives reaction C of calcium salts (2.3.1). millimoles per litre, the following approximate amounts of the
ions. sodium, 131, potassium, 5, calcium, 2, bicarbonate
Tests (as lactate), 29 and chloride, 111; (2) that the injection should
not be used if the solution contains visible solid particles.
pH (2.4.24). 5.0 to 7.0.
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin
Unit per ml.
Other tests. Complies with the tests stated under Parenteral Compound Sodium Lactate Solution
Preparations (Intravenous Infusions). For Irrigation
Assay. For sodium — Dilute suitably with water and determine
by Method A for flame photometry (2.4.4), or by atomic Ringer-Lactate Solution for Irrigation; Hartmann’s
absorption spectrophotometry (2.4.2), measuring at 589 nm Solution for Irrigation
and using sodium solution FP or sodium solution AAS, Compound Sodium Lactate Solution for Irrigation is a sterile
suitably diluted for the standard solutions. solution containing 0.24 per cent v/v of Lactic Acid (equivalent
For potassium — Dilute suitably with water and determine to 0.32 per cent w/v of sodium lactate) with 0.6 per cent w/v of
by Method A for flame photometry (2.4.4), or by atomic Sodium Chloride, 0.04 per cent w/v of Potassium Chloride and
absorption spectrophotometry (2.4.2), measuring at 767 nm 0.027 per cent w/v of Calcium Chloride in Water for Injections.
and using potassium solution FP or potassium solution AAS, Compound Sodium Lactate Solution for Irrigation contains
suitably diluted for the standard solutions. not less than 0.27 per cent and not more than 0.32 per cent
For total chlorides — To 20 ml add 30 ml of water, 50.0 ml of w/v of sodium, Na, not less than 0.019 per cent and not more
0.1 M silver nitrate and 2 ml of nitric acid, filter, wash the than 0.022 per cent w/v of potassium, K, not less than 0.37 per
precipitate with water slightly acidified with nitric acid and cent and not more than 0.42 per cent w/v of total chloride, Cl,
titrate the excess of silver nitrate with 0.1 M ammonium not less than 0.025 per cent and not more than 0.029 per cent
thiocyanate using ferric ammonium sulphate solution as w/v of calcium chloride, CaCl2,2H2O, and not less than 0.23
indicator. Carry out a blank titration. per cent and not more than 0.28 per cent w/v of lactate,
calculated as C3H6O3. It contains no antimicrobial agent.
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total
chloride, calculated as Cl. Description. A clear, colourless solution.

For calcium chloride — To 50 ml add 5.0 ml of 0.01 M Identification


magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and
titrate with 0.01 M disodium edetate using mordant black 11 A. When warmed with potassium permanganate gives
mixture as indicator. From the volume of 0.01 M disodium acetaldehyde, recognisable by its odour.
edetate required subtract the volume of 0.01 M magnesium B. The residue on evaporation, when moistened with
sulphate added. hydrochloric acid and introduced on a platinum wire into a
1 ml of the remainder of 0.01 M disodium edetate is equivalent flame imparts a yellow colour to the flame. When viewed
to 0.00147 g of CaCl2, 2H2O. through a suitable blue glass, the flame is tinged reddish
purple.
For lactate, calculated as C3H6O3 — Evaporate 50 ml to
dryness in a platinum dish and ignite very gently until C. Gives reaction C of calcium salts and reaction A of chlorides
completely carbonised. Boil the residue with 25.0 ml of 0.05 M (2.3.1).
sulphuric acid, filter, and wash thoroughly with hot
Tests
water.Titrate the excess of acid in the combined filtrate and
washings with 0.1 M sodium hydroxide using methyl orange pH (2.4.24). 5.0 to 7.0.
solution as indicator.
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
1 ml of 0.05 M sulphuric acid is equivalent to 0.009008 g of per ml.
C3H6O3.
Other tests. Complies with the tests stated under Parenteral
Storage. Store in single dose containers of glass or plastic. Preparations (Injections).

1102
IP 2007 SODIUM LAURYL SULPHATE

Assay. For sodium — Dilute suitably with water and determine in glass containers. The container may be designed to empty
by Method A for flame photometry (2.4.4), or by Method A for rapidly and may contain a volume of more than one litre.
atomic absorption spectrophotometry (2.4.2), measuring at Labelling. The label states (1) that the irrigation solution
589 nm and using sodium solution FP or sodium solution contains, in millimoles per litre, the following approximate
AAS respectively, suitably diluted with water for the standard amounts of the ions. sodium,131, potassium, 5, calcium,
solutions. 2, bicarbonate (as lactate), 29, and chloride, 111; (2) that the
For potassium — Dilute suitably with water and determine solution should not be used if it contains visible particles; (3)
by Method A for flame photometry (2.4.4), or by Method A for ‘For irrigation only’ and ‘Not for injection’; (4) that once the
atomic absorption spectrophotometry (2.4.2), measuring at container is opened, the unused portion should be discarded.
767 nm and using potassium solution FP or potassium
solution AAS respectively, suitably diluted with water for the
standard solutions.
Sodium Lauryl Sulphate
For calcium chloride — To 50.0 ml add 5.0 ml of 0.01 M
magnesium sulphate and 5 ml of ammonia buffer pH 10.9 and Sodium Lauryl Sulphate is a mixture of sodium alkyl sulphates
titrate with 0.01 M disodium edetate using eriochrome black consisting mainly of sodium dodecyl sulphate,
T mixture as indicator. From the volume of 0.01 M disodium CH3[CH2]10CH2OSO3Na.
edetate required subtract the volume of 0.01 M magnesium Sodium Lauryl Sulphate contains not less than 85.0 per cent
sulphate added. of sodium alkyl sulphates, calculated as C12H25NaO4S.
1 ml of the remainder of 0.01 M disodium edetate is equivalent Description. A white or pale yellow powder or crystals.
to 0.00147 g of CaCl2, 2H2O.
For total chlorides — To 20.0 ml add 30 ml of water, 50.0 ml of Identification
0.1 M silver nitrate and 2 ml of nitric acid. Filter, wash the A. A 1 per cent w/v solution, when shaken, produces plenty
precipitate with water slightly acidified with nitric acid and of foam.
titrate the excess of silver nitrate with 0.1 M ammonium
thiocyanate using ferric ammonium sulphate solution as B. Mix 0.1 ml of a 1 per cent w/v solution with 0.1 ml of a 0.1 per
indicator until a reddish yellow colour is produced. Carry out cent w/v solution of methylene blue and 2 ml of 1 M sulphuric
a blank titration. acid, add 2 ml of dichloromethane and shake; the
dichloromethane layer is intensely blue.
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of total
chloride, calculated as Cl. C. Mix about 10 mg with 10 ml of ethanol and heat to boiling
on a water-bath, shaking frequently. Filter immediately and
For lactate — Determine by liquid chromatography (2.4.14). evaporate the ethanol. Dissolve the residue in 8 ml of water,
Test solution. The preparation under examination. add 3 ml of 2 M hydrochloric acid, evaporate the solution to
half its volume and cool. Filter and to the filtrate add 1 ml of
Reference solution. A 0.28 per cent w/v solution of lithium
barium chloride solution; a white, crystalline precipitate is
lactate RS in the mobile phase.
produced.
Chromatographic system D. Gives reaction B of sodium salts (2.3.1).
– a stainless steel column 20 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm), Tests
– mobile phase: a mixture of 90 volumes of water and
10 volumes of a 2 per cent v/v solution of octylamine in Alkalinity. Dissolve 1.0 g in 100 ml of carbon dioxide-free
acetonitrile, the pH of which is adjusted to 7.0 with a water and add 0.1 ml of phenol red solution. Not more than
10 per cent v/v solution of phosphoric acid, 0.5 ml of 0.1 M hydrochloric acid is required to change the
– flow rate. 2 ml per minute, colour of the solution.
– spectrophotometer set at 210 nm, Non-esterified alcohols. Not more than 4 per cent, determined
– a 10 µl loop injector. by the following method. Dissolve 10.0 g in 100 ml of water,
Inject separately the test solution and the reference solution add 100 ml of ethanol (95 per cent) and extract the solution
and measure the responses for the major peak. with three quantities, each of 50 ml, of n-pentane, adding
sodium chloride, if necessary, to promote separation of the
Calculate the content of C3H6O3, in the injection.
two layers. Wash the combined organic layers with three
Storage. Store in single dose containers of glass or plastic. quantities, each of 50 ml, of water. Dry the organic solution
On keeping, small solid particles may separate from the solution over anhydrous sodium sulphate, filter and evaporate on a

1103
SODIUM METABISULPHITE IP 2007

water-bath until the odour of pentane is no longer detectable. Tests


Heat the residue at 105º for 15 minutes, cool and weigh.
Acidity. A solution is acidic to phenol red solution.
Sodium Chloride and Sodium Sulphate. Not more than a total
of 8.0 per cent, determined by the following methods. Arsenic (2.3.10). Mix 2.5 g in a porcelain dish with 10 ml of
water, 1.25 g of potassium chlorate and 16 ml of hydrochloric
For sodium chloride — Dissolve 5.0 g in 50 ml of water, add acid and heat to expel chlorine, remove the last traces of
2 M nitric acid dropwise until the solution is neutral to litmus chlorine with a few drops of stannous chloride solution AsT
paper, add 2 ml of potassium chromate solution and titrate and add 35 ml of water. The resulting solution complies with
with 0.1 M silver nitrate. the limit test for arsenic (4 ppm).
1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl.
Heavy metals (2.3.13). Dissolve 1.0 g in 10 ml of water, add 5 ml
For sodium sulphate — Dissolve 0.5 g in 20 ml of water, of hydrochloric acid and evaporate to dryness on a water-
warming gently if necessary, and add 1 ml of a 0.05 per cent bath. Dissolve the residue in 25 ml of water containing 2 ml of
w/v solution of dithizone in acetone. If the solution is red, dilute acetic acid. The solution complies with the limit test
add 1 M nitric acid, dropwise, until it becomes bluish green. for heavy metals, Method A ( 20 ppm).
Add 2 ml of dichloroacetic acid solution and 80 ml of acetone
Iron. To 0.5 g add 2 ml of hydrochloric acid and evaporate on
and titrate with 0.01 M lead nitrate until a permanent orange-
a water-bath to dryness. Dissolve the residue in 2 ml of
red colour is obtained.
hydrochloric acid and 20 ml of water and add a few drops of
1 ml of 0.01 M lead nitrate is equivalent to 0.001420 g of bromine solution, cool, dilute with water to 25 ml, then add
Na2SO4. 50 mg of ammonium persulphate and 5 ml of ammonium
Assay. Weigh accurately about 1.15 g, dissolve in sufficient thiocyanate solution. Any colour produced is not more intense
water to produce 1000.0 ml, warming if necessary. To 20.0 ml than that obtained by adding 5 ml of ammonium thiocyanate
add 15 ml of chloroform, 10 ml of dilute sulphuric acid and solution to a mixture of iron standard solution (20 ppm Fe),
1 ml of dimethyl yellow-oracet blue B solution and titrate 2 ml of hydrochloric acid, 50 mg of ammonium persulphate
with 0.004 M benzethonium chloride, shaking vigorously and sufficient water to produce 25 ml (40 ppm).
and allowing the layers to separate after each addition, until Thiosulphate. Dissolve 1.0 g in 10 ml of 2 M hydrochloric
the chloroform layer acquires a permanent clear green colour. acid and heat on a water-bath for 10 minutes; not more than a
1 ml of 0.004 M benzethonium chloride is equivalent to faint opalescence is produced.
0.001154 g of sodium alkyl sulphates, calculated as Assay. Weigh accurately about 0.1 g and dissolve in 50.0 ml
C12H25NaO4S. 0.05 M iodine, add 1 ml of hydrochloric acid and titrate the
Storage. Store protected from moisture. excess of iodine with 0.1 M sodium thiosulphate using starch
solution, added towards the end of the titration, as indicator.
1 ml of 0.05 M iodine is equivalent to 0.004753 g of Na2S2O5.
Sodium Metabisulphite
Storage. Store protected from light and moisture. On exposure
Sodium Pyrosulphite; Sodium Disulphite to air and moisture it is slowly oxidised to sulphate with
Na2S2O5 Mol. Wt. 190.1 disintegration of the crystals.

Sodium Metabisulphite may be prepared by saturating a


solution of Sodium Hydroxide with sulphur dioxide and
allowing crystallisation to occur. Sodium Methylparaben
Sodium Metabisulphite contains not less than 95.0 per cent
Sodium Methyl Hydroxybenzoate
and not more than 100.5 per cent of Na2S2O5.
Description. Colourless, prismatic crystals or a white or creamy O
white powder; odour, sulphurous. CH3
O
Identification
NaO
A. Gives the reactions of sodium salts (2.3.1).
C8H7NaO Mol. Wt. 174.1
B. A solution decolorises iodinated potassium iodide solution
and the resulting solution gives the reactions of sulphates Sodium Methylparaben is the sodium salt of methyl 4-
(2.3.1). hydroxybenzoate.

1104
IP 2007 SODIUM PHOSPHATE

Sodium Methylparaben contains not less than 99.0 per cent Sodium Phosphate
and not more than 102.0 per cent of C8H7NaO3, calculated on
the anhydrous basis. Disodium Hydrogen Phosphate; Disodium Hydrogen
Description. A white, crystalline powder; odourless or almost
Phosphate Dodecahydrate
odourless; hygroscopic. Na2HPO4,12H2O Mol. Wt. 358.1
Sodium Phosphate contains not less than 98.0 per cent and
Identification not more than 101.0 per cent of Na2HPO4, calculated on the
A. Dissolve 0.5 g in 5 ml of water and acidify to litmus paper anhydrous basis.
with hydrochloric acid; a white precipitate is produced. Wash Description. Colourless, transparent crystals; very
the precipitate with water and dry. efflorescent.
Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with methylparaben
Identification
RS. A 10.0 per cent w/v solution in distilled water (solution A)
B. The residue on ignition gives the reactions of sodium salts gives the reactions of sodium salts and of phosphates (2.3.1).
(2.3.1).
Tests
Tests Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Appearance of solution. A 10.0 per cent w/v solution in water
is clear (2.4.1). Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
of stannated hydrochloric acid AsT. The resulting solution
pH (2.4.24). 9.5 to 10.5, determined in a 0.1 per cent w/v solution. complies with the limit test for arsenic (2 ppm).
Chlorides (2.3.10). Dissolve 1.0 g in 20 ml of water, add 0.2 ml Heavy metals (2.3.13). Dissolve 2.0 g in 10 ml of water, adding
of nitric acid and filter. 15 ml of the filtrate complies with the 4 ml of 1 M acetic acid and diluting to 25 ml with water. The
limit test for chlorides (330 ppm). solution complies with the limit test for heavy metals, Method
Sulphates (2.3.17). Dissolve 0.5 g in 40 ml of water, add 3.5 ml A (10 ppm).
of 2 M hydrochloric acid, dilute to 60 ml with water and filter. Iron (2.3.14). 20 ml of solution A complies with the limit test for
15 ml of the filtrate complies with the limit test for sulphates iron (20 ppm).
(0.12 per cent).
Chlorides (2.3.10). To 10 ml of solution A add 10 ml of 2 M
Water (2.3.43). Not more than 5.0 per cent, determined on nitric acid and dilute to 20 ml with water. The resulting solution
1.0 g. complies with the limit test for chlorides (250 ppm).
Assay. Weigh accurately about 0.1 g, gently boil under a reflux Sulphates (2.3.17). To 2.5 ml of solution A add 2 ml of 2 M
condenser with 25 ml of 1.25 M sodium hydroxide for hydrochloric acid and dilute to 15 ml with distilled water.
30 minutes. Allow to cool, add 25.0 ml of 0.0333 M potassium The resulting solution complies with the limit test for sulphates
bromate, 5 ml of a 12.5 per cent w/v solution of potassium (600 ppm).
bromide and 10 ml of hydrochloric acid and immediately
Reducing substances. To 5 ml of solution A add 5 ml of 1 M
stopper the flask. Shake for 15 minutes and allow to stand for
sulphuric acid and 0.25 ml of 0.02 M potassium permanganate
15 minutes. Add 25 ml of dilute potassium iodide solution
and heat on a water-bath for 5 minutes; the red colour is not
and shake vigorously. Titrate the liberated iodine with 0.1 M
completely discharged.
sodium thiosulphate using starch solution, added towards
the end of the titration, as indicator. Repeat the operation Sodium dihydrogen phosphate. The value of the expression
without the substance under examination. The difference (n2 - 25)/(25 - n1),
between the titrations represents the amount of potassium where n1 and n2 are the titres of 1 M sodium hydroxide obtained
bromate required. The volume of 0.0333 M potassium bromate in the Assay, does not exceed 0.025.
used is equivalent to half of the volume of 0.1 M sodium
thiosulphate required for the titration. Water (2.3.43). 57.0 to 61.0 per cent, determined on 0.1 g
dissolved in a mixture of 10 volumes of methanol and 40
1 ml of 0.0333 M potassium bromate is equivalent to 0.005804 volumes of dimethylformamide.
g of C8H7NaO3.
Assay. Weigh accurately about 4.0 g (w), dissolve in 25 ml of
Storage. Store protected from moisture. water, add 25.0 ml of 1 M hydrochloric acid and titrate

1105
SODIUM PROPYLPARABEN IP 2007

potentiometrically with 1 M sodium hydroxide to the first 15 ml of the filtrate complies with the limit test for sulphates
inflection of the pH curve (n1 ml). Continue the titration until (0.12 per cent).
the second inflection of the curve is reached; the total volume Water (2.3.43). Not more than 5.0 per cen, determined on 1.0 g.
of sodium hydroxide required is n2 ml.
Assay. Weigh accurately about 0.1 g, gently boil under a reflux
Calculate the content of Na2HPO4 from the expression
condenser with 25 ml of 1.25 M sodium hydroxide for
1420(25 - n1)/w (100 - d),
30 minutes. Allow to cool, add 25.0 ml of 0.0333 M potassium
where d is the percentage water content. bromate, 5 ml of a 12.5 per cent w/v solution of potassium
bromide and 10 ml of hydrochloric acid and immediately
Storage. Store protected from moisture.
stopper the flask. Shake for 15 minutes and allow to stand for
15 minutes. Add 25 ml of dilute potassium iodide solution
and shake vigorously. Titrate the liberated iodine with 0.1 M
Sodium Propylparaben sodium thiosulphate using starch solution, added towards
the end of the titration, as indicator. Repeat the operation
Sodium Propyl Hydroxybenzoate without the substance under examination. The difference
between the titrations represents the amount of potassium
O bromate required. The volume of 0.0333 M potassium bromate
CH3 used is equivalent to half of the volume of 0.1 M sodium
O
thiosulphate required for the titration.
NaO 1 ml of 0.0333 M potassium bromate is equivalent to
0.00674 g of C10H11NaO3.
C10H11NaO3 Mol. Wt. 202.2
Storage. Store protected from moisture.
Sodium Propylparaben is the sodium salt of propyl 4-
hydroxybenzoate.
Sodium Propylparaben contains not less than 99.0 per cent
and not more than 102.0 per cent of C10H11NaO3, calculated on Sodium Salicylate
the anhydrous basis.
COONa
Description. A white, crystalline powder; odourless or almost
OH
odourless; hygroscopic.

Identification
A. Dissolve 0.5 g in 5 ml of water and acidify to litmus paper C7H5NaO3 Mol. Wt. 160.1
with hydrochloric acid; a white precipitate is produced. Wash Sodium Salicylate is sodium 2-hydroxybenzoate.
the precipitate with water and dry.
Sodium Salicylate contains not less than 99.0 per cent and not
Determine by infrared absorption spectrophotometry (2.4.6). more than 101.0 per cent of C7H5NaO3, calculated on the dried
Compare the spectrum with that obtained with propylparaben basis.
RS.
Description. Colourless, small crystals or shiny flakes or a
B. The residue on ignition gives the reactions of sodium salts white, crystalline powder.
(2.3.1).
Identification
Tests
Test A may be omitted if tests B and C are carried out. Test B
Appearance of solution. A 10.0 per cent w/v solution in water may be omitted if tests A and C are carried out.
is clear (2.4.1).
A. Determine by infrared absorption spectrophotometry (2.4.6).
pH (2.4.24). 9.5 to 10.5, determined in a 0.1 per cent w/v solution. Compare the spectrum with that obtained with sodium
Chlorides (2.3.10). Dissolve 1.0 g in 20 ml of water, add 0.2 ml salicylate RS or with the reference spectrum of sodium
of nitric acid and filter. 15 ml of the filtrate complies with the salicylate.
limit test for chlorides (330 ppm). B. A 10.0 per cent w/v solution in carbon dioxide-free water
Sulphates (2.3.17). Dissolve 0.5 g in 40 ml of water, add 3.5 ml prepared from distilled water (solution A) gives the reactions
of 2 M hydrochloric acid, dilute to 60 ml with water and filter. of salicylates (2.3.1).

1106
IP 2007 SODIUM STARCH GLYCOLLATE

C. Gives reaction B of sodium salts (2.3.1). Description. A very fine, white or off-white, free-flowing
powder; odourless or almost odourless.
Tests
Identification
Appearance of solution. Solution A is clear (2.4.1), and not
more intensely coloured than reference solution BYS6 (2.4.1). A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with sodium starch
Acidity. To 20 ml of solution A add 0.1 ml of phenol red solution;
glycollate RS or with the reference spectrum of sodium starch
the solution is yellow. Titrate with 0.01 M sodium hydroxide
glycollate.
to a reddish violet colour; not more than 2.0 ml of 0.01 M
sodium hydroxide is required. B. To 5 ml of a 2 per cent w/v dispersion in water add 0.05 ml
of 0.005 M iodine; a dark blue colour is produced.
Arsenic (2.3.10). Mix 5.0 g with 10 ml of bromine solution and
evaporate to dryness on a water-bath. Ignite gently, cool, C. The solution obtained in the test for Heavy metals gives
dissolve the residue, ignoring any carbon, in 50 ml of water the reactions of sodium salts (2.3.1).
and 14 ml of brominated hydrochloric acid AsT and remove
the excess of bromine with 2 ml of stannous chloride solution Tests
AsT. The resulting solution complies with the limit test for
pH (2.4.24). 5.5 to 7.5, determined in a 2.0 per cent w/v dispersion
arsenic (2 ppm).
in carbon dioxide-free water.
Heavy metals (2.3.13). Dissolve 2.0 g in 46 ml of water, add
Heavy metals (2.3.13). To 4.0 g in a silica or platinum dish add
with constant stirring 4 ml of dilute hydrochloric acid, filtering
2 ml of a 50 per cent w/v solution of sulphuric acid, heat in a
and using 25 ml of the filtrate. The solution complies with the
water-bath and then cautiously over a flame at about 600º.
limit test for heavy metals, Method A ( 20ppm).
Continue heating until all black particles have disappeared,
Chlorides (2.3.12). To 25 ml of solution A add 15 ml of water allow to cool, add 0.1 ml of 1 M sulphuric acid, heat to ignition
and 10 ml of 2 M nitric acid and filter. 25 ml of the filtrate once again and allow to cool. Add 0.1 ml of 2 M ammonium
complies with the limit test for chlorides (200 ppm). carbonate, evaporate to dryness and cautiously ignite. To
Sulphates (2.3.17). 2.5 ml of solution A diluted to 15 ml with the residue add 5 ml of hydrochloric acid, evaporate to
distilled water complies with the limit test for sulphates dryness on a water-bath and dissolve the residue in 100 ml of
(600 ppm). water. 25 ml of a solution complies with the limit test for heavy
metals, Method A (20 ppm).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105º. Iron (2.3.14). 50 ml of the solution obtained in the test for
Heavy metals complies with the limit test for iron (20 ppm).
Assay. Weigh accurately about 0.15 g, dissolve in 30 ml of
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric Sodium Chloride. Not more than 10.0 per cent, determined by
acid, determining the end-point potentiometrically (2.4.25). the following method. To 1.0 g add 20.0 ml of 0.1 M silver
Carry out a blank titration. nitrate and 30 ml of nitric acid and boil carefully for 30 minutes.
Cool and add a sufficient volume of a saturated solution of
1 ml of 0.1 M perchloric acid is equivalent to 0.01601 g of potassium permanganate to change the colour of the solution
C7H5NaO3. to red. Discharge the colour by the dropwise addition of
Storage. Store protected from light and moisture. hydrogen peroxide solution (10 vol), add 3 ml of dibutyl
phthalate and titrate with 0.1 M ammonium thiocyanate using
ferric ammonium sulphate solution as indicator, shaking
vigorously after each addition of titrant. Carry out a blank
Sodium Starch Glycollate titration.
1 ml of 0.1 M silver nitrate is equivalent to 0.005844 g of NaCl.
Sodium Carboxymethyl Starch
Sodium glycollate. To 0.2 g add 5 ml of glacial acetic acid,
Sodium Starch Glycollate is the sodium salt of a poly-a-
mix well and add 5 ml of water, stirring occasionally until
glucopyranose in which some of the hydroxyl groups are in
solution is complete. Slowly add 50 ml of acetone with stirring
the form of carboxymethyl ether.
and then add 1 g of sodium chloride. Filter, wash the residue
Sodium Starch Glycollate contains not less than 2.8 per cent with acetone and dilute the filtrate to 100 ml with acetone.
and not more than 4.5 per cent of sodium, Na, calculated on Transfer 2 ml of this solution to an open flask, heat on a water-
the material washed with Ethanol (95 per cent) and dried as bath for exactly 20 minutes, cool, add 5 ml of naphthalenediol
described under Assay. reagent and mix thoroughly. Add a further 15 ml of the same

1107
SODIUM STIBOGLUCONATE IP 2007

reagent, mix, cover the flask with aluminium foil and heat on a Sodium Stibogluconate contains not less than 30.0 per cent
water-bath for 20 minutes. Cool and dilute to 25 ml with and not more than 34.0 per cent of pentavalent antimony,
sulphuric acid. The absorbance (2.4.7) of the resulting solution calculated on the dried and methanol-free basis.
at the maximum at about 540 nm using water as the blank, is Description. A colourless, mostly amorphous powder;
not more than that of a solution prepared in the following odourless or almost odourless.
manner. To 5 ml of a 0.062 per cent w/v solution of glycollic
acid, previously dried at a pressure not exceeding 2 kPa for Identification
16 hours, add 5 ml of glacial acetic acid, dilute to 100 ml with
acetone and complete the procedure described above A. An aqueous solution is dextro-rotatory.
beginning at the words “Transfer 2 ml....” (2.0 per cent).
B. Pass hydrogen sulphide into a 5 per cent w/v solution for
Microbial Contamination (2.2.9). 1.0 g is free from Escherichia several minutes; an orange precipitate is produced.
coli and Salmonellae.
C. When heated, it chars without melting, emitting an odour
Loss on drying (2.4.19). Not more than 10.0 per cent, of burnt sugar and leaving a residue which gives the reactions
determined on 0.5 g by drying in an oven at 105º. of antimony compounds and the reactions of sodium salts
Assay. Weigh accurately about 4.0 g, add 350 ml of a mixture (2.3.1).
of 4 volumes of ethanol (95 per cent) and 1 volume of water,
add 0.25 ml of phenolphthalein solution and mix. Add 1 M
Tests
sodium hydroxide dropwise until the colour of the suspension pH (2.4.24). 5.0 to 5.6, determined in the solution obtained in
becomes faintly pink, shake for 30 minutes and decant through the test for Stability of solution.
a sintered glass crucible. Repeat the extraction three times, or
until a test for chloride ions is negative. Transfer the bulk of Stability of solution. Heat a solution containing 10.0 per cent
the residue to the crucible, wash the residue with ethanol w/v of pentavalent antimony in an autoclave at 115.5º and at a
(95 per cent) and dry at 110º to constant weight. Weigh pressure of 70 kPa for 30 minutes. The resulting solution is
accurately 0.5 g of the residue, add 80 ml of anhydrous glacial colourless or almost colourless.
acetic acid, heat under a reflux condenser for 2 hours, cool. Trivalent antimony. Dissolve 2.0 g in 30 ml of water, add 15 ml
Titrate with 0.1 M perchloric acid, determining the end-point of hydrochloric acid and titrate with 0.00833 M potassium
potentiometrically (2.4.25). bromate using methyl orange solution as indicator. Not more
1 ml of 0.1 M perchloric acid is equivalent to 0.0023 g of Na. than 1.3 ml of 0.00833 M potassium bromate is required.

Storage. Store protected from light and moisture. Chlorides. Dissolve 2.5 g in 50 ml of water and add 2 ml of 2 M
nitric acid and 75 ml of acetate buffer pH 5.0. Titrate with
0.1 M silver nitrate, determining the end-point
potentiometrically (2.4.25). Not more than 3.0 ml of 0.1 M silver
nitrate is required.
Sodium Stibogluconate
Methanol. Not more than 2.0 per cent w/w, determined by gas
Sodium Antimony Gluconate chromatography (2.4.13).
Test solution. Add 5 ml of water to 0.5 g of the substance
COONa COONa
under examination and mix with the aid of ultrasound until the
HC O O CH solution is complete.
O CH OH HO HC O
Reference solution (a). Add 5 ml of a 0.2 per cent v/v solution
HC O Sb O Sb O CH of ethanol (internal standard) to 0.5 g of the substance under
HC OH HO CH examination and mix with the aid of ultrasound until the solution
H2C OH HO CH2 is complete.
Reference solution (b). Add 1 ml of a 1.0 per cent v/v solution
Sodium Stibogluconate is mainly the disodium salt of µ-oxy- of methanol to 5 ml of a 0.2 per cent v/v solution of the internal
bis[gluconato(3-)O2,O3,O4-hydroxo-antimony]. standard.
The method of manufacture is such as to ensure consistently Chromatographic system
controlled reaction stoichiometry in order to yield sodium – a glass column 1.5 m x 4 mm, packed with porous polymer
stibogluconate that is satisfactory with regard to intrinsic beads (80 to 100 mesh),
toxicity. – temperature: column. 130º,

1108
IP 2007 SODIUM THIOSULPHATE

Calculate the percentage w/w of methanol taking 0.792 g as its Tests


weight per ml at 20º.
pH (2.4.24). 5.0 to 5.6.
Undue toxicity. Dissolve a suitable quantity of the substance
under examination in water for injections to give a solution Undue toxicity. Dilute a suitable volume of the injection under
containing 28 mg of pentavalent antimony per ml. Inject examination with sufficient water for injections to give a
intravenously 0.3 ml of the solution into each of 10 mice that solution containing the equivalent of 28 mg of pentavalent
have been deprived of food for not less than 17 hours. After antimony per ml. Inject intravenously 0.3 ml of the solution
injections allow the mice access to food and water. None of into each of 10 mice that have been deprived of food for not
the mice dies within 24 hours. If one of the mice dies within less than 17 hours. After injections allow the mice access to
24 hours, repeat the test. None of the second group of mice food and water. None of the mice dies within 24 hours. If one
dies within 24 hours. of the mice dies within 24 hours, repeat the test. None of the
second group of mice dies within 24 hours.
Loss on drying (2.4.19). Not more than 15.0 per cent,
determined on 0.25 g by drying in an oven at 130º at a pressure Other tests. Complies with the tests stated under Parenteral
not exceeding 0.7 kPa. Preparations (Injections).
Assay. To 1.0 ml, accurately measured, add 30 ml of
Assay. Weigh accurately about 0.16 g, dissolve in 30 ml of
hydrochloric acid and 70 ml of phosphoric acid and mix.
hydrochloric acid, add 70 ml of phosphoric acid and stir
Titrate with 0.05 M ferric ammonium sulphate prepared using
carefully until completely mixed. Titrate with 0.05 M ferrous
sulphuric acid (1 per cent), determining the end-point
ammonium sulphate prepared using sulphuric acid (1 per
potentiometrically (2.4.25), using a platinum electrode and a
cent), determining the end-point potentiometrically (2.4.25),
silver-silver chloride reference electrode.
using a platinum electrode and a silver-silver chloride reference
electrode. 1 ml of 0.05 M ferric ammonium sulphate is equivalent to
0.003044 g of pentavalent antimony.
1 ml of 0.05 M ferric ammonium sulphate is equivalent to
0.003044 g of pentavalent antimony. Labelling. The label states the strength in terms of the
equivalent amount of pentavalent antimony per ml.
Storage. Store protected from moisture.

Sodium Thiosulphate
Sodium Stibogluconate Injection Sodium Hyposulphite
Sodium Antimony Gluconate Injection Na2S2O3,5H2O Mol. Wt. 248.2
Sodium Stibogluconate Injection is a sterile solution of Sodium Sodium Thiosulphate contains not less than 99.0 per cent
Stibogluconate in Water for Injections. Injection in multiple and not more than 101.0 per cent of Na2S2O3,5H2O.
dose containers must not contain phenol as preservative.
Description. Colourless large crystals or a coarse, crystalline
Sodium Stibogluconate Injection contains not less than powder; odourless; deliquescent in moist air and effloresces
9.5 per cent w/v and not more than 10.5 per cent w/v of in dry air at temperature above 33º. It dissolves in its water of
pentavalent antimony, Sb. crystallisation at about 49º.
Description. A clear, colourless or faintly straw-coloured
Identification
solution.
A. To 0.5 ml of a 10.0 per cent w/v solution in carbon dioxide-
Identification free water (solution A) add 0.5 ml of water and 2 ml of 0.1 M
silver nitrate; a white precipitate is produced which quickly
A. It is dextro-rotatory.
becomes yellowish and finally black.
B. Dilute 2 ml to 10 ml with water and pass hydrogen sulphide
B. To a portion of solution A add a few drops of iodine
through the solution; no immediate precipitate is produced.
solution; the colour is discharged.
Continue to pass hydrogen sulphide through the solution for
several minutes; an orange precipitate is produced. C. Dilute 2.5 ml of solution A to 5 ml with water and add 1 ml of
hydrochloric acid; a gas is evolved which turns starch-iodate
C. The residue obtained by evaporation to dryness, after
paper blue and a precipitate of sulphur is produced.
incineration, gives the reactions of antimony compounds and
the reactions of sodium salts (2.3.1). D. 1 ml of solution A gives reaction A of sodium salts (2.3.1).

1109
SODIUM THIOSULPHATE INJECTION IP 2007

Tests Identification
Appearance of solution. Solution A is clear (2.4.1), and A. To 0.5 ml of a 10.0 per cent w/v solution in carbon dioxide-
colourless (2.4.1). free water (solution A) add 0.5 ml of water and 2 ml of 0.1 M
silver nitrate; a white precipitate is produced which quickly
pH (2.4.24). 6.0 to 8.4, determined in solution A.
becomes yellowish and finally black.
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
of stannated hydrochloric acid AsT. The resulting solution B. To a portion of solution A add a few drops of iodine
complies with the limit test for arsenic (2 ppm). solution; the colour is discharged.
C. Dilute 2.5 ml of solution A to 5 ml with water and add 1 ml of
Heavy metals (2.3.13). Dissolve 1.0 g in 10 ml of water. Slowly
hydrochloric acid; a gas is evolved which turns starch-iodate
add 5 ml of dilute hydrochloric acid and evaporate the mixture
paper blue and a precipitate of sulphur is produced.
to dryness on a water-bath. Gently boil the residue with 15 ml
of water for 2 minutes and filter. Heat the filtrate to boiling, D. 1 ml of solution A gives reaction A of sodium salts (2.3.1).
add sufficient bromine solution to the hot filtrate to produce
a clear solution and add a slight excess of bromine solution. Tests
Boil the solution to expel the bromine completely, cool to room
pH (2.4.24). 7.0 to 9.0.
temperature and add a drop of phenolphthalein solution and
sodium hydroxide solution until a slight pink colour is Other tests. Complies with the tests stated under Parenteral
produced. Add 2 ml of dilute acetic acid and dilute with water Preparations (Injections).
to 25 ml. The solution complies with the limit test for heavy Assay. To an accurately measured volume containing about
metals, Method A ( 20 ppm). 0.5 g of Sodium Thiosulphate add about 20 ml of water and
Chlorides (2.3.12). To 12.5 ml of solution A add 15 ml of 2 M titrate with 0.05 M iodine, using 3 ml of starch solution, added
nitric acid, boil gently for 3 to 4 minutes, cool and filter. The towards the end of the titration, as indicator.
filtrate complies with the limit test for chlorides (200 ppm). 1 ml of 0.05 M iodine is equivalent to 0.02482 g of
Sulphides. To 10 ml of solution A add 0.05 ml of a freshly Na2S2O3,5H2O.
prepared 5 per cent w/v solution of sodium nitroprusside; the Storage. Store in single dose containers.
solution does not become violet.
Sulphates and sulphites (2.3.17). Dilute 2.5 ml of solution A to
10 ml with distilled water. To 3 ml of this solution add 2 ml of
iodine solution and gradually add more iodine solution and
Sodium Valproate
dilute to 15 ml with distilled water. The resulting solution
complies with the limit test for sulphates (0.2 per cent). H3 C
Assay. Weigh accurately about 0.5 g, dissolve in 20 ml of
H3 C COONa
water and titrate with 0.05 M iodine using starch solution,
added towards the end of the titration, as indicator. C8H15NaO2 Mol. Wt. 166.2
1 ml of 0.05 M iodine is equivalent to 0.02482 g of Sodium Valproate is sodium 2-propylpentanoate.
Na2S2O3,5H2O.
Sodium Valproate contains not less than 98.5 per cent and not
Storage. Store protected from moisture. more than 101.0 per cent of C8H15NaO2, calculated on the dried
basis.
Description. A white or almost white, crystalline powder;
Sodium Thiosulphate Injection hygroscopic.
Sodium Hyposulphite Injection Identification
Sodium Thiosulphate Injection is a sterile solution of Sodium A. Determine by infrared absorption spectrophotometry (2.4.6).
Thiosulphate in Water for Injections. Compare the spectrum with that obtained with sodium
Sodium Thiosulphate Injection contains not less than valproate RS or with the reference spectrum of sodium
95.0 per cent and not more than 105.0 per cent of the stated valproate.
amount of sodium thiosulphate, Na2S2O3,5H2O.
B. In the test for Related substances, the principal peak in the
Description. A clear, colourless solution. chromatogram obtained with test solution (b) corresponds to

1110
IP 2007 SODIUM VALPROATE ORAL SOLUTION

the peak in the chromatogram obtained with the reference inlet port at 200º and detector at 300º,
solution. – flow rate. 20 ml per minute of the carrier gas.
C. Dissolve 1.25 g in 20 ml of distilled water in a separating Allow the chromatography to proceed for 2.5 times the
funnel, add 5 ml of 2 M nitric acid, shake and allow the mixture retention time of valproic acid. Adjust the sensitivity so that
to stand for 12 hours; the lower layer (solution A) gives the height of the peak due to the internal standard in the
reactions of sodium salts (2.3.1). chromatogram obtained with test solution (a) is not less than
70 per cent of the full-scale deflection on the recorder. In the
Tests chromatogram obtained with test solution (c), the sum of the
areas of any secondary peaks is not greater than the area of
Appearance of solution. A 20.0 per cent w/v solution is not the peak due to the internal standard. Ignore any peaks the
more opalescent than opalescence standard OS2 (2.4.1), and area of which is less than 1 per cent of the area of the peak due
is not more intensely coloured than reference solution to the internal standard. The test is not valid unless the
YS6 (2.4.1). resolution between the peak due to the internal standard and
Acidity or alkalinity. To 10 ml of a 10.0 per cent w/v solution the principal peak in the chromatogram obtained with test
in carbon dioxide-free water add 0.1 ml of dilute solution (a) is at least 12.
phenolphthalein solution; not more than 0.75 ml of either Chlorides (2.3.12). Dissolve 1.25 g in 10 ml of water. The
0.1 M sodium hydroxide or 0.1 M hydrochloric acid is resulting solution complies with the limit test for chlorides
required to change the colour of the solution. (200 ppm).
Related substances. Determine by gas chromatography Sulphates (2.3.17). Solution A complies with the limit test for
(2.3.13). sulphates (200 ppm).
Test solution (a). Add 5 ml of 1 M sulphuric acid to 10 ml of a Heavy metals (2.3.13). 1.0 g complies with the limit test for
0.04 per cent w/v solution of the substance under examination heavy metals, Method B. Use 2 ml of lead standard solution
and shake with three quantities, each of 20 ml, of ether. Add (10 ppm) to prepare the standard (20 ppm).
10 ml of a 0.02 per cent w/v solution of butyric acid (internal
standard) in ether to the combined ether extracts, shake with Loss on drying (2.4.19). Not more than 2.0 per cent, determined
anhydrous sodium sulphate, filter and evaporate the filtrate on 1.0 g by drying in an oven at 105º.
to a volume of about 10 ml at a temperature not exceeding 30º Assay. Weigh accurately about 0.15 g and dissolve in 25 ml of
using a rotary evaporator. anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
Test solution (b). Add 0.5 ml of 1 M sulphuric acid to 10 ml of acid, determining the end-point potentiometrically (2.4.25).
a 0.04 per cent w/v solution of the substance under examination Carry out a blank titration.
and shake with three quantities, each of 5 ml, of ether. Shake 1 ml of 0.1 M perchloric acid is equivalent to 0.01662 g of
the combined ether extracts with anhydrous sodium sulphate, C8H15NaO2.
filter and evaporate the filtrate to a volume of about 10 ml at a
temperature not exceeding 30º using a rotary evaporator. Storage. Store protected from moisture.

Test solution (c). Dissolve 0.5 g of the substance under


examination in 10 ml of water, add 5 ml of 1 M sulphuric acid
and treat as described for test solution (a) beginning at the Sodium Valproate Oral Solution
words “shake with three...”.
Sodium Valproate Elixir
Reference solution. Prepare in the same manner as test solution
Sodium Valproate Oral Solution is a solution of Sodium
(b) but using sodium valproate RS in place of the substance
Valproate in a suitable flavoured vehicle.
under examination.
Sodium Valproate Oral Solution contains not less than
Chromatographic system
95.0 per cent and not more than 105.0 per cent of the stated
– a glass column 2.6 m x 2 mm, packed with silanised
amount of sodium valproate, C8H15NaO2.
diatomaceous support (125 to 180 mesh) impregnated
with 5 per cent w/w of polyethylene glycol 20,000 2- Identification
nitroterephthalate and 1 per cent w/w of phosphoric
acid, A. Shake a quantity containing about 0.25 g of Sodium
– temperature. column.150º to 170º to obtain a retention Valproate with two quantities, each of 25 ml, of chloroform
time of about 12 minutes for valproic acid [the principal and discard the chloroform extracts. Add 10 ml of a saturated
peak in test solution (b)], solution of sodium chloride and 10 ml of 2 M hydrochloric

1111
SODIUM VALPROATE TABLETS IP 2007

acid, mix and shake with 25 ml of chloroform. Wash the ether and 1 volume of light petroleum (40º to 60º), shake,
chloroform layer with 5 ml of water, shake with anhydrous allow to separate and reserve the ether layer. Shake the
sodium sulphate, filter and evaporate to dryness. aqueous layer with a further 40 ml of the ether-light petroleum
On the residue determine by infrared absorption mixture and discard the aqueous layer. Shake each of the
spectrophotometry (2.4.6). Compare the spectrum with that ether extracts with the same three quantities, each of 10 ml, of
obtained with valproic acid RS or with the reference spectrum a saturated solution of sodium chloride, combine the extracts
of valproic acid. and evaporate to a volume of about 1 ml at a temperature not
exceeding 30º. Add 50 ml of ethanol (95 per cent), previously
B. Shake a quantity containing about 0.25 g of Sodium neutralised with 0.01 M sodium hydroxide using dilute
Valproate with a mixture of 10 ml of a saturated solution of phenolphthalein solution as indicator, and titrate with 0.1 M
sodium chloride, 10 ml of 2 M hydrochloric acid and 25 ml of sodium hydroxide.
chloroform. Evaporate the chloroform layer to dryness,
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01662 g of
dissolve the residue in 2 ml of water, make just alkaline with
C8H15NaO2.
2 M sodium hydroxide and add 0.5 ml of a 10 per cent w/v
solution of cobalt nitrate; a purple precipitate is produced Determine the weight per ml of the preparation (2.4.29) and
which is soluble in dichloromethane. calculate the content of C8H15NaO2 weight in volume.

Tests
Related substances. Determine by gas chromatography Sodium Valproate Tablets
(2.4.13). Sodium Valproate Tablets contain not less than 95.0 per cent
Test solution (a). Mix a quantity of the oral solution containing and not more than 105.0 per cent of the stated amount of
0.50 g of Sodium Valproate with 10 ml of water, acidify with sodium valproate, C8H15NaO2.
2 M sulphuric acid and shake with three quantities, each of
20 ml, of dichloromethane. Wash the combined Identification
dichloromethane extracts with 10 ml of water, shake with A. Shake a quantity of the powdered tablets containing about
anhydrous sodium sulphate, filter and evaporate the filtrate 0.5 g of Sodium Valproate with 10 ml of water and centrifuge.
to a volume of about 10 ml at a temperature not exceeding 30º Acidify 5 ml of the supernatant liquid with 2 M sulphuric
using a rotary evaporator. acid, shake with 25 ml of chloroform and wash the chloroform
Test solution (b). Mix a quantity of the oral solution containing layer with 5 ml of water. Dry by shaking with anhydrous
0.5 g of Sodium Valproate with 10 ml of a 0.02 per cent w/v sodium sulphate, filter and evaporate the chloroform.
solution of octanoic acid (internal standard) in 0.1 M sodium On the residue determine by infrared absorption
hydroxide and continue as described for test solution (a) spectrophotometry (2.4.6). Compare the spectrum with that
beginning at the words “acidify with 2 M sulphuric acid...”. obtained with valproic acid RS or with the reference spectrum
Reference solution. A 0.02 per cent w/v solution of the internal of valproic acid.
standard in dichloromethane. B. Shake a quantity of the powdered tablets containing about
0.25 g of Sodium Valproate with 3 ml of water and centrifuge.
Chromatographic system
To 2 ml of the supernatant liquid add 0.5 ml of a 10 per cent
– a glass column 1.5 m x 4 mm. packed with acid-washed,
w/v solution of cobalt nitrate; a purple precipitate is produced
silanised diatomaceous support (80 to 180 mesh) coated
which is soluble in dichloromethane.
with 15 per cent w/w of free fatty acid phase (such as
Supelco FFAP 2-1063) and 1 per cent w/w of phosphoric Tests
acid,
– temperature. column. 170º. Related substances. Determine by gas chromatography
(2.4.13).
In the chromatogram obtained with test solution (b) the sum
of the areas of any secondary peaks is not greater than the Test solution (a). Shake a quantity of the powdered tablets
area of the peak due to the internal standard. containing 0.50 g of Sodium Valproate with 10 ml of water,
acidify with 2 M sulphuric acid and shake with three quantities,
Other tests. Complies with the tests stated under Oral Liquids. each of 20 ml, of dichloromethane. Wash the combined
Assay. Weigh accurately a quantity containing about 0.15 g dichloromethane extracts with 10 ml of water, shake with
of Sodium Valproate, add 50 ml of water, mix thoroughly and anhydrous sodium sulphate, filter and evaporate the filtrate
add 10 ml of saturated solution of sodium chloride, 10 ml of to a volume of about 10 ml at a temperature not exceeding 30º
2 M hydrochloric acid and 40 ml of a mixture of 2 volumes of using a rotary evaporator.

1112
IP 2007 SORBIC ACID

Test solution (b). Shake a quantity of the powdered tablets Description. A white or almost white, crystalline powder.
containing 0.50 g of Sodium Valproate with 10 ml of a 0.020 per
cent w/v solution of octanoic acid (internal standard) in Identification
0.1 M sodium hydroxide and continue as described for test
Test A may be omitted if tests B and C are carried out. Tests B
solution (a) beginning at the words “acidify with 2 M sulphuric
and C may be omitted if test A is carried out.
acid...”.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution. A 0.02 per cent w/v of the internal standard
Compare the spectrum with that obtained with sorbic acid RS
in dichloromethane.
or with the reference spectrum of sorbic acid.
Chromatographic system
B. Dissolve 50 mg in sufficient water to produce 250 ml and
– a glass column 1.5 m x 4 mm, packed with acid-washed,
dilute 2 ml of this solution to 200 ml with 0.1 M hydrochloric
silanised diatomaceous support (80 to 180 mesh) coated
acid.
with 15 per cent w/w of free fatty acid phase (such as
Supelco FFAP 2-1063) and 1 per cent w/w of phosphoric When examined in the range 230 nm to 360 nm (2.4.7), the
acid, resulting solution shows an absorption maximum at about
– temperature. column. 170º, 264 nm; absorbance at about 264 nm, 0.43 to 0.51.
In the chromatogram obtained with test solution (b) the sum C. Dissolve 0.2 g in 2 ml of ethanol (95 per cent) and add
of the areas of any secondary peaks is not greater than the 0.2 ml of bromine water; the solution is decolorised.
area of the peak due to the internal standard.
Tests
Other tests. Comply with the tests stated under Tablets.
Appearance of solution. A 5.0 per cent w/v solution in ethanol
Assay. Weigh and powder 20 tablets. Weigh accurately a
(95 per cent) is clear (2.4.1), and colourless (2.4.1).
quantity of the powder containing about 0.3 g of Sodium
Valproate, add 70 ml of water, shake for 10 minutes, dilute to Heavy metals (2.3.13). 2.0 g complies with the limit test for
100.0 ml with water and filter. To 50.0 ml of the filtrate add 10 ml heavy metals, Method A (10 ppm).
of a 30 per cent w/v solution of sodium chloride and 10 ml of Aldehydes. Not more than 0.15 per cent, determined by the
2 M hydrochloric acid. Extract with 40 ml of a mixture of following method. Dissolve 1.0 g in a mixture of 50 ml of
2 volumes of ether and 1 volume of light petroleum (boiling 2-propanol and 30 ml of water, adjust the pH of the solution
range 40º to 60º) and allow to separate. Shake the aqueous to 4.0 with 0.1 M hydrochloric acid or 0.1 M sodium
layer with a further 40 ml of the ether-light petroleum mixture. hydroxide and dilute to 100 ml with water. To 10 ml of the
Shake each of the ether extracts with the same three quantities, resulting solution add 1 ml of decolorised fuchsin solution
each of 10 ml, of a saturated solution of sodium chloride, and allow to stand for 30 minutes. Any colour produced is not
combine the ether extracts and evaporate to a volume of about more intense than that obtained in a solution prepared
1 ml at a temperature not exceeding 30°. Add 50 ml of ethanol simultaneously by adding 1 ml of decolorised fuchsin solution
(95 per cent), previously neutralised with 0.01 M sodium to a mixture of 1.5 ml of acetaldehyde standard solution (100
hydroxide using dilute phenolphthalein solution as indicator, ppm C2H4O), 4 ml of 2-propanol and 4.5 ml of water.
and titrate with 0.1 M sodium hydroxide.
Sulphated ash (2.3.18). Not more than 0.2 per cent.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01662 g of
C8H15NaO2. Water (2.3.43). Not more than 1.0 per cent, determined on
2.0 g.
Assay. Weigh accurately about 0.2 g, dissolve in 20 ml of
ethanol (95 per cent) and titrate with 0.1 M sodium hydroxide,
Sorbic Acid using 0.2 ml of phenolphthalein solution as indicator, until a
pink colour is produced.
COOH
H3 C 1 ml of 0.1 M sodium hydroxide is equivalent to 0.01121 g of
C6H8O2.
C6H8O2 Mol. Wt. 112.1 Storage. Store protected from light and moisture.
Sorbic Acid is (2E,4E)-hexa-2,4-dienoic acid.
Sorbic Acid contains not less than 99.0 per cent and not more
than 101.0 per cent of C6H8O2, calculated on the anhydrous
basis.

1113
SORBITOL IP 2007

Sorbitol after washing rapidly with 5 ml of a mixture of equal volumes


of methanol and water and drying in a current of air, melts at
D-Glucitol about 175º (2.4.21).

CH2OH Tests
H C OH
Appearance of solution. Solution A is clear (2.4.1), and
HO C H colourless (2.4.1).
H C OH
Acidity or alkalinity. Dissolve 5.0 g in sufficient carbon
H C OH dioxide-free water prepared from distilled water to produce
CH2OH 50 ml (solution A). To 10 ml of solution A add 10 ml of carbon
dioxide-free water. To 10 ml of the resulting solution add
C6H14O6 Mol. Wt. 182.2 0.05 ml of phenolphthalein solution; not more than 0.2 ml of
Sorbitol is D-glucitol, a hexahydric alcohol related to 0.01 M sodium hydroxide is required to change the colour of
glucose. the solution to pink. To a further 10 ml of the solution add
0.05 ml of methyl red solution; Not more than 0.3 ml of 0.01 M
Sorbitol contains not less than 98.0 per cent and not more hydrochloric acid is required to change the colour of the
than 101.0 per cent of C6H14O6, calculated on the anhydrous solution to red.
basis.
Specific optical rotation (2.4.22). +4.0º to +7.0º, determined in
Description. A white, crystalline powder; odourless. a solution prepared in the following manner. Dissolve a mixture
of 5.0 g of the substance under examination and 6.4 g of borax
Identification
in 40 ml of water, allow to stand for 1 hour, shaking occasionally,
A. Determine by thin-layer chromatography (2.4.17), coating dilute to 50 ml with water and filter, if necessary.
the plate with a uniform 0.75-mm thick layer of the following Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
mixture. Mix 0.1 g of carbomer with 110 ml of water and allow of stannated hydrochloric acid AsT. The resulting solution
to stand, with gentle stirring, for 1 hour. Adjust to pH 7.0 by complies with the limit test for arsenic (2 ppm).
the gradual addition, with continuous shaking, of 2 M sodium
hydroxide and add 30 g of silica gel H. Heat the plate at 110º Heavy metals (2.3.13). 2.0 g complies with the limit test for
for 1 hour, allow to cool and use immediately. heavy metals, Method A (10 ppm).
Mobile phase. A mixture of 85 volumes of 2-propanol and Chlorides (2.3.12). 5.0 g complies with the limit test for chlorides
15 volumes of a 0.2 per cent w/v solution of boric acid. (50 ppm).
Test solution. Dissolve 0.25 g of the substance under Nickel. Dissolve 10.0 g in sufficient water to produce 20 ml,
examination in 100 ml of ethanol (95 per cent). add 3 ml of bromine water and 2 ml of a 20 per cent w/v
solution of citric acid, mix and add 10 ml of 6 M ammonia and
Reference solution. A 0.25 per cent w/v solution of sorbitol 1 ml of a 1 per cent w/v solution of dimethylglyoxime in ethanol
RS in ethanol (95 per cent). (95 per cent). Mix, dilute to 50 ml with water and allow to
Apply to the plate 2 µl of each solution. After development, stand for 5 minutes; any colour produced is not more intense
dry the plate at 100º to 105º for 15 minutes, allow to cool, spray than that produced by treating in the same manner and at the
with a 0.5 per cent w/v solution of potassium permanganate same time 1.0 ml of nickel standard solution (10 ppm Ni)
in 1 M sodium hydroxide and heat at 100º for 2 minutes. The diluted to 20 ml with water (1 ppm).
principal spot in the chromatogram obtained with the test Sulphates (2.3.17). 12 ml of solution A complies with the limit
solution corresponds to that in the chromatogram obtained test for sulphates (125 ppm).
with the reference solution.
Reducing sugars. Dissolve 5.0 g in 3 ml of water with the aid
B. Dissolve 50 mg in 3 ml of water, add 3 ml of catechol solution of gentle heat, cool, add 20 ml of cupri-citric solution and a
and pour the mixture into 6 ml of sulphuric acid; a pink colour few glass beads, heat in such a manner that the solution boils
is produced. in 4 minutes and continue boiling for a further 3 minutes. Cool
C. Dissolve 5 g in 3 ml of water with the aid of gentle heat, rapidly and add 100 ml of a 2.4 per cent v/v solution of glacial
cool, add 7 ml of methanol, 1 ml of benzaldehyde and 1 ml of acetic acid followed by 20.0 ml of 0.025 M iodine. Add,
hydrochloric acid, mix and shake continuously for 2 hours. shaking continuously, 25 ml of a 6 per cent v/v solution of
Filter, dissolve the crystals in 20 ml of boiling sodium hydrochloric acid and, when any precipitate has redissolved,
bicarbonate solution and allow to crystallise. The residue, titrate the excess iodine with 0.05 M sodium thiosulphate

1114
IP 2007 SORBITOL SOLUTION (70 PER CENT) (CRYSTALLISING)

using 1 ml of starch solution, added towards the end of the B. Dry 1 g over phosphorus pentoxide at a pressure of 1.5 to
titration, as indicator. Not less than 12.8 ml of 0.05 M sodium 2.5 kPa. Heat 0.5 g of the residue with a mixture of 5 ml of
thiosulphate is required. acetic anhydride and 0.5 ml pyridine with the aid of heat and
Sulphated ash (2.3.18). Not more than 0.1 per cent. allow to stand for 10 minutes. Pour the mixture into 25 ml of
water, allow to stand in ice for 2 hours and filter. The precipitate,
Water (2.3.43). Not more than 1.5 per cent, determined on after recrystallisation from a small volume of ethanol (95 per
1.0 g. cent) and drying over phosphorus pentoxide at a pressure of
Assay. Weigh accurately about 0.4 g and dissolve in sufficient 1.5 to 2.5 kPa, melts at about 100º (2.4.21).
water to produce 100.0 ml. To 10.0 ml of the solution add 20 ml C. Determine by thin-layer chromatography (2.4.17), coating
of a 2.14 per cent w/v solution of sodium periodate and 2 ml of the plate with a uniform 0.75-mm thick layer of the following
1 M sulphuric acid and heat on a water-bath for exactly mixture. Mix 0.1 g of carbomer with 110 ml of water and allow
15 minutes. Cool, add 3 g of sodium bicarbonate, in small to stand, with gentle stirring, for 1 hour. Adjust to pH 7.0 by
quantities, and 25.0 ml of 0.1 M sodium arsenite, mix, add 5 ml the gradual addition, with continuous shaking, of 2 M sodium
of a 20 per cent w/v solution of potassium iodide and allow to hydroxide and add 30 g of silica gel H. Heat the plate at 110°
stand for 15 minutes. Titrate with 0.05 M iodine until the first for 1 hour, allow to cool and use immediately.
trace of yellow colour appears. Repeat the operation without Mobile phase. A mixture of 85 volumes of 2-propanol and
the substance under examination. The difference between the 15 volumes of a 0.2 per cent w/v solution of boric acid.
titrations represents the amount of iodine required.
Test solution. Dissolve 0.35 g of the substance under
1 ml of 0.05 M iodine is equivalent to 0.001822 g of C6H14O6. examination in 100 ml of ethanol (95 per cent).
Sorbitol intended for use in the manufacture of parenteral Reference solution. A 0.25 per cent w/v solution of sorbitol
preparations without a further appropriate procedure for RS in ethanol (95 per cent).
removal of bacterial endotoxins complies with the following
Apply to the plate 2 µl of each solution. After development,
additional requirement.
dry the plate at 100º to 105º for 15 minutes, allow to cool, spray
Bacterial endotoxins (2.2.3). Not more than 4.0 Endotoxin Units with a 0.5 per cent w/v solution of potassium permanganate
per g for parenteral preparations having a concentration of in 1 M sodium hydroxide and heat at 100º for 2 minutes. The
less than 10 per cent w/v of sorbitol and not more than 2.5 principal spot in the chromatogram obtained with the test
Endotoxin Units per g for parenteral preparations containing solution corresponds to that in the chromatogram obtained
10 per cent w/v or more of sorbitol. with the reference solution.
Storage. Store protected from moisture.
Tests
Labelling. The label states whether or not the contents are
intended for use in the manufacture of parenteral preparations. Appearance of solution. A 14.0 per cent w/v solution in carbon
dioxide–free water (solution A) is clear (2.4.1), and colourless
(2.4.1).
Sorbitol Solution (70 Per Cent) Acidity or alkalinity. To 10 ml of solution A add 0.05 ml of
phenolphthalein solution; not more than 0.2 ml of 0.01 M
(Crystallising) sodium hydroxide is required to change the colour of the
Sorbitol (70 Per cent) (Crystallising) solution to pink. To a further 10 ml of the solution add 0.05 ml
of methyl red solution. Not more than 0.3 ml of 0.01 M
Sorbitol Solution (70 per cent) (Crystallising) is an aqueous hydrochloric acid is required to change the colour of the
solution of hexitols. solution to red.
Sorbitol Solution ( 70 per cent) (Crystallising) contains not Refractive index (2.4.27). 1.457 to 1.462.
less than 68.0 per cent w/w and not more than 72.0 per cent
Relative density (2.4.29). Not less than 1.290.
w/w of hexitols, calculated as D-glucitol (C6H14O6).
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
Description. A clear, colourless, syrupy liquid.
of stannated hydrochloric acid AsT. The resulting solution
Identification complies with the limit test for arsenic (2 ppm).
Heavy metals (2.3.13). 2.0 g complies with the limit test for
A. Dilute 7.0 g with 40 ml of water, add 6.4 g of borax, allow to
heavy metals, Method A (10 ppm).
stand for 1 hour, shaking occasionally, and dilute to 50.0 ml
with water. Filter if necessary. The optical rotation of the Chlorides (2.3.12). 5.0 g complies with the limit test for chlorides
resulting solution is 0º to +1.5º (2.4.22). (50 ppm).

1115
SORBITOL SOLUTION (70 PER CENT) (NON-CRYSTALLISING) IP 2007

Nickel. Dissolve 10.0 g in sufficient water to produce 20 ml, cent w/w of solid matter and not less than 62.0 per cent w/w of
add 3 ml of bromine water and 2 ml of a 20 per cent w/v polyols expressed as D-glucitol (C6H14O6).
solution of citric acid, mix and add 10 ml of 6 M ammonia and Description. A clear, colourless or faintly yellow, syrupy liquid;
1 ml of a 1 per cent w/v solution of dimethylglyoxime in ethanol odourless.
(95 per cent). Mix, dilute to 50 ml with water and allow to
stand for 5 minutes; any colour produced is not more intense Identification
than that produced by treating in the same manner and at the
same time 1.0 ml of nickel standard solution (10 ppm Ni) A. Dilute 7.0 g with sufficient carbon dioxide-free water to
diluted to 20 ml with water (1 ppm). produce 50 ml (solution A). To 3 ml of a freshly prepared 10 per
cent w/v solution of catechol add 6 ml of sulphuric acid while
Sulphates (2.3.17). 12 ml of solution A complies with the limit cooling in ice. To 3 ml of the mixture add 0.3 ml of solution A
test for sulphates (125 ppm). and heat gently over a naked flame for 30 seconds; a pink
Reducing sugars. Dissolve 5.0 g in 3 ml of water with the aid colour is produced which becomes deep brownish red.
of gentle heat, cool, add 20 ml of cupri-citric solution and a B. Dry 1 g over phosphorus pentoxide at 80º at a pressure of
few glass beads, heat in such a manner that the solution boils 1.5 to 2.5 kPa. Dissolve 0.5 g of the residue in a mixture of 5 ml
in 4 minutes and continue boiling for a further 3 minutes. Cool of acetic anhydride and 0.5 ml of pyridine with the aid of heat
rapidly and add 100 ml of a 2.4 per cent v/v solution of glacial and allow to stand for 10 minutes. Pour the mixture into 25 ml
acetic acid followed by 20.0 ml of 0.025 M iodine. Add, of water, allow to stand in ice for 2 hours and filter. The residue,
shaking continuously, 25 ml of a 6 per cent v/v solution of after recrystallisation from a small volume of ethanol (95 per
hydrochloric acid and, when any precipitate has redissolved, cent) and drying over phosphorus pentoxide at a pressure of
titrate the excess iodine with 0.05 M sodium thiosulphate 1.5 to 2.5 kPa, melts at about 100º (2.4.21).
using 1 ml of starch solution, added towards the end of the
titration, as indicator. Not less than 12.8 ml of 0.05 M sodium C. Determine by thin-layer chromatography (2.4.17), coating
thiosulphate is required. the plate with a uniform 0.75-mm thick layer of the following
mixture. Mix 0.1 g of carbomer with 110 ml of water and allow
Sulphated ash (2.3.18). Not more than 0.1 per cent. to stand, with gentle stirring, for 1 hour. Adjust to pH 7.0 by
Assay. Measure accurately a volume containing about 0.6 g the gradual addition, with continuous shaking, of 2 M sodium
of Sorbitol and dissolve in sufficient water to produce hydroxide and add 30 g of silica gel H. Heat the plate at 110º
100.0 ml. To 10.0 ml of the solution add 20 ml of a 2.14 per cent for 1 hour, allow to cool and use immediately.
w/v solution of sodium periodate and 2 ml of 1 M sulphuric Mobile phase. A mixture of 85 volumes of 2-propanol and
acid and heat on a water-bath for exactly 15 minutes. Cool, 15 volumes of a 0.2 per cent w/v solution of boric acid.
add 3 g of sodium bicarbonate, in small quantities, and
25.0 ml 0.1 M sodium arsenite, mix, add 5 ml of a 20 per cent Test solution. Dissolve 0.35 g of the substance under
w/v solution of potassium iodide and allow to stand for examination in 100 ml of ethanol (95 per cent).
15 minutes. Titrate with 0.05 M iodine until the first trace of Reference solution. A 0.25 per cent w/v solution of sorbitol
yellow colour appears. Repeat the operation without the RS in ethanol (95 per cent).
substance under examination. The difference between the
titrations represents the amount of iodine required. Apply to the plate 2 µl of each solution. After development,
dry the plate at 100º to 105º for 15 minutes, allow to cool, spray
1 ml of 0.05 M iodine is equivalent to 0.001822 g of C6H14O6. with a 0.5 per cent w/v solution of potassium permanganate
Storage. Store protected from moisture. in 1 M sodium hydroxide and heat at 100º for 2 minutes. The
principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
with the reference solution.

Sorbitol Solution (70 Per Cent) (Non- Tests


Crystallising) Appearance of solution. Solution A is clear (2.4.1), and
colourless (2.4.1).
Sorbitol (70 per cent) (Non-crystallising)
Acidity or alkalinity. To 10 ml of solution A add 10 ml of
Sorbitol Solution (70 per cent) (Non-crystallising) is an aqueous carbon dioxide-free water. To 10 ml of the resulting solution
solution of hydrogenated, partly hydrolysed starch. add 0.05 ml of phenolphthalein solution; not more than 0.2 ml
Sorbitol Solution (70 per cent) (Non-crystallising) contains of 0.01 M sodium hydroxide is required to change the colour
not less than 68.0 per cent w/w and not more than 72.0 per of the solution to pink. To a further 10 ml of the solution add

1116
IP 2007 SPIRONOLACTONE

0.05 ml of methyl red solution; Not more than 0.3 ml of 0.01 M add 3 g of sodium bicarbonate, in small quantities, and
hydrochloric acid is required to change the colour of the 25.0 ml of 0.1 M sodium arsenite, mix, add 5 ml of a 20 per cent
solution to red. w/v solution of potassium iodide and allow to stand for
Optical rotation (2.4.22). +1.5º to + 3.5º, determined in a solution 15 minutes. Titrate with 0.05 M iodine until the first trace of
prepared in the following manner. Dilute 7.0 g with 40 ml of yellow colour appears. Repeat the operation without the
water, add 6.4 g of borax, allow to stand for 1 hour, shaking substance under examination. The difference between the
occasionally, dilute to 50 ml with water and filter, if necessary. titrations represents the amount of iodine required.

Refractive index (2.4.27). 1.455 to 1.465. 1 ml of 0.05 M iodine is equivalent to 0.001822 g of C6H14O6.

Relative density (2.4.29). Not less than 1.285. Storage. Store protected from moisture.
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml
of stannated hydrochloric acid AsT. The resulting solution
complies with the limit test for arsenic (2 ppm).
Spironolactone
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method A (10 ppm).
Chlorides (2.3.12). 5.0 g complies with the limit test for chlorides H3C O
(50 ppm). O
H3C H
Nickel. Dissolve 10.0 g in sufficient water to produce 20 ml,
add 3 ml of bromine water and 2 ml of a 20 per cent w/v H H
solution of citric acid, mix and add 10 ml of 6 M ammonia and
O S
1 ml of a 1 per cent w/v solution of dimethylglyoxime in ethanol
(95 per cent). Mix, dilute to 50 ml with water and allow to O CH3
stand for 5 minutes; any colour produced is not more intense
than that produced by treating in the same manner and at the
same time 1.0 ml of nickel standard solution (10 ppm Ni) C24H32O4S Mol. Wt. 416.6
diluted to 20 ml with water (1 ppm).
Spironolactone is 7α-acetylthio-3-oxo-17α-pregn-4-ene-
Sulphates (2.3.17). 12 ml of solution A complies with the limit 21,17β-carbolactone.
test for sulphates (125 ppm).
Spironolactone contains not less than 97.0 per cent and not
Reducing sugars. Dissolve 5.0 g in 3 ml of water with the aid more than 102.0 per cent of C24H32O4S, calculated on the dried
of gentle heat, cool, add 20 ml of cupri-citric solution and a basis.
few glass beads, heat in such a manner that the solution boils
in 4 minutes and continue boiling for a further 3 minutes. Cool Description. A yellowish white to buff coloured powder;
rapidly and add 100 ml of a 2.4 per cent v/v solution of glacial odourless or with a slight odour of thioacetic acid. It exhibits
acetic acid followed by 20.0 ml of 0.025 M iodine. Add, polymorphism.
shaking continuously, 25 ml of a 6 per cent v/v solution of
hydrochloric acid and, when any precipitate has redissolved, Identification
titrate the excess iodine with 0.05 M sodium thiosulphate
Test A may be omitted if tests B and C are carried out. Test B
using 1 ml of starch solution, added towards the end of the
may be omitted if tests A and C are carried out.
titration, as indicator. Not less than 12.8 ml of 0.05 M sodium
thiosulphate is required. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with spironolactone
Sulphated ash (2.3.18). Not more than 0.1 per cent.
RS or with the reference spectrum of spironolactone. Examine
Assay. For solid matter — Weigh accurately about 1.0 g and the substances as 5 per cent w/v solutions in chloroform.
dry to constant weight over phosphorus pentoxide at 80º at a
B. Determine by thin-layer chromatography (2.4.17), coating
pressure of 1.5 to 2.5 kPa. Weigh the residue.
the plate with silica gel G.
For polyols — Measure accurately a volume containing about
Solvent mixture. A mixture of 90 volumes of acetone and
0.4 g of Sorbitol and dissolve in sufficient water to produce
10 volumes of 1,2-propanediol.
100.0 ml. To 10.0 ml of the solution add 20 ml of a 2.14 per cent
w/v solution of sodium periodate and 2 ml of 1 M sulphuric Mobile phase. A mixture of 40 volumes of cyclohexane and 10
acid and heat on a water-bath for exactly 15 minutes. Cool, volumes of toluene.

1117
SPIRONOLACTONE TABLETS IP 2007

Test solution. Dissolve 25 mg of the substance under iodine and 0.1 ml of starch solution and mix; a blue colour
examination in 10 ml of the solvent mixture. develops.
Reference solution (a). Dissolve 25 mg of spironolactone RS Sulphated ash (2.3.18). Not more than 0.1 per cent.
in 10 ml of the solvent mixture.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Reference solution (b). Mix equal volumes of the test solution on 1.0 g by drying in an oven at 105º for 3 hours,
and reference solution (a).
Assay. Weigh accurately about 50 mg, dissolve in sufficient
Place the dry plate in a tank containing a shallow layer of the methanol to produce 250.0 ml, dilute 5.0 ml to 100.0 ml with
solvent mixture, allow the solvent mixture to ascend to the methanol and measure the absorbance of the resulting
top, remove the plate from the tank and allow the solvent to solution at the maximum at about 238 nm (2.4.7).
evaporate. Use within 2 hours, with the flow of the mobile
Calculate the content of C24H32O4S taking 470 as the specific
phase in the direction in which the aforementioned treatment
absorbance at 238 nm.
was done.
Storage. Store protected from light and moisture.
Apply to the plate 2 µl of each solution. Allow the mobile
phase to rise 12 cm. Dry the plate in a current of warm air, allow
the solvent to evaporate, heat at 120º for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120º for a further 10 minutes, allow to cool and examine Spironolactone Tablets
in daylight and in ultraviolet light at 365 nm. The principal
Spironolactone Tablets contain not less than 95.0 per cent
spot in the chromatogram obtained with the test solution
and not more than 105.0 per cent of the stated amount of
corresponds to that in the chromatogram obtained with
spironolactone, C24H32O4S.The tablets may contain added
reference solution (a). The principal spot in the chromatogram
flavouring agents and may be coated.
obtained with reference solution (b) appears as a single,
compact spot.
Identification
C. Shake about 10 mg with 2 ml of sulphuric acid
(50 per cent); an orange solution with an intense yellowish A. Extract a quantity of the powdered tablets containing
fluorescence is produced. Heat the solution gently; the colour 0.125 g of Spironolactone with two quantities, each of 10 ml,
becomes deep red and hydrogen sulphide is evolved which of chloroform, filter, evaporate the combined filtrates to
turns lead acetate paper black. Add the solution to 10 ml of dryness and dissolve the residue in 2.5 ml of chloroform.
water; a greenish yellow solution is produced which shows On the resulting solution determine by infrared absorption
opalescence or a precipitate. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with spironolactone RS or with the reference
Tests
spectrum of spironolactone.
Specific optical rotation (2.4.22). –33.0º to –37.0º, determined B. Determine by thin-layer chromatography (2.4.17), coating
in a 1.0 per cent w/v solution in chloroform. the plate with silica gel G.
Chromium. Mix 0.2 g with 1 g of potassium carbonate and Solvent mixture. A mixture of 90 volumes of acetone and 10
0.3 g of potassium nitrate in a platinum crucible, heat gently volumes of 1,2-propanediol.
until fused and ignite at 600º to 650º until the carbon is removed.
Cool, dissolve the residue as completely as possible in 10 ml Mobile phase. A mixture of 40 volumes of cyclohexane and 10
of water with the aid of gentle heat, filter and dilute to 20 ml volumes of toluene.
with water. To 10 ml of the solution add 0.5 g of urea and just Test solution. Extract a quantity of the powdered tablets
acidify with sulphuric acid (14 per cent). When effervescence containing 50 mg of Spironolactone with 10 ml of chloroform,
ceases add a further 1 ml of sulphuric acid (14 per cent), filter and evaporate the filtrate to dryness. Dissolve 25 mg the
dilute to 20 ml with water and add 0.5 ml of diphenylcarbazide residue in 10 ml of the solvent mixture.
solution. Any colour produced is not more intense than that
obtained by adding 1 ml of sulphuric acid (14 per cent) to Reference solution (a). Dissolve 25 mg of spironolactone RS
0.5 ml of a freshly prepared 0.00283 per cent w/v solution of in 10 ml of the solvent mixture.
potassium dichromate, diluting to 20 ml with water and adding Reference solution (b). Mix equal volumes of the test solution
0.5 ml of diphenylcarbazide solution (50 ppm). and reference solution (a).
Free mercapto compounds. Shake 2.0 g with 20 ml of water for Place the dry plate in a tank containing a shallow layer of the
1 minute and filter. To 10 ml of the filtrate add 0.05 ml of 0.01 M solvent mixture, allow the solvent mixture to ascend to the

1118
IP 2007 STAVUDINE

top, remove the plate from the tank and allow the solvent to Stavudine
evaporate. Use within 2 hours, with the flow of the mobile
phase in the direction in which the aforementioned treatment
O
was done.
CH3
Apply to the plate 2 µl of each solution. Allow the mobile HN
phase to rise 12 cm. Dry the plate in a current of warm air, allow
the solvent to evaporate, heat at 120º for 15 minutes and spray O N
O
the hot plate with ethanolic sulphuric acid (20 per cent v/v). HO
Heat at 120º for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution C10H12N2O4 Mol. Wt. 224.2
corresponds to that in the chromatogram obtained with Stavudine is 2',3',didehydro-3'-deoxythymidine.
reference solution (a). The principal spot in the chromatogram
obtained with reference solution (b) appears as a single, Stavudine contains not less than 98.0 per cent and not more
compact spot. than 102.0 per cent of C10H12N2O4, calculated on the dried
basis.
C. Shake about 10 mg of the residue obtained in test B with
Description. A white or almost white powder.
2 ml of sulphuric acid (50 per cent); an orange solution with
an intense yellowish fluorescence is produced. Heat the Identification
solution gently; the colour becomes deep red and hydrogen
sulphide is evolved which turns lead acetate paper black. A. Determine by infrared absorption spectrophotometry (2.4.6).
Add the solution to 10 ml of water; a greenish yellow solution Compare the spectrum with that obtained with stavudine RS
is produced which shows opalescence or a precipitate. or with the reference spectrum of stavudine.
B. In the Assay, the principal peak in the chromatogram
Tests obtained with the test solution corresponds to the peak due
Dissolution (2.5.2). to stavudine in the chromatogram obtained with reference
solution (c).
Apparatus. No 1
C. Melts at about 164º (2.4.21).
Medium. 1000 ml of 0.1 M hydrochloric acid containing
0.1 per cent w/v of sodium dodecyl sulphate. Tests
Speed and time. 75 rpm and 60 minutes.
Specific optical rotation (2.4.22). –39.0º to – 46.0º, determined
Withdraw a suitable volume of the medium, filter through a in a 0.7 per cent w/v solution in water.
membrane filter disc with an average pore diameter not greater
than 1.0 µm. Measure the absorbance of the filtrate, suitably Related substances. Determine by liquid chromatography
diluted if necessary, at the maximum at about 242 nm (2.4.7). (2.4.14), as described in the Assay.
Calculate the content of C24H32O4S in the medium taking 445 Separately inject the test solution, reference solutions (a) and
as the specific absorbance at 242 nm. (b) and record the chromatograms for at least three times the
retention time of the principal peak. In the chromatogram
D. Not less than 70.0 per cent of the stated amount of C24H32O4S.
obtained with the test solution, the area of any peak
Other tests. Comply with the tests stated under Tablets. corresponding to thymine is not greater than the area of the
peak in the chromatogram obtained with reference solution
Assay. Weigh and powder 20 tablets. Weigh accurately a
(b) (1 per cent); the area of any peak corresponding to α-
quantity of the powder containing about 25 mg of
thymidine is not greater than the area of the corresponding
Spironolactone, add 100 ml of methanol and heat just to peak in the chromatogram obtained with reference solution
boiling, with swirling. Cool, add sufficient methanol to produce (a) (1 per cent); the area of any individual impurity peak other
250.0 ml, dilute 10.0 ml to 100.0 ml with methanol and measure than thymine and α-thymidine is not more than 0.5 per cent
the absorbance of the resulting solution at the maximum at and the sum of the areas of all the impurity peaks other than
about 238 nm (2.4.7). thymine and α-thymidine is not more than 1.0 per cent.
Calculate the content of C24H32O4S taking 470 as the specific Heavy metals (2.3.13). 1.0 g complies with the limit test for
absorbance at 238 nm. heavy metals, Method B (20 ppm).
Storage. Store protected from light and moisture. Sulphated ash (2.3.18). Not more than 0.3 per cent.

1119
STAVUDINE CAPSULES IP 2007

Loss on drying (2.4.19). Not more than 1.0 per cent, determined When examined in the range 200 nm to 300 nm (2.4.7), the
on 1.0 g by drying at 105º for 3 hours. resulting solution shows an absorption maximum at about
Assay. Determine by liquid chromatography (2.4.14). 265 nm.
B. In the Assay, the principal peak in the chromatogram
Test solution. Dissolve 50.0 mg of the substance under
obtained with the test solution corresponds to the peak due
examination in 100.0 ml of the mobile phase.
to stavudine in the chromatogram obtained with the reference
Reference solution (a). A solution containing 0.05 per cent solution.
w/v of stavudine RS, 0.01 per cent w/v of thymine and
0.0005 per cent w/v of â-thymidine in the mobile phase. Tests
Reference solution (b). A 0.0005 per cent w/v solution of Related substances. Determine by liquid chromatography
thymine in the mobile phase. (2.4.14).
Reference solution (c). A 0.05 per cent w/v solution of Test solution. Mix well the contents of 20 capsules and transfer
stavudine RS in the mobile phase. an accurately weighed quantity containing about 50 mg of
Chromatographic system Stavudine to a 100-ml volumetric flask. Add about 60 ml of
– a stainless steel column 25 cm x 4.6 mm, packed with water, mix with the aid of ultrasound for 10 minutes, dilute to
octadecylsilane bonded to porous silica (5 µm), volume with water, mix and filter.
– column temperature 50º, Reference solution. Weigh 5 mg each of stavudine RS and
– mobile phase: a mixture of 20 volumes of methanol and thymine RS and transfer to a 25-ml volumetric flask. Add 5 ml
80 volumes of water, of methanol, mix with the aid of ultrasound to dissolve and
– flow rate. 1 ml per minute, dilute to volume with water.
– spectrophotometer set at 265 nm,
Chromatographic system
– a 20 µl loop injector.
– a stainless steel column 25 cm x 4.6 mm, packed with
Inject separately reference solutions (a) and (c). The order of octylsilane bonded to porous silica (5 µm),
elution in the chromatogram obtained with reference solution – mobile phase: gradient mixtures of acetonitrile and
(a) is thymine, α-thymidine and stavudine with relative 0.1 M ammonium acetate,
retention times of about 0.5, 0.7 and 1.0 respectively. The test – flow rate. 1 ml per minute,
is not valid unless the column efficiency determined from the – a linear gradient programme using the conditions given
stavudine peak in the chromatogram obtained with reference below,
solution (a) is not less than 3000 theoretical plates, the tailing – spectrophotometer set at 270 nm,
factor is not more than 1.5 and the relative standard deviation – a 20 µl loop injector.
for replicate injections of reference solution (c) is not more Time 0.1 M Ammonium acetate Acetonitrile
than 2.0 per cent. (in min.) (per cent v/v) (per cent v/v)
Inject separately the test solution and reference solution (c) 0 95 5
and measure the responses of the principal peak. 5 95 5
Calculate the content of C10H12N2O4. 25 20 80
Storage. Store protected from light and moisture. 30 20 80
31 95 5
35 95 5
Stavudine Capsules Inject the reference solution. The test is not valid unless the
Stavudine Capsules contain not less than 90.0 per cent and column efficiency determined from the peaks of stavudine
not more than 110.0 per cent of the stated amount of stavudine, and thymine is not less than 7000 theoretical plates and the
C10H12N2O4. tailing factor is not more than 2.0.
Inject separately water and the test solution. Examine the
Identification water chromatogram for any extraneous peaks and ignore the
A.Shake a quantity of the mixed contents of the capsules corresponding peaks observed in the chromatogram obtained
containing about 50 mg of Stavudine with 80 ml of water for with the test solution.
10 minutes. Add sufficient water to produce 100 ml, mix and Any secondary peak observed in the chromatogram obtained
filter. Dilute 10 ml of the filtrate to 100 ml with water. with the test solution corresponding to thymine is not more

1120
IP 2007 STAVUDINE ORAL SOLUTION

than 3.0 per cent. Any other secondary peak observed in the volume with water, mix and filter. Further dilute 10.0 ml of the
chromatogram obtained with the test solution is not more filtrate to 25.0 ml with water.
than 0.5 per cent and the sum of the areas of all the secondary Reference solution. Weigh accurately about 50 mg of stavudine
peaks is not more than 4.0 per cent when calculated by RS and transfer to a 100-ml volumetric flask. Add about 60 ml
percentage area normalisation. of water, mix with the aid of ultrasound to dissolve and dilute
Dissolution (2.5.2). to volume with water. Dilute 10.0 ml of this solution to 25.0 ml
with water.
Apparatus. No 1
Medium. 900 ml of 0.01 M hydrochloric acid Chromatographic system
Speed and time. 75 rpm and 45 minutes. – a stainless steel column 25 cm x 4.6 mm, packed with
octylsilane bonded to porous silica (5 µm),
Withdraw a suitable volume of the medium and filter.
– mobile phase: a mixture of 5 volumes of acetonitrile
Determine by liquid chromatography (2.4.14). and 95 volumes of 0.1 M ammonium acetate,
– flow rate. 1 ml per minute,
Test solution. The filtrate obtained as given above.
– spectrophotometer set at 270 nm,
Reference solution. In the case of capsules containing 30 mg – a 20 µl loop injector
of Stavudine, weigh accurately about 30 mg of stavudine RS
and transfer to a 100-ml volumetric flask. Dissolve and dilute Inject the reference solution. The test is not valid unless the
to volume with 0.01 M hydrochloric acid. Dilute 5.0 ml of this column efficiency determined from the stavudine peak is not
solution to 50.0 ml with 0.01 M hydrochloric acid; in the case less than 3000 theoretical plates, the tailing factor is not more
of capsules containing 40 mg of stavudine, weigh accurately than 2.0 and the relative standard deviation for replicate
about 40 mg of stavudine RS and transfer to a 100-ml volumetric injections is not more than 2.0 per cent.
flask. Dissolve and dilute to volume with 0.01 M hydrochloric Inject separately the test solution and the reference solution
acid. Dilute 5.0 ml of this solution to 50.0 ml with 0.01 M and measure the responses for the major peak.
hydrochloric acid.
Calculate the content of C10H12N2O4 in the capsules.
Chromatographic system
Storage. Store protected from moisture.
– a stainless steel column 15 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 85 volumes of water and
15 volumes of methanol, Stavudine Oral Solution
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 266 nm, Stavudine Oral Solution is a mixture consisting of Stavudine
– a 20 µl loop injector. with buffering agents and other excipients. It contains a
suitable flavouring agent. It is filled in a sealed container.
Inject the reference solution and record the chromatograms
for twice the retention time of stavudine. The test is not valid The oral solution is constituted by dispersing the contents of
unless the column efficiency determined from the stavudine the sealed container in the specified volume of water just
peak is not less than 3000 theoretical plates, the tailing factor before issue.
is not more than 1.5 and the relative standard deviation for Stavudine Oral Solution contains not less than 90.0 per cent
replicate injections is not more than 1.5 per cent and not more than 110.0 per cent of the stated amount of
Inject separately the test solution and the reference solution stavudine C10H12N2O4.
and measure the responses for the major peak. Storage. Store the constituted solution in a refrigerator
Calculate the content of C10H12N2O4 in the medium. (2º to 8º). Discard any unused portion after 30 days of
reconstitution.
D. Not less than 70 per cent of the stated amount of
C10H12N2O4. The contents of the sealed container comply with the
requirements stated under Oral Liquids and with the
Other tests. Comply with the tests stated under Capsules. following requirements.
Assay. Determine by liquid chromatography (2.4.14).
Identification
Test solution. Weigh accurately a quantity of the mixed
contents of 20 capsules containing about 50 mg of Stavudine In the Assay, the principal peak in the chromatogram obtained
and transfer to a 100-ml volumetric flask. Add about 60 ml of with the test solution corresponds to the peak in the
water, mix with the aid of ultrasound for 10 minutes, dilute to chromatogram obtained with reference solution (a).

1121
STAVUDINE AND LAMIVUDINE TABLETS IP 2007

Tests Chromatographic system


– a stainless steel column 3.3 cm x 4.6 mm, packed with
pH (2.4.24). 5.0 to 7.0, determined in the reconstituted solution. octadecylsilane bonded to porous silica (5 µm),
Related substances. Determine by liquid chromatography – mobile phase: A. 95 volumes of 25 mM ammonium
(2.4.14). acetate and 5 volumes of methanol,
B. 50 volumes of 25 mM ammonium
NOTE — Prepare the solutions immediately before use. acetate and 50 volumes of methanol.
Test solution. Weigh accurately a quantity of the reconstituted – flow rate. 1 ml per minute,
solution containing 10 mg of stavudine and dissolve in 20 ml – a linear gradient programme using the conditions given
of water. below,
– spectrophotometer set at 268 nm,
Reference solution (a). A 0.05 per cent w/v solution of – a 20 µl loop injector.
stavudine RS in water.
Time mobile phase A mobile phase B
Reference solution (b). Dilute 1 ml of reference solution (a) to (in min.) (per cent v/v) (per cent v/v)
100 ml with water. 0 100 0
Reference solution (c). A solution containing 0.0125 per cent 60 100 0
w/v each of thymine and thymidine in water. Dilute 2 ml of the 120 0 100
solution to 100 ml of water. 155 100 0
Use the chromatographic system described under Assay. Inject reference solution (b). The test is not valid unless the
Inject reference solution (a). The test is not valid unless the resolution between thymine and thymidine is not less than
tailing factor is not more than 2.0 and the column efficiency in 8.4.
not less than 2000 theoretical plates. Inject reference solution (a). The test is not valid unless the
Inject reference solution (c). The test is not valid unless the tailing factor is not more than 2.0, the column efficiency in not
resolution between thymine and thymidine peak is not less less than 2000 theoretical plates and the relative standard
than 8.4. deviation for replicate injections is not more than 2.0 per cent.

Inject the test solution and reference solution (b). Run the Inject the test solution and reference solution (a).
chromatogram for 4 times the retention time (about 9 minutes) Calculate the content of C10H12N2O4 in the oral solution.
of the principal peak. In the chromatogram obtained with the
Storage. Store protected from moisture, at a temperature not
test solution, the area of any secondary peak is not more than
exceeding 30º.
1.5 times the area of the peak in the chromatogram obtained
with the reference solution (b) (1.5 per cent) and the sum of
areas of all the secondary peaks is not more than 3 times the
area of the peak in the chromatogram obtained with the Stavudine And Lamivudine Tablets
reference solution (b) (3.0 per cent).
Stavudine and Lamivudine Tablets contain not less than
Other tests. Complies with the tests stated under Oral Liquids. 90.0 per cent and not more than 110.0 per cent of the stated
amounts of stavudine, C 10 H 12 N 2 O 4 and lamivudine,
Water (2.3.43). Not more than 2.0 per cent, determined on
C8H11N3O3S. The tablets may be coated.
0.5 g.
Assay. Determine by liquid chromatography (2.4.14). Identification
NOTE — Prepare the solutions immediately before use. In the Assay, the two principal peaks in the chromatogram
obtained with the test solution correspond to the peaks due
Test solution. Weigh accurately a quantity of the reconstituted to stavudine and lamivudine in the chromatogram obtained
solution containing 10 mg of Stavudine, disperse in sufficient with the reference solution.
water and diluted to 100.0 ml with water.
Reference solution (a). A 0.01 per cent w/v solution of
Tests
stavudine RS in water. Related substances. Determine by liquid chromatography
Reference solution (b). A solution containing 0.0125 per cent (2.4.14).
w/v each of thymine and thymidine in water. Dilute 2 ml of the Test solution. Weigh accurately a quantity of the powdered
solution to 100 ml with water. tablets containing about 100 mg of lamivudine and transfer to

1122
IP 2007 STAVUDINE AND LAMIVUDINE TABLETS

a 200-ml volumetric flask. Add about 100 ml of water, mix with Dissolution (2.5.2).
the aid of ultrasound for 10 minutes with occasional shaking Apparatus. No 1
to obtain a uniform dispersion, cool to room temperature, dilute Medium. 900 ml of 0.01 M hydrochloric acid
to volume with water and mix. Filter through a membrane filter Speed and time. 50 rpm and 45 minutes.
disc with an average pore diameter not greater than 1.0 µm,
rejecting the first few ml of the filtrate. Withdraw a suitable volume of the medium and filter.
Reference solution (a). Weigh accurately about 65 mg of Determine by liquid chromatography (2.4.14).
stavudine RS and 6.5 mg of thymine RS, and transfer to a Test solution. The filtrate obtained as given above.
25-ml volumetric flask. Add 5 ml of methanol, sonicate to
dissolve, dilute to volume with water and mix. Reference solution. In the case of tablets containing 30 mg of
stavudine, weigh accurately about 33 mg of stavudine RS and
Reference solution (b). Weigh accurately about 100 mg of 167 mg of lamivudine RS and transfer to a 100-ml volumetric
lamivudine RS, transfer to a 200-ml volumetric flask, add about flask. Add about 20 ml of methanol, sonicate to dissolve and
100 ml of water and mix with the aid of ultrasound to dissolve. dilute to volume with a solvent mixture of equal volumes of
Add 10 ml of reference solution (a) to this solution and dilute methanol and water (diluent). Dilute 5.0 ml of this solution to
to volume with water and mix. 50.0 ml with 0.01 M hydrochloric acid; in the case of tablets
Chromatographic system containing 40 mg of stavudine, weigh accurately about 44 mg
– a stainless steel column 25 cm x 4.6 mm, packed with of stavudine RS and 167 mg of lamivudine RS and transfer to
octylsilane bonded to porous silica (5 µm), a 100-ml volumetric flask. Add about 20 ml of methanol, mix
– mobile phase: gradient mixtures of acetonitrile and with the aid of ultrasound to dissolve and dilute to volume
0.1 M ammonium acetate, with the diluent. Dilute 5.0 ml of this solution to 50.0 ml with
– flow rate. 1 ml per minute, 0.01 M hydrochloric acid.
– a linear gradient programme using the conditions given Chromatographic system
below, – a stainless steel column 15 cm x 4.6 mm, packed with
– spectrophotometer set at 270 nm, octadecylsilane bonded to porous silica (5 µm),
– a 20 µl loop injector. – mobile phase: a mixture of 35 volumes of methanol and
Time 0.1 M Ammonium acetate Acetonitrile 65 volumes of a buffer prepared by dissolving 0.68 g of
(in min.) (per cent v/v) (per cent v/v) potassium dihydrogen phosphate and 1.0 g of sodium
0 95 5 octanesulphonate in 1000.0 ml of water to which 1 ml of
triethylamine is added and the pH of which is adjusted
5 95 5
to 2.5 with phosphoric acid,
25 20 80 – flow rate. 1 ml per minute,
30 20 80 – spectrophotometer set at 266 nm,
31 95 5 – a 10 µl loop injector.
35 95 5 Inject the reference solution. The test is not valid unless the
Inject separately reference solutions (b) and (c). The test is column efficiency determined from the lamivudine peak is not
not valid unless the column efficiency determined from the less than 2000 theoretical plates, the tailing factor for the
thymine, stavudine and lamivudine peaks is not less than individual stavudine and lamivudine peaks is not more than
3000 theoretical plates and the tailing factor for the same peaks 1.5 and the relative standard deviation for replicate injections
is not more than 2.0. for each of the peaks corresponding to stavudine and
lamivudine is not more than 1.0 per cent.
Inject separately water and the test solution. Examine the
chromatogram obtained with water for any extraneous peaks Inject separately the test solution and the reference solution
and ignore the corresponding peaks observed in the and measure the responses for the major peaks due to
chromatogram obtained with the test solution. stavudine and lamivudine.

Any secondary peak observed in the chromatogram obtained Calculate the contents of C10H12N2O4 and C8H11N3O3S in the
with the test solution corresponding to thymine is not more medium.
than 3.0 per cent. Any other secondary peak observed in the D. Not less than 70 per cent of the stated amounts of
chromatogram obtained with the test solution is not more C10H12N2O4 and C8H11N3O3S.
than 0.5 per cent and the sum of the areas of all the secondary
Other tests. Comply with the tests stated under Tablets.
peaks is not more than 4.0 per cent when calculated by
percentage area normalisation. Assay. Determine by liquid chromatography (2.4.14).

1123
STEARIC ACID IP 2007

Test solution. Weigh and powder 20 tablets. Transfer an principal peaks in the chromatogram obtained with reference
accurately weighed quantity of the powder containing about solution (c).
150 mg of Lamivudine to a 100-ml volumetric flask, add about
20 ml of methanol and 50 ml of a mixture of equal volumes of Tests
water and methanol, mix with the aid of ultrasound for Congealing temperature (2.4.10). Not lower than 54º.
5 minutes with occasional shaking to obtain a uniform
dispersion. Cool to room temperature and dilute to volume Acid value (2.3.23). 200 to 212, determined on 1.0 g.
with the same solvent mixture. Filter the solution through a Iodine value (2.3.28). Not more than 4.0 (iodine monochloride
membrane filter disc with an average pore diameter not greater method).
than 1.0 µm, rejecting the first few ml of the filtrate. Dilute Mineral acid. Shake 5 g of the melted substance with an equal
suitably with the same solvent mixture to obtain a solution volume of hot water for 2 minutes, cool and filter. To the
with a concentration of 0.15 mg per ml of lamivudine. filtrate add 0.05 ml of methyl orange solution; no red colour is
Reference solution. A similarly prepared solution containing produced.
0.015 per cent w/v of lamivudine RS and a concentration of Heavy metals (2.3.13). 1.0 g complies with the limit test for
stavudine RS similar to that of the concentration of stavudine heavy metals, Method B (20 ppm).
in test solution.
Sulphated ash (2.3.18). Not more than 0.1 per cent, determined
Use the chromatographic system described under Dissolution. on 2.0 g.
Inject the reference solution. The test is not valid unless the Assay. Determine by gas chromatography (2.4.13).
column efficiency determined from the lamivudine peak is not
less than 2000 theoretical plates, the tailing factor for the Test solution. Add 5 ml of boron trifluoride solution to 0.1 g
individual peaks due to lamivudine and stavudine is not more of the substance under examination and heat under a reflux
than 1.5 and the relative standard deviation for replicate condenser for 15 minutes, cool and transfer to a separating
injections for each of the peaks corresponding to stavudine funnel with the aid of 10 ml of hexane. Add 10 ml of a saturated
and lamivudine is not more than 2.0 per cent. solution of sodium chloride and 10 ml of water, shake, discard
the lower aqueous layer and dry the upper layer over
Inject separately the test solution and the reference solution anhydrous sodium sulphate.
and measure the responses for the major peaks.
Reference solution (a). Add 5 ml of a 1 per cent w/v solution
Calculate the contents of C10H12N2O4 and C8H11N3O3S in the of nonadecanoic acid (internal standard) in toluene to a
tablets. mixture of 50 mg of stearic acid RS and 50 mg of palmitic acid
Storage. Store protected from moisture. RS and evaporate to dryness. Add 5 ml of boron trifluoride
solution and complete the procedure described for the test
solution beginning at the words “heat under a reflux
condenser....”..

Stearic Acid Reference solution (b). Add 5 ml of the internal standard


solution to 0.1 g of the substance under examination, evaporate
to dryness, add 5 ml of boron trifluoride solution and complete
COOH
H3C the procedure described for the test solution beginning at the
words “heat under a reflux condenser......”.
Stearic Acid consists of a mixture of fatty acids, chiefly stearic Reference solution (c). Prepare in the same manner as reference
acid (C18H36O2) and palmitic acid (C16H32O2). It may contain a solution (a) but omitting the internal standard.
suitable antioxidant.
Chromatographic system
Stearic Acid contains not less than 40.0 per cent of C18H36O2 – a glass column 1.5 m x 4 mm, packed with acid-washed,
and the sum of the contents of C18H36O2 and C16H32O2 is not silanised diatomaceous support (100 to 120 mesh) coated
less than 90.0 per cent. with 15 per cent w/w of diethylene glycol succinate
Description. A white powder or white, greasy, flaky crystals polyester,
or hard masses showing signs of crystallisation. – temperature: column 170º,
inlet port and detector at 240°,
Identification – flow rate. 30 ml per minute of the carrier gas.

In the Assay, the chromatogram obtained with the test solution Calculate the content of C18H36O2, and C16H32O2.
shows two principal peaks which correspond to the two Storage. Store protected from moisture.

1124
IP 2007 STILBOESTROL

Stearyl Alcohol B. When examined in the range 230 nm to 450 nm (2.4.7), the
irradiated solution prepared as directed in the Assay shows
Stearyl Alcohol is a mixture of solid alcohols consisting chiefly absorption maxima at about 292 nm and 418 nm.
of 1-octadecanol, (C18H38O).
C. In the test for Mono- and di-methyl ethers, the principal
Description. A white, unctuous mass or almost white flakes or spot in the chromatogram obtained with the test solution
granules; odour, faint and characteristic. corresponds to that in the chromatogram obtained with
reference solution (a).
Tests
D. Dissolve 0.5 mg in 0.2 ml of glacial acetic acid, add 1 ml of
Appearance of solution. Dissolve 0.5 g in 20 ml of ethanol phosphoric acid and heat in a water-bath for 3 minutes; a
(95 per cent) by heating to boiling and allow to cool. The deep yellow colour is produced which almost disappears on
solution is clear (2.4.1), and not more intensely coloured than dilution with 3 ml of glacial acetic acid (distinction from
reference solution BS6 (2.4.1). dienoestrol).
Melting range (2.4.21). 55º to 60º.
Tests
Acid value (2.3.23). Not more than 2.0.
4,4'-Dihydroxystilben and related ethers. Absorbance of a
Hydroxyl value (2.3.27). 195 to 220. 1.0 per cent w/v solution in ethanol at about 325 nm, not more
Iodine value (2.3.28). Not more than 2.0 (iodine bromide than 0.50 (2.4.7).
method), determined on 2.0 g dissolved in 25 ml of chloroform, Mono- and di-methyl ethers. Determine by thin-layer
warming if necessary to effect solution. chromatography (2.4.17), coating the plate with silica gel.
Saponification value (2.3.37). Not more than 2.0, determined Mobile phase. A mixture of 90 volumes of toluene and
on 10.0 g. 10 volumes of diethylamine.
Storage. Store protected from moisture. Test solution. Dissolve 0.5 g of the substance under
examination in 100 ml of ethanol (95 per cent).
Reference solution (a). A 0.5 per cent w/v solution of
stilboestrol RS in ethanol (95 per cent).
Stilboestrol
Reference solution (b). A 0.05 per cent w/v solution of
Diethylstilboestrol stilboestrol monomethyl ether RS in ethanol (95 per cent).
Reference solution (c). A 0.05 per cent w/v solution of
H3C OH stilboestrol dimethyl ether RS in ethanol (95 per cent).
Reference solution (d). A solution containing 0.25 per cent
w/v each of dienoestrol RS and stilboestrol RS
HO CH3 Apply to the plate 1 µl of each solution. After development,
dry the plate in air, spray with ethanolic sulphuric acid
C18H20O2 Mol. Wt. 268.4 (20 per cent) and heat at 120º for 10 minutes. Any secondary
spots in the chromatogram obtained with the test solution
Stilboestrol is (E)-α,β-diethylstilbene-4,4'-diol. corresponding to the mono-and di-methyl ethers of
Stilboestrol contains not less than 97.0 per cent and not more stilboestrol are not more intense than the spots in the
than 101.0 per cent of C18H20O2, calculated on the dried basis. chromatograms obtained with reference solutions (b) and (c)
respectively. Stilboestrol sometimes produces two spots. The
Description. A white or almost white, crystalline powder;
test is not valid unless the chromatogram obtained with
odourless.
reference solution (d) shows two clearly separated spots of
Identification approximately the same intensity.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Test A may be omitted if tests B, C and D are carried out. Tests
B and C may be omitted if tests A and D are carried out. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105º.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with stilboestrol RS Assay. Weigh accurately about 20 mg, dissolve in sufficient
or with the reference spectrum of stilboestrol. ethanol to produce 100.0 ml and dilute 10.0 ml of this solution

1125
STILBOESTROL TABLETS IP 2007

to 100.0 ml with ethanol. To 25.0 ml of the resulting solution Assay. Weigh and powder 20 tablets. Weigh accurately a
add 25.0 ml of a solution prepared by dissolving 1 g of quantity of the powder containing about 5 mg of Stilboestrol,
dipotassium hydrogen phosphate in 55 ml of water, transfer a add 5 ml of ethanol, shake for 15 minutes, add sufficient
portion of the mixture to a 1-cm closed quartz cell, place the ethanol to produce 100.0 ml and centrifuge. Dilute 20.0 ml of
cell 10 cm from a 15-watt short-wave, ultraviolet light of mercury the clear, supernatant liquid to 50.0 ml with ethanol and mix.
lamp and irradiate for 10 minutes. Measure the absorbance of To 25.0 ml of the resulting solution, add 25.0 ml of a solution
the irradiated solution at the maximum at about 418 nm (2.4.7). prepared by dissolving 1 g of dipotassium hydrogen
Calculate the content of C 18H20O2 from the absorbance phosphate in 55 ml of water, transfer a portion of the mixture
obtained by repeating the operation using stilboestrol RS in to a 1-cm closed quartz cell, place the cell 10 cm from a 15-watt
place of the substance under examination. short-wave, ultraviolet light of mercury lamp and irradiate for
10 minutes. Measure the absorbance of the irradiated solution
Storage. Store protected from light and moisture. at 418 nm (2.4.7).
Calculate the content of C18H20O2 in the tablet from the
absorbance obtained by repeating the operation using
Stilboestrol Tablets stilboestrol RS in place of the substance under examination.

Diethylstilboestrol Tablets Storage. Store protected from light and moisture.

Stilboestrol Tablets contain not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
stilboestrol, C18H20O2. The tablets may be coated. Prepared Storax
Identification Storax
A. When examined in the range 230 nm to 450 nm (2.4.7), the Prepared Storax is a balsam obtained from the wounded trunk
irradiated solution prepared as directed in the Assay shows of Liquidambar orientalis Miller (Fam.Hamameli-daceae) and
absorption maxima at about 292 nm and 418 nm. subsequently purified by solution in Ethanol (95 per cent),
filtration and evaporation of the solvent.
B. Extract a quantity of the powdered tablets containing 3 mg
of Stilboestrol with ether, filter and evaporate the filtrate to Prepared Storax contains not less than 30.0 per cent of total
dryness. Dissolve 0.5 mg of the residue in 0.2 ml of glacial balsamic acids calculated as cinnamic acid, C9H8O2, on the
acetic acid, add 1 ml of phosphoric acid and heat in a water- dried basis.
bath for 3 minutes; a deep yellow colour is produced which Description. A brown, viscous substance, transparent in thin
almost disappears on dilution with 3 ml of glacial acetic acid layers; odour, agreeable and balsamic.
(distinction from dienoestrol).
Identification
Tests
Shake 1 g with a 10 per cent w/v solution of potassium
Uniformity of content. For tablets containing 10 mg or less
chromate and 1 ml of sulphuric acid; the odour of
— Comply with the test stated under Tablets.
benzaldehyde is produced.
Finely crush one tablet, add 10 ml of ethanol, shake for
30 minutes, add sufficient ethanol to produce 25.0 ml and Tests
centrifuge. Pipette an aliquot of the supernatant liquid
containing 0.5 mg of Stilboestrol add 25.0 ml of a solution Acid value (2.3.23). 50 to 80, calculated on the dried basis.
prepared by dissolving 1 g of dipotassium hydrogen Ester value (2.3.26). 100 to 133, calculated on the dried basis.
phosphate in 55 ml of water, transfer a portion of the mixture
Saponification value (2.3.37). 170 to 200, calculated on the
to a 1-cm closed quartz cell, place the cell 10 cm from a 15-watt
dried basis.
short-wave, ultraviolet light of mercury lamp and irradiate for
10 minutes. Measure the absorbance of the irradiated solution Ethanol-soluble matter. Not less than 70 per cent, determined
at the maximum at about 418 nm (2.4.7). by the following method. Weigh accurately about 10.0 g in a
beaker and heat at 105º for 30 minutes. Dissolve the residue in
Calculate the content of C18H20O2 in the tablet from the
100 ml of hot ethanol (95 per cent), filter through a tared
absorbance obtained by repeating the operation using
sintered glass crucible, wash the residue with small amounts
stilboestrol RS in place of the substance under examination.
of hot ethanol (95 per cent) until the last washing is nearly
Other tests. Comply with the tests stated under Tablets. colourless. Reserve the residue for the Ethanol-insoluble matter

1126
IP 2007 STREPTOKINASE

test. Combine the filtrate and the washings and evaporate at a Identification
temperature not exceeding 60º. Dry the residue at 105º for
1 hour, cool and weigh. A. Place 0.5 ml of citrated human, canine or rabbit plasma in a
haemolysis tube maintained in a water-bath at 37º. Add 0.1 ml
Ethanol-insoluble matter. Not more than 5 per cent, determined of a solution of the substance under examination containing
by the following method. Dry the residue obtained in the test 10,000 Units per ml in citro-phosphate buffer pH 7.2 and
for Ethanol-soluble matter at 105º for 1 hour, cool and weigh. 0.1 ml of a solution of thrombin containing 20 Units per ml in
Loss on drying (2.4.19). Not more than 5 per cent, determined citro-phosphate buffer pH 7.2 and shake immediately; a clot
on 1.0 g by drying in a thin layer over phosphorus pentoxide forms and lyses within 30 minutes. Repeat the procedure using
at 60º at a pressure not exceeding 2.7 kPa. citrated bovine plasma; lysis does not occur within 1 hour.
Assay. Weigh accurately about 1.25 g and boil with 25 ml of B. Dissolve 0.6 g of agar in 50.0 ml of mixed barbitone buffer
dilute ethanolic potassium hydroxide solution under a reflux pH 8.6, heating until a clear solution is obtained. Place glass
condenser for 1 hour. Remove the ethanol and digest the plates (50 mm x 50 mm) that are free from traces of grease on a
residue with 50 ml of hot water until diffused. Cool the liquid, level surface. Apply to each plate 4 ml of the agar solution and
add 150 ml of water and 1.5 g of magnesium sulphate allow to cool until set. Bore a hole 6 mm in diameter in the
dissolved in 50 ml of water. Mix thoroughly and set aside for centre of the agar and an appropriate number of holes (not
10 minutes. Filter, wash the residue on the filter with 20 ml of exceeding six) at distances of 11 mm from the central hole
water, acidify the combined filtrate and washings with removing the residual agar by means of a cannula connected
hydrochloric acid and extract with successive quantities of to a vacuum pump. Place a quantity of 80 µl of goat or rabbit
50, 40, 30, 30 and 30 ml of ether. Combine the ether extracts and antistreptokinase serum containing 10,000 Units of
discard the aqueous portion. Extract with successive antistreptokinase activity per ml in the central hole and 80 µl
quantities of 20, 20, 10, 10 and 10 ml of sodium bicarbonate of a solution of the substance under examination containing
solution, washing each aqueous extract with the same 20 ml 125,000 Units of streptokinase activity per ml of each of the
of ether. Discard the ether layers, acidify the combined surrounding holes. Place the plates in a humidified tank for
aqueous extracts with hydrochloric acid and extract with 24 hours.
successive quantities of 30, 20, 20 and 10 ml of chloroform, Only one precipitation arc is produced which is well-defined
filtering each chloroform extract through a plug of cotton wool and localised between the application point of the serum and
on which a layer of anhydrous sodium sulphate is placed. each hole containing the solution of the substance under
Evaporate the chloroform on a water-bath until about 10 ml examination.
remains and remove the remainder in a current of air stopping
immediately when the last trace of solvent is removed. Dissolve Tests
the residue by warming with 10 ml of ethanol (95 per cent),
pH (2.4.24). 6.8 to 7.5, determined on a solution prepared in
previously neutralised to phenol red solution, cool and titrate
freshly boiled and cooled water containing 5000 Units per ml.
with 0.1 M sodium hydroxide using phenol red solution as
indicator. Streptodornase. Introduce 0.5 ml of a 0.1 per cent w/v solution
of sodium deoxyribonucleate in imidazole buffer pH 6.5 into
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01482 g of
each of eight centrifuge tubes. To each of the first two tubes
total balsamic acids, calculated as cinnamic acid, C9H8O2.
add 0.25 ml of imidazole buffer pH 6.5 and 0.25 ml of solution
Storage. Store protected from moisture. of the substance under examination in imidazole buffer pH
6.5 containing 150,000 Units per ml (solution A) followed
immediately by 3.0 ml of 0.25 M perchloric acid. Mix the
Streptokinase contents of each tube, centrifuge for 5 minutes at 3000 rpm
and measure the absorbance of each of the supernatant liquids
Streptokinase is a preparation of a protein obtained from culture
at about 260 nm (2.4.7), using as the blank a mixture of 1.0 ml of
filtrates of certain strains of Streptococcus haemolyticus group
imidazole buffer pH 6.5 and 3.0 ml of 0.25 M perchloric acid.
C. It has the property of combining with human plasminogen
Calculate the sum of the two absorbances (A1). To each of the
to form plasminogen activator and is purified to contain not
remaining six tubes add, respectively, 0.25, 0.25, 0.125, 0.125, 0
less than 600 Units of Streptokinase activity per µg of nitrogen
and 0 ml of imidazole buffer pH 6.5 followed by 0.25 ml of
before addition of any stabiliser or carrier. It usually contains
solution A and finally 0, 0, 0.125, 0.125, 0.25 and 0.25 ml
a buffer and may be stabilised by the addition of suitable
respectively of a solution of the Standard Preparation
substances such as Human Albumin.
containing 20 Units of streptodornase activity per ml in
Description. A white powder or a white, friable solid; imidazole buffer pH 6.5. [The Standard Preparation is the 1st
hygroscopic. International Standard Preparation for Streptodornase,

1127
STREPTOKINASE IP 2007

established in 1964, consisting of a freeze-dried mixture of Prepare a solution of the Standard Preparation to contain
streptodornase and streptokinase with lactose (supplied in 1000 Units of streptokinase activity per ml and prepare a
ampoules containing 2400 Units of streptodornase activity), solution of the preparation under examination expected to
or another suitable preparation the activity of which has been have the same concentration; keep the solutions in ice and
determined in relation to the International Standard]. Mix the use within 6 hours. Prepare three 1.5-fold serial dilutions of
contents of each tube, incubate at 37º for 15 minutes and add the solution of the Standard Preparation so that the longest
to each tube 3.0 ml of 0.25 M perchloric acid. Mix the contents clot-lysis time is less than 20 minutes and prepare three similar
of each tube, centrifuge and measure the absorbance of each dilutions of the solution of the preparation under examination.
of the supernatant liquids at about 260 nm (2.4.7), using as the Keep the solutions in ice and use within 1 hour. Using 24
blank the mixture specified above. If the sum of the absorbances tubes, 8 mm in diameter, label the tubes S1, S2, S3 for the dilutions
of the liquids in the third and fourth tubes is A2, that of the of the Standard Preparation and T1, T2, T3 for the dilutions of
liquids in the fifth and six tubes is A3, and that of the liquids in the preparation under examination, allocating four tubes to
the seventh and eighth tubes is (A4), (A1–A2) is less than each dilution. Place the tubes in ice. Into each tube introduce
0.5(A3+A4)–A2. 0.2 ml of the appropriate dilution, 0.2 ml of citro-phosphate
Streptolysin. Dissolve a quantity of the substance under buffer pH 7.2 containing 3 per cent w/v of bovine serum
examination containing 500,000 Units in 0.5 ml of a mixture of albumin and 0.1 ml of a solution containing 20 Units of
90 volumes of saline solution and 10 volumes of citro- thrombin per ml. Place the tubes in a water-bath at 37º and
phosphate buffer pH 7.2 in a haemolysis tube. Add 0.4 ml of a allow to stand for 2 minutes to attain temperature equilibrium.
2.3 per cent w/v solution of sodium thioglycollate and Using an automatic pipette, introduce into the bottom of the
incubate in a water-bath at 37º for 10 minutes. Add 0.1 ml of a first tube 0.5 ml of a 1 per cent w/v solution to human
solution of a reference preparation of human antistreptolysin euglobulins ensuring mixing. At 5-second intervals introduce
O containing 5 Units per ml and incubate at 37º for 5 minutes. successively into the remaining tubes 0.5 ml of a 1 per cent w/
Add 1 ml of rabbit erythrocyte suspension, continue the v solution of human euglobulins. Using a stop-watch, measure
incubation for 30 minutes and centrifuge at about 1000 rpm. for each tube the time in seconds that elapses between the
The absorbance of the supernatant liquid at about 550 nm addition of the euglobulin and the lysis of the clot.
(2.4.7), is not more than 1.5 times the absorbance obtained by Using the logarithms of the lysis times, calculate the result of
repeating the above procedure using 0.5 ml of the mixture of the assay by standard statistical methods.
saline solution and citro-phosphate buffer pH 7.2 in place of
The estimated potency is not less than 90 per cent and not
the solution containing the substance under examination.
more than 111 per cent of the stated potency. The fiducial
Loss on drying (2.4.19). Not more than 4.0 per cent, determined limits of error are not less than 80 per cent and not more than
on 1.0 g by drying over phosphorus pentoxide at a pressure 125 per cent of the stated potency.
not exceeding 2.7 kPa for 24 hours.
Streptokinase intended for use in the manufacture of parenteral
Assay. The potency of streptokinase is determined by preparations complies with the following additional
comparing its ability to activate human plasminogen to form requirements.
plasmin with that of the Standard Preparation. The plasmin Abnormal toxicity (2.2.1). Determine by Method A, using a
generated is determined by measurement of the time taken to solution containing 50,000 Units in 0.5 ml of water for
lyse a fibrin clot under the conditions of a suitable method of injections administered in 15 to 20 seconds.
assay.
Bacterial endotoxins (2.2.3). Dissolve the contents of the
Standard Preparation sealed container in water BET to give a solution containing
10,000 Units of Streptokinase per ml. Carry out the test on the
The Standard Preparation is the 2nd International Standard
resulting solution; the maximum allowable endotoxin
for Streptokinase, established in 1989, consisting of freeze-
concentration of the solution is 23.33 Units of endotoxin per
dried streptokinase (supplied in ampoules containing
ml. Carry out the test using the maximum valid dilution of the
700 Units of streptokinase activity), or another suitable
prepared solution calculated from the declared sensitively of
preparation the activity of which has been determined in
the lysate used in the test.
relation to the International reference preparation.
Sterility (2.2.11). Complies with the test for sterility.
Method
Storage. Store in sealed containers, protected from light. The
Use citro-phosphate buffer pH 7.2 containing 3 per cent w/v containers should be sterile, tamper-evident and sealed so as
of bovine serum albumin for the preparation of solutions and to exclude micro-organisms. Under these conditions the
dilutions. contents may be expected to retain their potency for 2 years.

1128
IP 2007 STREPTOKINASE INJECTION

Labelling. The label states (1) the number of Units of Only one precipitation arc is produced which is well-defined
streptokinase activity in the container; (2) the number of Units and localised between the application point of the serum and
of streptokinase activity per mg, calculated with reference to each hole containing the solution of the substance under
the dried preparation; (3) the name and quantity of any added examination.
substances; (4) the storage conditions; (5) whether or not it is
intended for use in the manufacture of parenteral preparations. Tests
pH (2.4.24). 6.8 to 7.5, determined on a freshly constituted
injection containing 5000 Units per ml.
Streptokinase Injection Streptodornase. Introduce 0.5 ml of a 0.1 per cent w/v solution
of sodium deoxyribonucleate in imidazole buffer pH 6.5 into
Streptokinase Injection is a sterile material consisting of each of eight centrifuge tubes. To each of the first two tubes
Streptokinase with or without auxiliary agents. It is filled in a add 0.25 ml of imidazole buffer pH 6.5 and 0.25 ml of solution
sealed container. of the contents of the container with the substance under
The injection is constituted by dissolving the contents of the examination in imidazole buffer pH 6.5 containing
sealed container in the requisite amount of sterile Water for 150,000 Units per ml (solution A) followed immediately by
Injections, immediately before use. 3.0 ml of 0.25 M perchloric acid. Mix the contents of each
tube, centrifuge for 5 minutes at 3000 rpm and measure the
The constituted solution complies with the requirements for
absorbance of each of the supernatant liquids at about
Clarity of solution and Particulate matter stated under
260 nm (2.4.7), using as the blank a mixture of 1.0 ml of imidazole
Parenteral Preparations (Injections).
buffer pH 6.5 and 3.0 ml of 0.25 M perchloric acid. Calculate
Storage. The constituted solution should be used immediately the sum of the two absorbances (A1). To each of the remaining
after preparation but, in any case, within the period six tubes add, respectively, 0.25, 0.25, 0.125, 0.125, 0 and 0 ml
recommended by the manufacturer. of imidazole buffer pH 6.5 followed by 0.25 ml of solution A
The contents of the sealed container comply with the and finally 0, 0, 0.125, 0.125, 0.25 and 0.25 ml respectively of a
requirements stated under Parenteral Preparations (Powders solution of the Standard Preparation containing 20 Units of
for Injection) and with the following requirements. streptodornase activity per ml in imidazole buffer pH 6.5.
[The Standard Preparation is the 1st International Standard
Identification Preparation for Streptodornase, established in 1964, consisting
of a freeze-dried mixture of streptodornase and streptokinase
A. Place 0.5 ml of citrated human, canine or rabbit plasma in a with lactose (supplied in ampoules containing 2400 Units of
haemolysis tube maintained in a water-bath at 37º. Add 0.1 ml streptodornase activity), or another suitable preparation the
of a solution of the contents of the container containing activity of which has been determined in relation to the
10,000 Units per ml in citro-phosphate buffer pH 7.2 and International Standard]. Mix the contents of each tube,
0.1 ml of a solution of thrombin containing 20 Units per ml in incubate at 37o for 15 minutes and add to each tube 3.0 ml of
citro-phosphate buffer pH 7.2 and shake immediately; a clot 0.25 M perchloric acid. Mix the contents of each tube,
forms and lyses within 30 minutes. Repeat the procedure using centrifuge and measure the absorbance of each of the
citrated bovine plasma; lysis does not occur within 1 hour. supernatant liquids at about 260 nm (2.4.7), using as the blank
B. Dissolve 0.6 g of agar in 50.0 ml of mixed barbitone buffer the mixture specified above. If the sum of the absorbances of
pH 8.6, heating until a clear solution is obtained. Place glass the liquids in the third and fourth tubes is A2, that of the
plates (50 mm x 50 mm) that are free from traces of grease on a liquids in the fifth and six tubes is A3, and that of the liquids in
level surface. Apply to each plate 4 ml of the agar solution and the seventh and eighth tubes is (A4), (A1–A2) is less than
allow to cool until set. Bore a hole 6 mm in diameter in the 0.5(A3+A4)–A2.
centre of the agar and an appropriate number of holes Streptolysin. Dissolve a quantity of the contents of the
(not exceeding six) at distances of 11 mm from the central hole container containing 500,000 Units in 0.5 ml of a mixture of
removing the residual agar by means of a cannula connected 90 volumes of saline solution and 10 volumes of citro-
to a vacuum pump. Place a quantity of 80 µl of goat or rabbit phosphate buffer pH 7.2 in a haemolysis tube. Add 0.4 ml of a
antistreptokinase serum containing 10,000 Units of 2.3 per cent w/v solution of sodium thioglycollate and
antistreptokinase activity per ml in the central hole and 80 µl incubate in a water-bath at 37º for 10 minutes. Add 0.1 ml of a
of a solution of the contents of the container containing solution of a reference preparation of human antistreptolysin
125,000 Units of streptokinase activity per ml of each of the O containing 5 Units per ml and incubate at 37º for 5 minutes.
surrounding holes. Place the plates in a humidified tank for Add 1 ml of rabbit erythrocyte suspension, continue the
24 hours. incubation for 30 minutes and centrifuge at about 1000 rpm.

1129
STREPTOMYCIN SULPHATE IP 2007

The absorbance of the supernatant liquid at about 550 nm 1 per cent w/v solution to human euglobulins ensuring mixing.
(2.4.7), is not more than 1.5 times the absorbance obtained by At 5-second intervals introduce successively into the
repeating the above procedure using 0.5 ml of the mixture of remaining tubes 0.5 ml of a 1 per cent w/v solution of human
saline solution and citro-phosphate buffer pH 7.2 in place of euglobulins. Using a stop-watch, measure for each tube the
the solution containing the substance under examination. time in seconds that elapses between the addition of the
Bacterial endotoxins (2.2.3). Dissolve the contents of the euglobulin and the lysis of the clot.
sealed container in water BET to give a solution containing Using the logarithms of the lysis times, calculate the result of
10,000 Units of Streptokinase per ml. Carry out the test on the the assay by standard statistical methods.
resulting solution; the maximum allowable endotoxin
The estimated potency is not less than 90 per cent and not
concentration of the solution is 23.33 Units of endotoxin per
more than 111 per cent of the stated potency. The fiducial
ml. Carry out the test using the maximum valid dilution of the
limits of error are not less than 80 per cent and not more than
prepared solution calculated from the declared sensitively of
125 per cent of the stated potency.
the lysate used in the test.
Storage. Store in sealed containers, protected from light in a
Assay. Determine on the mixed contents of ten containers.
refrigerator (2º to 8º). The containers should be sterile and
The potency of streptokinase is determined by comparing its sealed so as to exclude micro-organisms. Under these
ability to activate human plasminogen to form plasmin with conditions the contents may be expected to retain their
that of the Standard Preparation. The plasmin generated is potency for 2 years.
determined by measurement of the time taken to lyse a fibrin Labelling. The label states the total number of Units of
clot under the conditions of a suitable method of assay. streptokinase activity contained in it.
Standard Preparation
The Standard Preparation is the 2nd International Standard
for Streptokinase, established in 1989, consisting of freeze- Streptomycin Sulphate
dried streptokinase (supplied in ampoules containing
700 Units of streptokinase activity), or another suitable
preparation the activity of which has been determined in
relation to the International reference preparation. NH
Method HO HN NH2
Use citro-phosphate buffer pH 7.2 containing 3 per cent w/v
of bovine serum albumin for the preparation of solutions and HO OH
dilutions.
O HN NH2
Prepare a solution of the Standard Preparation to contain O
1000 Units of streptokinase activity per ml and prepare a CHO , 3H2 SO4
NH
solution of the contents of the container expected to have the H3C
same concentration; keep the solutions in ice and use within OH
OH O O
6 hours. Prepare three 1.5-fold serial dilutions of the solution
of the Standard Preparation so that the longest clot-lysis time H3CHN
is less than 20 minutes and prepare three similar dilutions of HO
the solution of the preparation under examination. Keep the OH
solutions in ice and use within 1 hour. Using 24 tubes, 8 mm in
2
diameter, label the tubes S1, S2, S3 for the dilutions of the
Standard Preparation and T1, T2, T3 for the dilutions of the
preparation under examination, allocating four tubes to each
dilution. Place the tubes in ice. Into each tube introduce 0.2 ml (C21H39N7O12)2,3H2SO4 Mol. Wt. 1457.4
of the appropriate dilution, 0.2 ml of citro-phosphate buffer Streptomycin Sulphate is the sulphate of O-2-deoxy-2-
pH 7.2 containing 3 per cent w/v of bovine serum albumin methylamino-α-L-glucopyranosyl-(1→2)-O-5-deoxy-3-C-
and 0.1 ml of a solution containing 20 Units of thrombin per formyl-α-L-lyxofuranosyl-(1→4)-N1,N3-diamidino-D-
ml. Place the tubes in a water-bath at 37º and allow to stand for streptamine, a substance produced by the growth of certain
2 minutes to attain temperature equilibrium. Using an automatic strains of Streptomyces griseus or obtained by any other
pipette, introduce into the bottom of the first tube 0.5 ml of a means.

1130
IP 2007 STREPTOMYCIN SULPHATE

Streptomycin Sulphate has a potency equivalent to not less E. Gives the reactions of sulphates (2.3.1).
than 700 µg and not more than 850 µg of streptomycin per mg.
It contains not less than 90.0 per cent of the stated amount of Tests
streptomycin, C21H39N7O12, calculated on the dried basis.
Appearance of solution. A 25.0 per cent w/v solution in carbon
Description. A white or almost white powder; odourless or dioxide-free water is not more intensely coloured than degree
with slight odour; hygroscopic. 3 of the appropriate range of reference solutions (2.4.1). The
solution, after standing protected from light at a temperature
Identification of about 20º for 24 hours, is not more opalescent than
A. Determine by thin-layer chromatography (2.4.17). Prepare opalescence standard OS2 (2.4.1).
the plate by mixing 0.3 g of carbomer with 240 ml of water, pH (2.4.24). 4.5 to 7.0, determined in a 25.0 per cent w/v solution.
allowing to stand with moderate stirring for 1 hour, adjusting
Sulphates. 18.0 to 21.5 per cent, calculated on the dried basis,
the pH to 7.0 by the gradual addition with constant shaking of
when determined by the following method. Dissolve 0.25 g in
2 M sodium hydroxide and adding 30 g of silica gel H. Spread
100 ml of water, adjust the pH to 11 with strong ammonia
a uniform layer of the resulting suspension 0.75 mm thick.
solution and add 10.0 ml of 0.1 M barium chloride and 0.5 mg
Heat the plate at 110º for 1 hour, allow to cool and use
of metalphthalein. Titrate the excess of barium chloride with
immediately.
0.1 M disodium edetate, adding 50 ml of ethanol (95 per
Mobile phase. A 7 per cent w/v solution of potassium cent) when the colour of the solution begins to change and
dihydrogen phosphate. continuing the titration until the violet-blue colour disappears.
Test solution. Dissolve 0.1 g of the substance under 1 ml of 0.1 M barium chloride is equivalent to 0.009606 g of
examination in 100 ml of water. sulphate, SO4.
Reference solution (a). A 0.1 per cent w/v of streptomycin Colorimetric test. Dissolve 0.1 g in sufficient water to produce
sulphate RS in water. 100 ml. To 5 ml, add 5 ml of 0.2 M sodium hydroxide and heat
Reference solution (b). A solution containing 0.1 per cent in a water-bath for exactly 10 minutes. Cool in ice for exactly
w/v of streptomycin sulphate RS, 0.1 per cent w/v of neomycin 5 minutes, add 3 ml of a 1.5 per cent w/v solution of ferric
sulphate RS and 0.1 per cent w/v of kanamycin monosulphate ammonium sulphate in 0.25 M sulphuric acid and sufficient
RS in water. water to produce 25 ml and mix. Exactly 20 minutes after the
addition of the ferric ammonium sulphate solution, measure
Apply to the plate 10 µl of each solution. Allow the mobile
the absorbance of a 2-cm layer of the solution at the maximum
phase to rise 12 cm. Dry the plate in warm air, spray with a
at about 525 nm (2.4.7), using as the blank a solution prepared
mixture of equal volumes of a 0.2 per cent w/v solution of
in the same manner but omitting the substance under
naphthalene-1,3-diol in ethanol (95 per cent) and sulphuric
examination. The absorbance is not less than 90.0 per cent of
acid (45 per cent) and heat at 150º for 5 to 10 minutes. The
that obtained by carrying out the procedure at the same time
principal spot in the chromatogram obtained with the test
and in the same manner using streptomycin sulphate RS in
solution corresponds to that in the chromatogram obtained
place of the substance under examination, each absorbance
with reference solution (a). The test is not valid unless the
being calculated on the dried basis.
chromatogram obtained with reference solution (b) shows
three clearly separated spots. Streptomycin B. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
B. Dissolve 5 to 10 mg in 4 ml of water and add 1 ml of 1 M
sodium hydroxide. Heat in a water-bath for 4 minutes. Add a Mobile phase. A mixture of 50 volumes of toluene, 25 volumes
slight excess of 2 M hydrochloric acid and 0.1 ml of a 10 per of glacial acetic acid and 25 volumes of methanol.
cent w/v solution of ferric chloride; a violet colour is produced. Test solution. Dissolve 0.2 g of the substance under
C. Dissolve 0.1 g in 2 ml of water and add 1 ml of dilute examination in 5 ml of a freshly prepared mixture of 97 volumes
1-naphthol solution and 2 ml of a mixture of equal volumes of of methanol and 3 volumes of sulphuric acid, heat under a
dilute sodium hypochlorite solution and water; a red colour reflux condenser for 1 hour, cool, wash down the condenser
is produced. with methanol and add sufficient methanol to produce 20 ml.
D. Dissolve 10 mg in 5 ml of water and add 1 ml of 1 M Reference solution. Dissolve 36 mg of D-mannose in 5 ml of a
hydrochloric acid. Heat in a water-bath for 2 minutes. Add freshly prepared mixture of 97 volumes of methanol and
2 ml of a 0.5 per cent w/v solution of 1-naphthol in 1 M sodium 3 volumes of sulphuric acid, heat under a reflux condenser
hydroxide and heat in a water-bath for 1 minute; a faint yellow for 1 hour, cool, wash down the condenser with methanol and
colour is produced. add sufficient methanol to produce 50 ml. Dilute 5 ml of the

1131
STREPTOMYCIN INJECTION IP 2007

resulting solution to 50 ml with methanol; this solution Storage. Store protected from light and moisture. If it is
contains the equivalent of 0.03 per cent w/v of streptomycin B intended for use in the manufacture of parenteral preparations
(1 mg of D-mannose is equivalent to 4.13 mg of streptomycin the container should be sterile and sealed so as to exclude
B). micro-organisms.
Apply to the plate 10 µl of each solution. Allow the mobile Labelling. The label states (1) the equivalent weight of
phase to rise 13 to 15 cm. Dry the plate in air, and spray with a streptomycin contained in it; (2) whether or not the contents
freshly prepared mixture of equal volumes of a 0.2 per cent are intended for use in the manufacture of parenteral
w/v solution of naphthalene-1,3-diol in ethanol (95 per cent) preparations; (3) the name and quantity of any added stabiliser.
and a 20 per cent v/v solution of sulphuric acid and heat at
110º for 5 minutes. Any spot in the chromatogram obtained
with the test solution is not more intense than the Streptomycin Injection
corresponding spot in the chromatogram with the reference
solution. Streptomycin Sulphate Injection
Methanol. Determine by gas chromatography (2.4.13). Streptomycin Injection is a sterile material consisting of
Streptomycin Sulphate with or without auxiliary agents. It is
Test solution. A 4 per cent w/v solution of the substance
filled in a sealed container.
under examination in water.
The injection is constituted by dissolving the contents of the
Reference solution. A 0.012 per cent w/v solution of methanol sealed container in the requisite amount of sterile Water for
in water. Injections, immediately before use.
Chromatographic system The constituted solution complies with the requirements for
– a glass column 1.5 to 2.0 m x 2 to 4 mm, packed with Clarity of solution and Particulate matter stated under
ethylvinylbenzene-divinylbenzene copolymer (150 to Parenteral Preparations (Injections).
180 µm) porous polymer beads (such as Porapak Q),
Storage. The constituted solution should be used immediately
– temperature: column 120º to 140º,
after preparation but, in any case, within the period
inlet port and detector at least 50º higher than that of
recommended by the manufacturer.
the column,
– flow rate. 30 to 40 ml per minute of the carrier gas. Streptomycin Injection contains not less than 90.0 per cent
and not more than 115.0 per cent of the stated amount of
The area of any peak corresponding to methanol in the streptomycin, C21H39N7O12, calculated on the dried basis.
chromatogram obtained with the test solution is not greater
The contents of the sealed container comply with the
than that of the peak in the chromatogram obtained with
requirements stated under Parenteral Preparations
reference solution (0.3 per cent).
(Powders for Injection) and with the following requirements.
Sulphated ash (2.3.18). Not more than 1.0 per cent.
Description. A white or almost white powder which yields a
Loss on drying (2.4.19). Not more than 7.0 per cent, determined clear, colourless or faintly yellow coloured solution when
on 1.0 g by drying over phosphorus pentoxide at 60º at a dissolved in water.
pressure not exceeding 0.1 kPa for 24 hours.
Identification
Assay. Determine by the microbiological assay of antibiotics,
Method A or B (2.2.10), and express the results in µg of A. Determine by thin-layer chromatography (2.4.17). Prepare
streptomycin per mg. the plate by mixing 0.3 g of carbomer with 240 ml of water,
allowing to stand with moderate stirring for 1 hour, adjusting
Streptomycin Sulphate intended for use in the manufacture the pH to 7.0 by the gradual addition with constant shaking of
of parenteral preparations without a further appropriate 2 M sodium hydroxide and adding 30 g of silica gel H. Spread
procedure for removal of bacterial endotoxins complies with a uniform layer of the resulting suspension 0.75 mm thick.
the following additional requirement. Heat the plate at 110º for 1 hour, allow to cool and use
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin immediately.
Unit per mg of streptomycin. Mobile phase. A 7 per cent w/v solution of potassium
Streptomycin Sulphate intended for use in the manufacture dihydrogen phosphate.
of parenteral preparations without a further appropriate Test solution. Dissolve 0.1 g of the substance under
sterilisation procedure complies with the following examination in 100 ml of water.
additional requirement. Reference solution (a). A 0.1 per cent w/v of streptomycin
Sterility (2.2.11). Complies with the test for sterility. sulphate RS in water.

1132
IP 2007 SUCCINYLCHOLINE CHLORIDE

Reference solution (b). A solution containing 0.1 per cent Identification


w/v of streptomycin sulphate RS, 0.1 per cent w/v of neomycin
sulphate RS and 0.1 per cent w/v of kanamycin monosulphate A. Triturate a quantity of the powdered tablets containing
RS in water. 0.2 g of streptomycin in a mixture of 2 ml of methanol and
0.1 ml of sulphuric acid, filter, if necessary, and allow to stand
Apply to the plate 10 µl of each solution. Allow the mobile at about 25º; crystals of streptidine sulphate separate in the
phase to rise 12 cm. Dry the plate in warm air, spray with a course of 2 to 3 days. Dissolve the crystals in a solution of
mixture of equal volumes of a 0.2 per cent w/v solution of 0.1 g of picric acid in 10 ml of hot water and cool; the
naphthalene-1,3-diol in ethanol (95 per cent) and sulphuric precipitate, after recrystallisation from hot water, melts at about
acid (45 per cent) and heat at 150º for 5 to 10 minutes. The 283º (2.4.21).
principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained B. Boil a small quantity of the powdered tablets containing
with reference solution (a). The test is not valid unless the 0.1 g of streptomycin with 5 ml of 1 M sodium hydroxide for a
chromatogram obtained with reference solution (b) shows few minutes, add a slight excess of 2 M hydrochloric acid
three clearly separated spots. and 0.15 ml of a 10 per cent w/v solution of ferric chloride; a
brilliant violet colour is produced.
B. Dissolve 5 to 10 mg in 4 ml of water and add 1 ml of 1 M
sodium hydroxide. Heat in a water-bath for 4 minutes. Add a Tests
slight excess of 2 M hydrochloric acid and 0.1 ml of a 10 per
Other tests. Comply with the tests stated under Tablets.
cent w/v solution of ferric chloride; a violet colour is produced.
Assay. Weigh and powder 20 tablets. Weigh accurately a
C. Dissolve 0.1 g in 2 ml of water and add 1 ml of dilute
quantity of the powder containing about 0.3 g of streptomycin
1-naphthol solution and 2 ml of a mixture of equal volumes of
and, triturate with 20 ml of buffer solution pH 8.0. Dilute to
dilute sodium hypochlorite solution and water; a red colour
100.0 ml with water.
is produced.
Determine by the microbiological assay of antibiotics, Method
D. Dissolve 10 mg in 5 ml of water and add 1 ml of 1 M
A or B (2.2.10).
hydrochloric acid. Heat in a water-bath for 2 minutes. Add
2 ml of a 0.5 per cent w/v solution of 1-naphthol in 1 M sodium Storage. Store protected from moisture.
hydroxide and heat in a water-bath for 1 minute; a faint yellow Labelling. The label states the strength in terms of the
colour is produced. equivalent amount of streptomycin.
Tests
pH (2.4.24). 4.5 to 7.0, determined in a 25.0 per cent w/v solution.
Succinylcholine Chloride
Loss on drying (2.4.19). Not more than 7.0 per cent, determined
on 1.0 g by drying in an oven at 60º over phosphorus pentoxide Suxamethonium Chloride
at a pressure not exceeding 0.1 kPa for 24 hours.
H3C O
Other tests. Complies with the tests stated under Parenteral CH3
Preparations (Injections). H3 C N O
O
N CH3 2Cl , 2H2 O
H3C CH3
Assay. Determine on the mixed contents of ten containers by O
the microbiological assay of antibiotics, Method A or B (2.2.10). C14H30Cl2N2O4,2H2O Mol. Wt. 397.3
Bacterial endotoxins (2.2.3): Not more than 0.25 Endotoxin Succinylcholine Chloride is 2,2'-
Unit per mg of streptomycin. succinyldioxybis(ethyltrimethylammonium) dichloride
Storage. Store protected from moisture. dihydrate.
Labelling. The label states the strength in terms of the Succinylcholine Chloride contains not less than 98.0 per cent
equivalent amount of streptomycin in a suitable dose-volume. and not more than 101.0 per cent of C14H30Cl2N2O4, calculated
on the anhydrous basis.

Streptomycin Tablets Description. A white or almost white, crystalline powder;


almost odourless; hygroscopic.
Streptomycin Sulphate Tablets
Identification
Streptomycin Tablets contain not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of Test A may be omitted if tests B, C and D are carried out. Tests
streptomycin, C21H39N7O12. B and C may be omitted if tests A and D are carried out.

1133
SUCCINYLCHOLINE INJECTION IP 2007

A. Determine by infrared absorption spectrophotometry (2.4.6). 1 ml of 0.1 M perchloric acid is equivalent to 0.01807 g of
Compare the spectrum with that obtained with succinylcholine C14H30Cl2N2O4.
chloride RS or with the reference spectrum of succinylcholine
Storage. Store protected from light and moisture.
chloride.
B. Dissolve about 25 mg in 1 ml of water, add 0.1 ml of a 1 per
cent w/v solution of cobalt chloride and 0.1 ml of potassium
ferrocyanide solution; a green colour is produced.
Succinylcholine Injection
C. Dissolve 1 g in sufficient carbon dioxide-free water to
produce 20 ml. To 1 ml of this solution add 9 ml of water, 10 ml Suxamethonium Chloride Injection
of 1 M sulphuric acid and 30 ml of ammonium reineckate Succinylcholine Injection is a sterile solution of
solution; a pink precipitate is produced. Allow to stand for 30 Succinylcholine Chloride in Water for Injections.
minutes, filter and wash with water, then with ethanol (95 per
Succinylcholine Injection contains not less than 90.0 per cent
cent) and finally with ether. The residue, after drying at 80º
and not more than 107.5 per cent of the stated amount of
melts at 180º to 185º (2.4.21).
succinylcholine chloride, C14H30Cl2N2O4,2H2O.
D. Gives the reactions of chlorides (2.3.1).
Identification
Tests
Dilute a volume containing 20 mg of Succinylcholine Chloride
Appearance of solution. A 5.0 per cent w/v solution in carbon to 50 ml with water. To 0.5 ml add 2 ml of chloroform, 2 ml of a
dioxide-free water (solution A) is clear (2.4.1). 4 ml of solution solution containing 0.16 per cent w/v of citric acid and 6.6 per
A diluted to 10 ml with water is colourless (2.4.1). cent w/v of disodium hydrogen phosphate and 0.1 ml of a
pH (2,4,24). 4.0 to 5.0, determined in a 0.5 per cent w/v solution. solution containing 0.15 per cent w/v of each of bromothymol
Choline chloride. Determine by thin-layer chromatography blue and anhydrous sodium carbonate. Shake for 2 minutes
(2.4.17), coating the plate with microcrystalline cellulose. and allow to separate. The chloroform layer is yellow.

Mobile phase. A mixture of 50 volumes of 1-butanol, Tests


40 volumes of water and 10 volumes of anhydrous formic
acid, shake for 10 minutes, allow to separate. Use the upper pH (2.4.28). 3.0 to 5.0.
layer as the mobile phase. Hydrolysis products. The volume of 0.1 M sodium hydroxide
Test solution. Dissolve 0.4 g of the substance under required for the preliminary neutralisation in the Assay is not
examination in 10 ml of methanol. more than one tenth of the total volume of 0.1 M sodium
hydroxide required for the preliminary neutralisation and the
Reference solution. A solution containing 4 per cent w/v of
hydrolysis.
succinylcholine chloride RS and 0.02 per cent w/v of choline
chloride in methanol. Other tests. Complies with the tests stated under Parenteral
Apply to the plate 5 µl of each solution. After development, Preparations (Injections).
dry the plate in air, spray with potassium iodobismuthate Assay. To an accurately measured volume containing about
solution. Any secondary spot in the chromatogram obtained 0.25 g of Succinylcholine Chloride add 30 ml of carbon dioxide-
with the test solution is not more intense than the spot due to free water and shake with five quantities, each of 25 ml, of
choline chloride in the chromatogram obtained with the ether. Wash the combined ether solutions with two quantities,
reference solution. The test is not valid unless the each of 10 ml, of water and discard the ether. Shake the
chromatogram obtained with the reference solution shows combined washings with two quantities, each of 10 ml, of
two clearly separated spots. ether, add the washings to the original aqueous solution and
Sulphated ash (2.3.18). Not more than 0.1 per cent. neutralise with 0.1 M sodium hydroxide using bromothymol
blue solution as indicator. Add 25.0 ml of 0.1 M sodium
Water (2.3.43). 8.0 to 10.0 per cent, determined on 0.3 g. hydroxide, heat under a reflux condenser for 40 minutes, allow
Assay. Weigh accurately about 0.3 g, dissolve in 30 ml of to cool and titrate the excess of alkali with 0.1 M hydrochloric
anhydrous glacial acetic acid, add 30 ml of acetic anhydride acid using bromothymol blue solution as indicator. Repeat
and 10 ml of mercuric acetate solution. Titrate with 0.1 M the operation using 40 ml of carbon dioxide-free water
perchloric acid, using crystal violet solution as indicator, beginning at the words “Add 25.0 ml of 0.1 M sodium
until a bluish green colour is produced. Carry out a blank hydroxide.....”. The difference between the titrations
titration. represents the amount of sodium hydroxide required.

1134
IP 2007 SULPHACETAMIDE SODIUM

1 ml of 0.1 M sodium hydroxide is equivalent to 0.01987 g of Calcium. To 1 ml of solution A add 9 ml of water and 1 ml of
C14H30Cl2N2O4,2H2O. ammonium oxalate solution; the solution remains clear for at
Storage. Store protected from light. The injection should not least 1 minute.
be allowed to freeze. Heavy metals (2.3.13). Add 0.1 ml of dilute hydrochloric acid
to 4 ml of solution A and dilute with sufficient water to produce
25 ml. The solution complies with the limit test for heavy metals,
Method A (10 ppm).
Sucrose
Sulphites. To 4 ml of solution A add sufficient water to produce
Refined Sugar 20 ml, add 0.05 ml of 0.1 M iodine and 0.05 ml of starch solution;
a blue colour develops.
Dextrins. To 2 ml of solution A add 8 ml of water, 0.05 ml of
HO
2 M hydrochloric acid and 0.05 ml of 0.05 M iodine; the
O solution remains yellow or becomes faint bluish green.

OH Glucose and invert sugar. Dissolve 20 g in sufficient water to


make 100 ml and filter if necessary. Place 50 ml of the clear
OH O
HO solution in a 250-ml beaker, add 50 ml of alkaline cupric
O OH tartarate solution, cover the beaker with a watch glass, heat
HO the mixture at such a rate that it comes to a boil in approximately
4 minutes and continue boiling for exactly 2 minutes. Add
OH OH immediately 100 ml of recently boiled and cooled water and
collect the precipitated cuprous oxide on a tared sintered -
glass crucible. Wash the residue with the hot water, then with
C12H22O11 Mol. Wt. 342.3 10 ml of ethanol (95 per cent) and finally with 10 ml of ether.
Sucrose is β-D-fructofuranosyl-α-D-glucopyranoside. Dry at 105º for 1 hour; the weight of the cuprous oxide is not
more than 112 mg.
Description. An almost white or colourless crystals, dry
crystalline powder; odourless; taste, sweet. Colouring matter. A. To 100 ml of solution A in a ground-
glass-stoppered tube add 1 ml of dilute hypophosphorous
Identification acid and allow to stand for 1 hour; no unpleasant odour is
detectable.
Dissolve 150.0 g in sufficient carbon dioxide-free water
prepared from distilled water to produce 300 ml (solution A). B. Examine solution A in ultraviolet light at 365 nm. Any
Dilute 1 ml of solution A to 100 ml with water. To 5 ml of the fluorescence is not more intense than that of a solution
solution add 2 ml of freshly prepared 2 M sodium hydroxide containing 0.4 mg of quinine sulphate in 0.005 M sulphuric
and 0.15 ml of a freshly prepared copper sulphate solution; acid.
the solution is clear and blue and remains so on boiling. To Sulphated ash (2.3.18). Not more than 0.1 per cent determined
the hot solution add 4 ml of 2 M hydrochloric acid, heat to by dissolving 5.0 g in 5 ml of water, adding 2 ml of sulphuric
boiling and add 4 ml of 2 M sodium hydroxide; an orange acid, evaporating to dryness and igniting to constant weight.
precipitate is produced immediately.
Storage. Store protected from light and moisture.
Tests
Acidity or alkalinity. To 10 ml of solution A add 0.3 ml of Sulphacetamide Sodium
phenolphthalein solution. The solution is colourless and not
more than 0.6 ml of 0.01 M sodium hydroxide is required to
O O O
change the colour of the solution to pink. S
N CH3 ,H2O
Specific optical rotation (2.4.22). +65.9º to +67.0º, determined
in a 10 per cent w/v solution. Na
H2N
Barium. To 10 ml of solution A add 1 ml of 1 M sulphuric acid.
C8H9N2NaO3S,H2O Mol. Wt. 254.2
When examined immediately and after 1 hour any opalescence
is not more intense than that of a mixture of 1 ml of distilled Sulphacetamide Sodium is the monohydrate of the sodium
water and 10 ml of solution A. salt of N1-acetylsulphanilamide.

1135
SULPHACETAMIDE EYE DROPS IP 2007

Sulphacetamide Sodium contains not less than 99.0 per cent Reference solution (b). A 0.025 per cent w/v solution of
and not more than 101.0 per cent of C8H9N2NaO3S, calculated sulphanilamide in water.
on the anhydrous basis.
Reference solution (c). A 0.05 per cent w/v solution of
Description. A white or yellowish white, crystalline powder; sulphanilamide in the test solution.
odourless.
Apply to the plate 5 µl of each solution. After development,
Identification dry the plate in air and spray with a freshly prepared 2 per cent
w/v solution of dimethylaminobenzaldehyde in a mixture of
Test A may be omitted if tests B, C, D, E and F are carried out. 55 volumes of hydrochloric acid and 45 volumes of water.
Tests B, C, D and E may be omitted if tests A and F are carried Any secondary spot in the chromatogram obtained with the
out. test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a) and not
A. Determine by infrared absorption spectrophotometry (2.4.6).
more than one such spot is more intense than the spot in the
Compare the spectrum with that obtained with sulphacetamide
chromatogram obtained with reference solution (b). The
sodium RS or with the reference spectrum of sulphacetamide
chromatogram obtained with reference solution (c) shows two
sodium.
clearly separated spots.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Heavy metals (2.3.13). 1.0 g complies with the limit test for
0.001 per cent w/v solution in citro-phosphate buffer pH 7.0
heavy metals, Method B (20 ppm).
shows an absorption maximum at about 255 nm; absorbance
at about 255 nm, 0.66 to 0.72. Sulphates (2.3.17). Dissolve 1.5 g in sufficient distilled water
to produce 25 ml, add 25 ml of 2 M acetic acid, shake for
C. Dissolve 1 g in 10 ml of water, add 6 ml of 2 M acetic acid
30 minutes and filter. 25 ml of the filtrate complies with the limit
and filter. Wash the precipitate with a small volume of water
test for sulphates (200 ppm).
and dry at 105º for 4 hours. The melting range of the precipitate
is 181º to 185º (2.4.21). Water (2.3.43). 6.0 to 8.0 per cent, determined on 0.2 g.
D. Dissolve 0.1 g of the precipitate obtained in test C in 5 ml of Assay. Weigh accurately about 0.25 g, dissolve in a mixture of
ethanol (95 per cent), add 0.2 ml of sulphuric acid and heat; 50 ml of water and 20 ml of 2 M hydrochloric acid, add 3 g of
ethyl acetate, recognizable by its odour, is produced. potassium bromide, cool in ice and carry out the nitrite titration
(2.3.31).
E. Dissolve 1 mg of the precipitate obtained in test C in 5 ml of
water with the aid of heat. The solution gives the reaction of 1 ml of 0.1 M sodium nitrite is equivalent to 0.02362 g of
primary aromatic amines (2.3.1), producing an orange-red C8H9N2NaO3S.
precipitate.
Storage. Store protected from light and moisture.
F. A 5 per cent w/v solution gives the reactions of sodium
salts (2.3.1).

Tests
Sulphacetamide Eye Drops
Appearance of solution. A 5.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely Sulphacetamide Sodium Eye Drops
coloured than reference solution GYS4 (2.4.1). Sulphacetamide Eye Drops are a sterile solution of
pH (2.4.28). 8.0 to 9.5, determined in a 5.0 per cent w/v solution. Sulphacetamide Sodium in Purified Water. It may contain a
suitable antimicrobial agent.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel HF254. Sulphacetamide Eye Drops contain not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
Mobile phase. A mixture of 50 volumes of 1-butanol, sulphacetamide sodium, C8H9N2NaO3S,H2O.
25 volumes of ethanol, 25 volumes of water and 10 volumes
of strong ammonia solution. Identification
Test solution. A 10 per cent w/v solution of the substance
To a volume containing 0.5 g of Sulphacetamide Sodium add 6
under examination in water.
ml of 5 M acetic acid, stirring constantly. Filter the precipitate,
Reference solution (a). A 0.05 per cent w/v solution of wash with water and dry at 105º for 4 hours. The residue
sulphanilamide in water. complies with the following tests.

1136
IP 2007 SULPHADIAZINE

A. Determine by infrared absorption spectrophotometry (2.4.6). Sulphadiazine


Compare the spectrum with that obtained with sulphacetamide
RS or with the reference spectrum of sulphacetamide.
B. When examined in the range 230 nm to 360 nm (2.4.7), a O O N
0.001 per cent w/v solution in citro-phosphate buffer pH 7.0 S
N N
shows an absorption maximum at about 255 nm; absorbance H
at about 255 nm, 0.66 to 0.72. H2N
C. Dissolve 10 mg in 2 ml of 2 M hydrochloric acid. The C10H10N4O2S Mol. Wt. 250.3
solution gives the reaction of primary aromatic amines (2.3.1).
Sulphadiazine is N1-(pyrimidin-2-yl)sulphanilamide.
Tests Sulphadiazine contains not less than 99.0 per cent and not
Appearance of solution. Dilute the eye drops, if necessary, to more than 101.0 per cent of C10H10N4O2S, calculated on the
contain 10.0 per cent w/v of Sulphacetamide Sodium. The dried basis.
solution is not more intensely coloured than reference solution Description. White, yellowish white or pinkish white crystals
BYS4 (2.4.1). or crystalline powder; almost odourless.
pH (2.4.24). 6.6 to 8.6.
Identification
Related substances. Determine by thin-layer chromatography
(2.4.14), coating the plate with silica gel HF254. Test A may be omitted if tests B, C and D are carried out. Tests
C and D may be omitted if tests A and B are carried out.
Mobile phase. A mixture of 50 volumes of 1-butanol,
25 volumes of ethanol, 25 volumes of water and 10 volumes A. Determine by infrared absorption spectrophotometry (2.4.6).
of strong ammonia solution. Compare the spectrum with that obtained with sulphadiazine
RS or with the reference spectrum of sulphadiazine.
Test solution. Dilute a suitable volume with water to produce
a solution containing 4 per cent w/v of Sulphacetamide B. In the test for Related substances, the principal spot in the
Sodium. chromatogram obtained with test solution (b) corresponds to
Reference solution. A 0.2 per cent w/v solution of that in the chromatogram obtained with the reference solution.
sulphanilamide in water. C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
Apply to the plate 5 µl of each solution. After development, dilute 1 ml of this solution to 10 ml with water. The solution,
dry the plate in air and spray with a freshly prepared 2 per cent without further acidification, gives the reaction of primary
w/v solution of dimethylaminobenzaldehyde in a mixture of aromatic amines (2.3.1).
55 volumes of hydrochloric acid and 45 volumes of water. D. Heat 3 g in a test-tube inclined at an angle of 45º with the
Any secondary spot in the chromatogram obtained with the lower part immersed in a silicone oil-bath at about 270º. It
test solution is not more intense than the spot in the decomposes and a white or yellowish white sublimate is
chromatogram obtained with the reference solution. produced. The sublimate, after recrystallisation from toluene
Other tests. Comply with the tests stated under Eye Drops. and drying at 100º melts at 123º to 127º (2.4.21).
Assay. To an accurately measured volume containing about Tests
0.5 g of Sulphacetamide Sodium add 75 ml of water and 10 ml
of hydrochloric acid. Add 3 g of potassium bromide, cool in Appearance of solution. Dissolve 0.8 g in 10 ml of 1 M sodium
ice and carry out the nitrite titration (2.3.31). hydroxide. The solution is not more intensely coloured than
reference solution YS5, BYS5 or GYS5 (2.4.1).
1 ml of 0.1 M sodium nitrite is equivalent to 0.02542 g of
C8H9N2NaO3S,H2O. Acidity. Heat 1.25 g of the finely powdered substance at about
70º with 25 ml of carbon dioxide-free water for 5 minutes.
Storage. Store protected from light and moisture. The Eye
Cool for about 15 minutes in ice and filter. To 20 ml of the
Drops should not be allowed to freeze.
filtrate add 0.1 ml of bromothymol blue solution. Not more
Labelling. The label states (1) the name and concentration of than 0.2 ml of 0.1 M sodium hydroxide is required to change
any antimicrobial agent used; (2) that it is not meant for the colour of the solution.
injection; (3) that the solution should be used within one
month of opening the container; (4) that the solution should Related substances (2.3.7). Complies with test C.
not be used if it is dark brown in colour; (5) that it should not Heavy metals (2.3.13). 1.0 g complies with the limit test for
be allowed to freeze. heavy metals, Method B (20 ppm).

1137
SULPHADIAZINE TABLETS IP 2007

Sulphated ash (2.3.18). Not more than 0.1 per cent. Dissolution (2.5.2).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Apparatus. No 1
on 1.0 g by drying in an oven at 105º. Medium. 900 ml of 0.1 M hydrochloric acid
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of Speed and time. 100 rpm and 60 minutes.
20 ml of 2 M hydrochloric acid and 50 ml of water. Add 3 g of Withdraw a suitable volume of the sample and filter promptly
potassium bromide, cool in ice and carry out the nitrite titration through a membrane filter disc with an average pore diameter
(2.3.31). not greater than 1.0 mm. Reject the first few ml of the filtrate
1 ml of 0.1 M sodium nitrite is equivalent to 0.02503 g of and dilute a suitable volume of the filtrate with the same
C10H10N4O2S. solvent. Dilute suitably with 0.01 M sodium hydroxide.
Measure the absorbances of the resulting solution and of a
Storage. Store protected from light and moisture.
standard solution of sulphadiazine RS of similar concentration
in the same medium at the maximum at about 254 nm (2.4.7).
Calculate the content of C10H10N4O2S in the medium.
Sulphadiazine Tablets
D. Not less than 70.0 per cent of the stated amount of
Sulphadiazine Tablets contain not less than 95.0 per cent and C10H10N4O2S.
not more than 105.0 per cent of the stated amount of
sulphadiazine, C10H10N4O2S. Other tests. Comply with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Identification quantity of the powder containing about 0.5 g of Sulphadiazine
A. Triturate a quantity of the powdered tablets containing and dissolve as completely as possible in a mixture of 50 ml of
0.5 g Sulphadiazine with two successive quantities, each of water and 10 ml of hydrochloric acid. Carry out the nitrite
5 ml, of chloroform and reject the chloroform. Triturate the titration (2.3.31).
residue with 10 ml of dilute ammonia solution for 5 minutes, 1 ml of 0.1 M sodium nitrite is equivalent to 0.02503 g of
add 10 ml of water and filter. Warm the filtrate until most of the C10H10N4O2S.
ammonia has been expelled, cool and acidify with acetic acid.
Storage. Store protected from light and moisture.
Collect the precipitate, wash with water and dry at about 100º;
the residue melts at about 256º, with decomposition (2.4.21).
B. On the residue obtained in test A determine by infrared Sulphadimethoxine
absorption spectrophotometry (2.4.6). Compare the spectrum
with that obtained with sulphadiazine RS or with the reference OCH3
spectrum of sulphadiazine.
C. In the test for Related substances, the principal spot in the O O N
chromatogram obtained with test solution (b) corresponds to S
N N OCH3
that in the chromatogram obtained with the reference solution.
H
Tests H2N

Related substances (2.3.7). Complies with test C, but using C12H14N4O4S Mol. Wt. 310.3
the following solutions. 1
Sulphadimethoxine is N -(2,6-dimethoxypyrimidin-4-yl)
Test solution (a). Extract a quantity of the powdered tablets sulphanilamide.
containing 0.5 g of Sulphadiazine with 25 ml of a mixture of Sulphadimethoxine contains not less than 99.0 per cent and
90 volumes of methanol and 10 volumes of strong ammonia not more than 101.0 per cent of C12H14N4O4S, calculated on the
solution by shaking for 10 minutes, filter and use the filtrate. dried basis.
Test solution (b). Dilute 1 volume of test solution (a) to
Description. A white or creamy-white, crystalline powder.
5 volumes with a mixture of 24 volumes of methanol and
1 volume of strong ammonia solution. Identification
Test solution (c). Dilute 1 volume of test solution (a) to A. Determine by infrared absorption spectrophotometry (2.4.6).
200 volumes with the same solvent mixture. Compare the spectrum with that obtained with
Reference solution. A 0.4 per cent w/v solution of sulphadimethoxine RS or with the reference spectrum of
sulphadiazine RS in the same solvent mixture. sulphadimethoxine

1138
IP 2007 SULPHADIMIDINE

B. Dissolve about 0.1 g in 3 ml of 5 M sodium hydroxide and B. Dissolve about 0.1 g in 3 ml of 5 M sodium hydroxide and
50 ml of water and dilute to 100 ml with water. To 5 ml of the 50 ml of water and dilute to 100 ml with water. To 5 ml of the
solution add 100 mg of phenol, heat to boiling, cool and add solution add 100 mg of phenol, heat to boiling, cool and add
0.5 ml of sodium hypochlorite solution and 0.15 ml of 5 M 0.5 ml of sodium hypochlorite solution and 0.15 ml of 5 M
sodium hydroxide; a yellow colour is produced. sodium hydroxide; a yellow colour is produced.
C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
dilute 1 ml to 10 ml with water. The resulting solution, without dilute 1 ml to 10 ml with water. The resulting solution, without
further acidification, gives the reaction of primary aromatic further acidification, gives the reaction of primary aromatic
amines (2.3.1). amines (2.3.1).
D. Suspend about 20 mg in 5 ml of water and add 5 M sodium D. Suspend about 20 mg in 5 ml of water and add 5 M sodium
hydroxide until completely dissolved. Add 0.2 ml of cupric hydroxide until completely dissolved. Add 0.2 ml of cupric
sulphate solution; the solution turns yellow and a yellow sulphate solution; the solution turns yellow and a yellow
precipitate is formed. precipitate is formed.
E. Melting point (2.4.21). 197º to 204º. E. Melting range (2.4.21). 197º to 204º.

Tests Tests
Related substances (2.3.7). Complies with test B. Related substances (2.3.7). Complies with test B, but using
the following solutions.
Heavy metals (2.3.1.3). Dissolve 1.0 g in 5 ml of 5 M sodium
hydroxide and 20 ml of water. The solution complies with the Test solution. Extract a quantity of the powdered tablets
limit test for heavy metals, Method A (20 ppm). containing 0.25 g of Sulphadimethoxine with 100 ml of a mixture
Sulphated ash (2.3.18). Not more than 0.1 per cent. of 9 volumes of methanol and 1 volume of strong ammonia
solution by shaking for 10 minutes, filter and use the filtrate.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Reference solution. A 0.00125 per cent w/v solution of
on 1.0 g by drying in an oven at 105º.
sulphanilamide.
Assay. Weigh accurately about 0.3 g, dissolve in a mixture of
Other tests. Comply with the tests stated under Tablets.
20 ml of 2 M hydrochloric acid and 50 ml of water. Add 3 g of
potassium bromide, cool in ice and carry out the nitrite titration Assay. Weigh and powder 20 tablets. Weigh accurately a
(2.3.31). quantity of the powder containing about 0.3 g of
1 ml of 0.1 M sodium nitrite is equivalent to 0.03103 g of Sulphadimethoxine, dissolve as completely as possible in a
C12H14N4O4S. mixture of 50 ml of water and 10 ml of hydrochloric acid and
carry out the nitrite titration (2.3.31).
Storage. Store protected from light and moisture.
1 ml of 0.1 M sodium nitrite is equivalent to 0.03103 g of
C12H14N4O4S.
Storage. Store protected from light and moisture.
Sulphadimethoxine Tablets
Sulphadimethoxine Tablets contain not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
sulphadimethoxine, C12H14N4O4S. Sulphadimidine
Identification CH3
Triturate a quantity of the powdered tablets containing 0.5 g
of Sulphadimethoxine with 5 ml of 0.5 M hydrochloric acid, O O N
filter and neutralise the filtrate to litmus paper with 0.5 M S
N N CH3
sodium hydroxide. The precipitate, after washing with water H
and drying at 105º complies with the following tests. H2N
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with C12H14N4O2S Mol. Wt. 278.3
sulphadimethoxine RS or with the reference spectrum of 1
Sulphadimidine is N -(4,6-dimethylpyrimidin-2-yl)
sulphadimethoxine sulphanilamide.

1139
SULPHADIMIDINE SODIUM IP 2007

Sulphadimidine contains not less than 99.0 per cent and not Sulphadimidine Sodium
more than 101.0 per cent of C12H14N4O2S, calculated on the
dried basis. C12H13N4NaO2S Mol. Wt. 300.3
Description. White or almost white crystals or powder. Sulphadimidine Sodium is the sodium salt of N 1-(4,6-
dimethylpyrimidin-2-yl)sulphanilamide.
Identification Sulphadimidine Sodium contains not less than 98.0 per cent
Test A may be omitted if tests B, C and D are carried out. Tests and not more than 101.0 per cent of C12H13N4NaO2S, calculated
C and D may be omitted if tests A and B are carried out. on the dried basis.

A. Determine by infrared absorption spectrophotometry (2.4.6). Description. White or creamy white crystals or powder;
Compare the spectrum with that obtained with sulphadimidine odourless or almost odourless; hygroscopic.
RS or with the reference spectrum of sulphadimidine. Identification
B. In the test for Related substances, the principal spot in the
A. Dissolve 0.1 g in 10 ml water, acidify with 1 M hydrochloric
chromatogram obtained with test solution (b) corresponds to
acid, filter, wash the precipitate with water and dry the residue
that in the chromatogram obtained with the reference solution.
at 105º.
C. Heat 3 g in a test-tube inclined at an angle of about 45º with
On the residue determine by infrared absorption
the lower part immersed in a silicone oil-bath at about 270º. It
spectrophotometry (2.4.6). Compare the spectrum with that
decomposes and a white or yellowish white sublimate is
obtained with sulphadimidine RS or with the reference
produced. The sublimate, after recrystallisation from toluene
spectrum of sulphadimidine.
and drying at 100º melts at 150º to 154º (2.4.31).
B. Acidify a solution of 0.1 g in 5 ml of water with 6 M acetic
D. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
acid. A precipitate is produced which, after washing with cold
dilute 1 ml of this solution to 10 ml with water. The solution,
water and drying at 105º, gives the reaction of primary aromatic
without further acidification, gives the reaction of primary
amines (2.3.1), producing a bright orange-red precipitate.
aromatic amines (2.3.1).
C. The washed and dried precipitate obtained in test B melts
Tests at about 198º (2.4.21).

Appearance of solution. A 5.0 per cent w/v solution in 1 M D. Incinerate 0.5 g. The residue, when moistened with
sodium hydroxide is not more intensely coloured than hydrochloric acid and introduced on a platinum wire into the
reference solution YS5, BYS5 or GYS5 (2.4.1). flame of a Bunsen burner, imparts a yellow colour to the flame.

Acidity. Heat 1.25 g of the finely powdered substance at about Tests


70º with 25 ml of carbon dioxide-free water for 5 minutes.
Appearance of solution. A 33.3 per cent w/v solution is clear
Cool for about 15 minutes in ice and filter. To 20 ml of the
(2.4.1), and not more intensely coloured than reference solution
filtrate add 0.1 ml of bromothymol blue solution. Not more
YS4 (2.4.1).
than 0.2 ml of 0.1 M sodium hydroxide is required to change
the colour of the solution. pH (2.4.24). 10.0 to 11.0, determined in a 10.0 per cent w/v
solution.
Related substances (2.3.7). Complies with test C.
Related substances (2.3.7). Complies with test A, but using as
Heavy metals (2.3.13). 1.0 g complies with the limit test for the test solution a solution prepared by dissolving the
heavy metals, Method B (20 ppm). substance under examination in 1 volume of strong ammonia
Sulphated ash (2.3.18). Not more than 0.1 per cent. solution and then diluting with 9 volumes of ethanol (95 per
cent) to produce a 1.0 per cent w/v solution.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105º. Loss on drying (2.4.19). Not more than 2.0 per cent, determined
on 1.0 g by drying in an oven at 105º.
Assay. Weigh accurately about 0.25 g, dissolve in 20 ml of 2 M
hydrochloric acid and 50 ml of water, add 3 g of potassium Assay. Weigh accurately about 0.3 g, dissolve in a mixture of
bromide, cool in ice and carry out the nitrite titration (2.3.31). 75 ml of water and 10 ml of hydrochloric acid, add 3 g of
potassium bromide, cool in ice and carry out the nitrite titration
1 ml of 0.1 M sodium nitrite is equivalent to 0.02783 g of (2.3.31).
C12H14N4O2S.
1 ml of 0.1 M sodium nitrite is equivalent to 0.03003 g of
Storage. Store protected from light and moisture. C12H13N4NaO2S.

1140
IP 2007 SULPHADIMIDINE TABLETS

Storage. Store protected from light and moisture. Add 3 g of potassium bromide, cool in ice and carry out the
nitrite titration (2.3.31).
1 ml of 0.1 M sodium nitrite is equivalent to 0.03003 g of
Sulphadimidine Injection C12H13N4NaO2S.
Sulphadimidine Sodium Injection Storage. Store protected from light, in single dose containers.

Sulphadimidine Injection is a sterile solution of Sulphadimidine Labelling. When the injection is prepared from Sulphadimidine
Sodium in Water for Injections free from dissolved air. It is the strength is stated as the amount of sulphadimidine sodium
prepared either from Sulphadimidine Sodium or by the in a suitable dose-volume.
interaction of Sulphadimidine and Sodium Hydroxide.
Sulphadimidine Injection contains not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
Sulphadimidine Tablets
sulphadimidine sodium, C12H13N4NaO2S. Sulphadimidine Tablets contain not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
Identification sulphadimidine, C12H14N4O2S.
A. Acidify a volume containing 0.1 g of Sulphadimidine Sodium
Identification
with 6 M acetic acid, filter, reserving the filtrate, wash the
precipitate with water and dry at 105º. A. Triturate a quantity of the powdered tablets containing
On the residue determine by infrared absorption 0.5 g of Sulphadimidine with two quantities, each of 5 ml, of
spectrophotometry (2.4.6). Compare the spectrum with that chloroform and discard the chloroform. Triturate the residue
obtained with sulphadimidine RS or with the reference with 10 ml of 5 M ammonia for 5 minutes, add 10 ml of water
spectrum of sulphadimidine. and filter. Warm the filtrate until most of the ammonia has been
removed, cool, acidify with 6 M acetic acid, wash the
B. The residue obtained in test A gives the reaction of primary precipitate with water and dry at 105º.
aromatic amines (2.3.1).
On the residue determine by infrared absorption
C. Evaporate the filtrate obtained in test A and incinerate. The
spectrophotometry (2.4.6). Compare the spectrum with that
residue, when moistened with hydrochloric acid and
obtained with sulphadimidine RS or with the reference
introduced on a platinum wire into the flame of a Bunsen
spectrum of sulphadimidine.
burner, imparts a yellow colour to the flame.
B. In the test for Related substances, the principal spot in the
Tests chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with the reference solution.
Appearance of solution. An injection containing 1.0 g of
Sulphadimidine Sodium in 3 ml is not more intensely coloured C. The residue obtained in test A gives the reaction of primary
than reference solution YS4 (2.4.1). aromatic amines (2.3.1), producing a bright orange-red
precipitate.
pH (2.4.24). 10.0 to 11.0.
Related substances (2.3.7). Complies with test A, using Tests
following solutions. Related substances (2.3.7). Complies with test C, but using
Test solution. The injection under examination diluted with the following solutions.
water to contain 0.2 per cent w/v of sulphadimidine sodium Test solution (a). Extract a quantity of the powdered tablets
Reference solution. A 0.002 per cent w/v solution of containing 0.5 g of Sulphadimidine with 25 ml of a mixture of
sulphanilamide in a mixture of 1 volume of strong ammonia 9 volumes of methanol and 1 volume of strong ammonia
solution and 9 volumes of ethanol (95 per cent). solution by shaking for 10 minutes, filter and use the filtrate.
Other tests. Complies with the tests stated under Parenteral Test solution (b). Dilute 1 volume of test solution (a) to
Preparations (Injections). 5 volumes with a mixture of 24 volumes of methanol and
1 volume of strong ammonia solution.
Assay. Dilute an accurately measured volume containing about
0.5 g of Sulphadimidine Sodium to 75 ml with water, add 10 ml Test solution (c). Dilute 1 volume of test solution (a) to
of hydrochloric acid and pass air slowly through the solution 200 volumes with the same solvent mixture.
until the odour of sulphur dioxide is no longer detectable and Reference solution. A 0.4 per cent w/v of sulphadimidine RS
the vapours do not turn moistened starch iodate paper blue. in the same solvent mixture.

1141
SULPHADOXINE IP 2007

Other tests. Comply with the tests stated under Tablets. D. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
Assay. Weigh and powder 20 tablets. Weigh accurately a dilute 1 ml to 10 ml with water. The resulting solution, without
quantity of the powder containing about 0.25 g of further acidification, gives the reaction of primary aromatic
Sulphadimidine, dissolve as completely as possible in a mixture amines (2.3.1).
of 50 ml of water and 10 ml of hydrochloric acid, add 3 g of Tests
potassium bromide, cool in ice and carry out the nitrite titration
(2.3.31). Appearance of solution. Dissolve 0.8 g in 10 ml of 1 M sodium
hydroxide. The solution is not more intensely coloured than
1 ml of 0.1 M sodium nitrite is equivalent to 0.02783 g of
reference solution YS5, BYS5 or GYS5 (2.4.1).
C12H14N4O2S.
Acidity. Heat 1.25 g of the finely powdered substance at about
Storage. Store protected from light and moisture.
70º with 25 ml of carbon dioxide-free water for 5 minutes.
Cool for about 15 minutes in ice and filter. To 20 ml of the
filtrate add 0.1 ml of bromothymol blue solution. Not more
Sulphadoxine than 0.2 ml of 0.1 M sodium hydroxide is required to change
the colour of the solution.
Sulphormethoxine; Sulphoethomidine
Related substances (2.3.7). Complies with test C, but using
OCH3 the following solution.
H3CO Test solution (a). A 2 per cent w/v solution of the substance
O O N
under examination in a mixture of 24 volumes of methanol and
S 1 volume of strong ammonia solution.
N N
H Heavy metals (2.3.13).1.0 g complies with the limit test for
H2N heavy metals, Method B (20 ppm).
C12H14N4O4S Mol. Wt. 310.3 Sulphated ash (2.3.18). Not more than 0.1 per cent.
Sulphadoxine is N1-(5,6-dimethoxypyrimidin-4- Loss on drying (2.4.19). Not more than 0.5 per cent, determined
yl)sulphanilamide. on 1.0 g by drying in an oven at 105º.
Sulphadoxine contains not less than 99.0 per cent and not Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of 2 M
more than 101.0 per cent of C12H14N4O4S, calculated on the hydrochloric acid, add 3 g of potassium bromide, cool in ice
dried basis. and carry out the nitrite titration (2.3.31).
Description. White or yellowish white crystals or crystalline 1 ml of 0.1 M sodium nitrite is equivalent to 0.03103 g of
powder. C12H14N4O4S.
Storage. Store protected from light and moisture.
Identification
Test A may be omitted if tests B, C and D are carried out. Tests
B and D may be omitted if tests A and C are carried out.
Sulphafurazole
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with sulphadoxine Sulfisoxazole
RS or with the reference spectrum of sulphadoxine.
B. In the test for Related substances, the principal spot in the O O O N
S CH3
chromatogram obtained with test solution (b) corresponds to N
that in the chromatogram obtained with the reference solution. H
CH3
H2N
C. Dissolve 0.5 g in 1 ml of sulphuric acid (40 per cent),
heating gently to effect solution, and continue heating until a C11H13N3O3S Mol. Wt. 267.3
crystalline precipitate is produced. Allow to cool, add 10 ml of 1
2 M sodium hydroxide, cool again, add 25 ml of ether and Sulphafurazole is N -(3,4-dimethylisoxazol-5-
shake for 5 minutes. Dry the upper layer over anhydrous yl)sulphanilamide.
sodium sulphate, filter and evaporate the solvent by heating Sulphafurazole contains not less than 99.0 per cent and not
on a water-bath. The residue melts either at 80º to 82º or at 90º more than 101.0 per cent of C11H13N3O3S, calculated on the
to 92º (2.4.21). dried basis.

1142
IP 2007 SULPHAFURAZOLE TABLETS

Description. White or yellowish white crystals or crystalline Reference solution (b). A 0.4 per cent w/v of sulphafurazole
powder; odourless. RS in a mixture of 1 volume of strong ammonia solution and
24 volumes of methanol.
Identification
Apply to the plate 5 µl of each solution. After development,
Test A may be omitted if tests B, C and D are carried out. Tests dry the plate at 105º and examine in ultraviolet light at 254 nm.
B and D may be omitted if tests A and C are carried out. Any secondary spot in the chromatogram obtained with the
test solution (a) is not more intense than the spot in the
A. Determine by infrared absorption spectrophotometry (2.4.6).
chromatogram obtained with reference solution (a).
Compare the spectrum with that obtained with sulphafurazole
RS or with the reference spectrum of sulphafurazole. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Sulphated ash (2.3.18). Not more than 0.1 per cent.
that in the chromatogram obtained with reference solution (b). Loss on drying (2.4.19). Not more than 0.5 per cent, determined
C. Dissolve 0.5 g in 1 ml of sulphuric acid (40 per cent), on 1.0 g by drying in an oven at 105º.
heating gently to effect solution, and continue heating until a Assay. Weigh accurately about 0.5 g, dissolve in 50 ml of dry
crystalline precipitate is produced. Allow to cool, add 10 ml of acetone. Titrate with 0.1 M tetrabutylammonium hydroxide,
2 M sodium hydroxide, cool again, add 25 ml of ether and using 0.4 per cent w/v solution of thymol blue in methanol as
shake for 5 minutes. Dry the upper layer over anhydrous indicator. Carry out a blank titration.
sodium sulphate, filter and evaporate the solvent by heating
on a water-bath. The residue melts at 119º to 123º (2.4.21). 1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to
0.02673 g of C11H13N3O3S.
D. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
dilute 1 ml to 10 ml with water. The resulting solution, without Storage. Store protected from light and moisture.
further acidification, gives the reaction of primary aromatic
amines (2.3.1).

Tests Sulphafurazole Tablets


Appearance of solution. Dissolve 0.8 g in 10 ml of 1 M sodium Sulphafurazole Tablets contain not less than 95.0 per cent and
hydroxide. The solution is not more intensely coloured than not more than 105.0 per cent of the stated amount of
reference solution YS5, BYS5 or GYS5 (2.4.1). sulphafurazole, C11H13N3O3S.
Acidity. Heat 1.25 g of the finely powdered substance at about Identification
70º with 25 ml of carbon dioxide-free water for 5 minutes.
Cool for about 15 minutes in ice and filter. To 20 ml of the A. Shake a quantity of the powdered tablets containing 0.6 g
filtrate add 0.1 ml of bromothymol blue solution. Not more of Sulphafurazole with 30 ml of acetone, filter and evaporate
than 0.2 ml of 0.1 M sodium hydroxide is required to change the filtrate to dryness; Dry the residue for 2 hours at 105º.
the colour of the solution. On the residue determine by infrared absorption
Related substances. Determine by thin-layer chromatography spectrophotometry (2.4.6). Compare the spectrum with that
(2.4.17), coating the plate with silica gel GF254. obtained with sulphafurazole RS or with the reference
spectrum of sulphafurazole.
Mobile phase. A mixture of 75 volumes of dichloromethane,
25 volumes of methanol and 1 volume of strong ammonia B. Dissolve 0.5 g of residue obtained in test A in 1 ml of
solution. sulphuric acid (40 per cent), heating gently to effect solution,
and continue heating until a crystalline precipitate is produced.
Test solution (a). Dissolve 0.2 g of the substance under
Allow to cool, add 10 ml of 2 M sodium hydroxide, cool again,
examination in 10 ml of a mixture of 1 volume of strong ammonia
add 25 ml of ether and shake for 5 minutes. Dry the upper layer
solution and 24 volumes of methanol.
over anhydrous sodium sulphate, filter and evaporate the
Test solution (b). Dissolve 0.4 g of the substance under solvent by heating on a water-bath. The residue melts at 119º
examination in 100 ml of a mixture of 1 volume of strong to 123º (2.4.21).
ammonia solution and 24 volumes of methanol.
C. Extract a quantity of the powdered tablets containing 50 mg
Reference solution (a). Dissolve 10 mg of the substance under of Sulphafurazole with 2 ml of warm dilute hydrochloric acid
examination in 100 ml of a mixture of 1 volume of strong and filter. The filtrate gives the reaction of primary aromatic
ammonia solution and 24 volumes of methanol. amines (2.3.1).

1143
SULPHALENE IP 2007

Tests Sulphalene contains not less than 99.0 per cent and not more
than 101.0 per cent of C11H12N4O3S, calculated on the dried
Related substances. Determine by thin-layer chromatography basis.
(2.4.17), coating the plate with silica gel GF254.
Description. A white or yellowish white powder; odourless or
Mobile phase. A mixture of 75 volumes of dichloromethane, almost odourless.
25 volumes of methanol and 1 volume of strong ammonia
solution. Identification
Test solution (a). Extract a quantity of the powdered tablets A. Determine by infrared absorption spectrophotometry (2.4.6).
containing 0.5 g of Sulphafurazole with 25 ml of a mixture of Compare the spectrum with that obtained with sulphalene RS
90 volumes of methanol and 10 volumes of strong ammonia or with the reference spectrum of sulphalene.
solution by shaking for 10 minutes, filter and use the filtrate.
B. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
Test solution (b). Dissolve 0.4 g of the substance under dilute 1 ml of this solution to 10 ml with water. The solution,
examination in 100 ml of a mixture of 1 volume of strong without further acidification, gives the reaction of primary
ammonia solution and 24 volumes of methanol. aromatic amines (2.3.1).
Reference solution (a). Dissolve 10 mg of the substance under C. Melting range (2.4.21). 175º to 178º.
examination in 100 ml of a mixture of 1 volume of strong
ammonia solution and 24 volumes of methanol. Tests
Reference solution (b). A 0.4 per cent w/v solution of Acidity. Heat 1.25 g of the finely powdered substance at about
sulphafurazole RS in a mixture of 1 volume of strong ammonia 70º for 5 minutes with 25 ml of carbon dioxide-free water.
solution and 24 volumes of methanol. Cool in ice for about 15 minutes and filter. To 20 ml of the
filtrate add 0.1 ml of bromothymol blue solution. Not more
Apply to the plate 5 µl of each solution. After development,
than 0.2 ml of 0.1 M sodium hydroxide is required to change
dry the plate at 105º and examine in ultraviolet light at 254 nm.
the colour of the solution.
Any secondary spot in the chromatogram obtained with test
solution (a) is not more intense than the spot in the Related substances (2.3.7). Complies with test B.
chromatogram obtained with reference solution (a). Heavy metals (2.3.13). Dissolve 1.0 g in 5 ml of 5 M sodium
Other tests. Comply with the tests stated under Tablets. hydroxide and 20 ml of water. The solution complies with the
limit test for heavy metals, Method A (20 ppm).
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 0.5 g of Sulphated ash (2.3.18). Not more than 0.1 per cent.
Sulphafurazole, dissolve as completely as possible in 50 ml of Loss on drying (2.4.19). Not more than 0.5 per cent, determined
dry acetone. Titrate with 0.1 M tetrabutylammonium on 1.0 g by drying in an oven at 105º.
hydroxide, using 0.4 per cent w/v solution of thymol blue in
Assay. Weigh accurately about 0.3 g, dissolve in a mixture of
methanol as indicator. Carry out a blank titration.
20 ml of 2 M hydrochloric acid and 50 ml of water. Add 3 g of
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to potassium bromide, cool in ice and carry out the nitrite titration
0.02673 g of C11H13N3O3S. (2.3.31).
Storage. Store protected from light and moisture. 1 ml of 0.1 M sodium nitrite is equivalent to 0.02803 g of
C11H12N4O3S.
Storage. Store protected from light and moisture.
Sulphalene
Sulphamethopyrazine; Sulphamethoxypyrazine Sulphamethizole
H3CO N N
N O O
O O S
CH3
S N S
N N H
H H2N
H2N
C9H10N4O2S2 Mol. Wt. 270.3
C11H12N4O3S Mol. Wt. 280.3 1
Sulphamethizole is N -(5-methyl-1,3,4-thiadiazol-2-
1
Sulphalene is N -(3-methoxypyrazin-2-yl)sulphanilamide. yl)sulphanilamide.

1144
IP 2007 SULPHAMETHOXAZOLE

Sulphamethizole contains not less than 99.0 per cent and not Apply to the plate 2 µl of each solution. After development,
more than 101.0 per cent of C9H10N4O2S2, calculated on the dry the plate at 105º and examine in ultraviolet light at 254 nm.
dried basis. Any secondary spot in the chromatogram obtained with test
Description. White or yellowish white crystals or crystalline solution (a) is not more intense than the spot in the
powder; odourless. chromatogram obtained with reference solution (a).
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Identification heavy metals, Method B (20 ppm).
Test A may be omitted if tests B, C and D are carried out. Tests Sulphated ash (2.3.18). Not more than 0.1 per cent.
C and D may be omitted if tests A and B are carried out. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
A. Determine by infrared absorption spectrophotometry (2.4.6). on 1.0 g by drying in an oven at 105º.
Compare the spectrum with that obtained with sulphamethizole Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of 2 M
RS or with the reference spectrum of sulphamethizole. hydrochloric acid, add 3 g of potassium bromide, cool in ice
B. In the test for Related substances, the principal spot in the and carry out the nitrite titration (2.3.31).
chromatogram obtained with test solution (b) corresponds to 1 ml of 0.1 M sodium nitrite is equivalent to 0.02703 g of
that in the chromatogram obtained with reference solution C9H10N4O2S2.
(b).
Storage. Store protected from light and moisture.
C. Dissolve 50 mg in 4 ml of methanol and add 0.2 ml of a
4.0 per cent w/v solution of cupric acetate; a flocculent,
yellowish green precipitate is produced which becomes dark
green. Sulphamethoxazole
D. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and CH3
dilute 1 ml of this solution to 10 ml with water. The solution,
O O
without further acidification, gives the reaction of primary S O
aromatic amines (2.3.1). N N
H
Tests H2N

Appearance of solution. Dissolve 0.8 g in 10 ml of 1 M sodium C10H11N3O3S Mol. Wt. 253.3


hydroxide. The solution is not more intensely coloured than 1
Sulphamethoxazole is N -(5-methylisoxazol-3-
reference solution YS5, BYS5 or GYS5 (2.4.1). yl)sulphanilamide.
Acidity. Heat 1.25 g of the finely powdered substance at about Sulphamethoxazole contains not less than 99.0 per cent and
70º with 25 ml of carbon dioxide-free water for 5 minutes. not more than 101.0 per cent of C10H11N3O3S, calculated on the
Cool for about 15 minutes in ice and filter. To 20 ml of the dried basis.
filtrate add 0.1 ml of bromothymol blue solution. Not more
than 0.2 ml of 0.1 M sodium hydroxide is required to change Description. A white or almost white, crystalline powder;
the colour of the solution. almost odourless.
Related substances. Determine by thin-layer chromatography Identification
(2.4.17), coating the plate with silica gel GF254.
Test A may be omitted if tests B, C and D are carried out. Tests
Mobile phase. A mixture of 80 volumes of chloroform and B and C may be omitted if tests A and D are carried out.
15 volumes of methanol.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Test solution (a). Dissolve 0.3 g of the substance under Compare the spectrum with that obtained with
examination in 10 ml of acetone. sulphamethoxazole RS or with the reference spectrum of
Test solution (b). Dissolve 0.3 g of the substance under sulphamethoxazole.
examination in 100 ml of acetone. B. In the test for Related substances, the principal spot in the
Reference solution (a). Dissolve 15 mg of the substance under chromatogram obtained with test solution (b) corresponds to
examination in 100 ml of acetone. that in the chromatogram obtained with the reference solution.
Reference solution (b). A 0.3 per cent w/v solution of C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
sulphamethizole RS in acetone. dilute 1 ml to 10 ml with water. The resulting solution, without

1145
SULPHAPHENAZOLE IP 2007

further acidification, gives the reaction of primary aromatic Description. A white or almost white, crystalline powder.
amines (2.3.1).
D. Melting range (2.4.21). 169º to 172º.
Identification
Test A may be omitted if tests B, C and D are carried out. Tests
Tests
B and C may be omitted if tests A and D are carried out.
Appearance of solution. Dissolve 0.8 g in 10 ml of 1 M sodium
A. Determine by infrared absorption spectrophotometry (2.4.6).
hydroxide. The solution is not more intensely coloured than
Compare the spectrum with that obtained with
reference solution YS5, BYS5 or GYS5 (2.4.1).
sulphaphenazole RS.
Acidity. Heat 1.25 g of the finely powdered substance with
25 ml of carbon dioxide-free water at 70º for 5 minutes. Cool B. In the test for Related substances, the principal spot in the
for about 15 minutes in ice and filter. To 20 ml of the filtrate add chromatogram obtained with test solution (b) corresponds to
0.1 ml of bromothymol blue solution. Not more than 0.3 ml of that in the chromatogram obtained with the reference solution.
0.1 M sodium hydroxide is required to change the colour of C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
the solution. dilute 1 ml of this solution to 10 ml with water. The solution,
Related substances (2.3.7). Complies with test C, but using without further acidification, gives the reaction of primary
the following solution. aromatic amines (2.3.1).
Test solution (a). Dissolve 0.1 g of the substance under D. Melting range (2.4.21). 178º to 183º.
examination in sufficient of a mixture of 1 volume of strong
ammonia solution and 24 volumes of methanol to produce Tests
5 ml.
Appearance of solution. Dissolve 0.8 g in 10 ml of 1 M sodium
Heavy metals (2.3.13). 1.0 g complies with the limit test for
hydroxide. The solution is not more intensely coloured than
heavy metals, Method B (20 ppm).
reference solution YS5, BYS5 or GYS5 (2.4.1).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Acidity. Heat 1.25 g of the finely powdered substance at about
Loss on drying (2.4.19). Not more than 0.5 per cent, determined 70º with 25 ml of carbon dioxide-free water for 5 minutes.
on 1.0 g by drying in an oven at 105º. Cool for about 15 minutes in ice and filter. To 20 ml of the
Assay. Weigh accurately about 0.2 g, dissolve in 50 ml of 2 M filtrate add 0.1 ml of bromothymol blue solution. Not more
hydrochloric acid, add 3 g of potassium bromide, cool in ice than 0.2 ml of 0.1 M sodium hydroxide is required to change
and carry out the nitrite titration (2.3.31). the colour of the solution.
1 ml of 0.1 M sodium nitrite is equivalent to 0.02533 g of Related substances (2.3.7). Complies with test C.
C10H11N3O3S.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Storage. Store protected from light and moisture.
heavy metals, Method B (20 ppm).
Iron (2.3.14). Ignite 2.0 g with 1 g of anhydrous sodium
carbonate, cool, dissolve the residue in 5 ml of hydrochloric
Sulphaphenazole acid and dilute to 35 ml with water. The resulting solution
complies with the limit test for iron (20 ppm).
O O
S N Sulphated ash (2.3.18). Not more than 0.1 per cent.
N N
H Loss on drying (2.4.19). Not more than 0.5 per cent, determined
H 2N on 1.0 g by drying in an oven at 105º.
Assay. Weigh accurately about 0.2 g, dissolve in a mixture of
20 ml of 2 M hydrochloric acid and 50 ml of water. Add 3 g of
C15H14N4O2S Mol. Wt. 314.4
potassium bromide, cool in ice and carry out the nitrite titration
1
Sulphaphenazole is N -(1-phenyl-1H pyrazol-5- (2.3.31).
yl)sulphanilamide.
1 ml of 0.1 M sodium nitrite is equivalent to 0.03144 g of
Sulphaphenazole contains not less than 99.0 per cent and not C15H14N4O2S.
more than 101.0 per cent of C15H14N4O2S, calculated on the
dried basis. Storage. Store protected from light and moisture.

1146
IP 2007 SULPHOBROMOPHTHALEIN SODIUM

Sulphaphenazole Tablets mixture of 50 ml of water and 10 ml of hydrochloric acid, cool


in ice and carry out the nitrite titration (2.3.31).
Sulphaphenazole Tablets contain not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of 1 ml of 0.1 M sodium nitrite is equivalent to 0.03144 g of
sulphaphenazole, C15H14N4O2S. C15H14N4O2S.
Storage. Store protected from light and moisture.
Identification
Mix a quantity of the powdered tablets containing about 1.5 g
of Sulphaphenazole with 30 ml of acetone and heat under a Sulphobromophthalein Sodium
reflux condenser for 5 minutes. Filter, evaporate the filtrate on
a water-bath to a volume of about 10 ml, add 40 ml of water,
Br O
heat for a further 15 minutes and cool in ice; a precipitate is
formed which, after washing with water and drying at 105º, Br
complies with the following tests. O
SO3Na
Test A may be omitted if tests B, C and D are carried out. Tests Br
B and C may be omitted if tests A and D are carried out. Br
OH
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with SO3Na
sulphaphenazole RS or with the reference spectrum of HO
sulphaphenazole.
C20H8Br4Na2O10S2 Mol. Wt. 837.9
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Sulphobromophthalein Sodium is disodium 5,5' -(4,5,6,7-
that in the chromatogram obtained with the reference solution. tetrabromo-1,3-dihydro-3-oxo-isobenzofuran-1,1-diyl)bis(2-
hydroxybenzenesulphonate).
C. Dissolve about 5 mg in 10 ml of 1 M hydrochloric acid and
dilute 1 ml of this solution to 10 ml with water. The solution, Sulphobromophthalein Sodium contains not less than 7.4 per
without further acidification, gives the reaction of primary cent and not more than 8.2 per cent of sulphur, S, and not less
aromatic amines (2.3.1). than 36.0 per cent and not more than 39.0 per cent of bromine,
Br, both calculated on the dried basis.
D. Melting range (2.4.21). 178º to 183º.
Description. A white, crystalline powder; hygroscopic.
Tests
Identification
Related substances (2.3.7). Comply with test C, but using the
A. Absorbance of a 0.0005 per cent w/v solution in 0.05 M
following solutions.
sodium hydroxide at the maximum at about 580 nm, about 0.4
Test solution (a). Extract a quantity of the powdered tablets (2.4.7).
containing 0.5 g of Sulphaphenazole with 25 ml of a mixture of
B. Mix 0.1 g with 0.5 g of sodium carbonate and ignite until
90 volumes of methanol and 10 volumes of strong ammonia
thoroughly charred. Cool, add 5 ml of hot water, heat for
solution by shaking for 10 minutes, filter and use the filtrate.
5 minutes on a water-bath and filter; the filtrate gives the
Test solution (b). Dilute 1 volume of test solution (a) to reactions of bromides (2.3.1).
5 volumes with a mixture of 24 volumes of methanol and
C. Gives reaction A of sodium salts (2.3.1).
1 volume of strong ammonia solution.
Test solution (c). Dilute 1 volume of test solution (a) to Tests
200 volumes with the same solvent mixture.
Appearance of solution. A 2.0 per cent w/v solution in carbon
Reference solution. A 0.4 per cent w/v of sulphaphenazole dioxide-free water is clear (2.4.1), and colourless (2.4.1).
RS in the same solvent mixture.
Halide ions. To 5 ml of a 1.0 per cent w/v solution add 1 ml of
Other tests. Comply with the tests stated under Tablets. dilute nitric acid and 1 ml of silver nitrate solution; not more
Assay. Weigh and powder 20 tablets. Weigh accurately a than a slight opalescence is produced.
quantity of the powder containing about 0.25 g of Calcium. Ignite 5.0 g in a platinum dish until free from carbon,
Sulphaphenazole, dissolve as completely as possible in a cool, add 1 ml of hydrochloric acid and 10 ml of water, heat

1147
SULPHOBROMOPHTHALEIN SODIUM INJECTION IP 2007

on a water-bath for 5 minutes, add 1 g of ammonium sulphate Sulphobromophthalein Sodium


and 4 ml of dilute ammonia solution and heat for a further
5 minutes. Transfer the contents of the dish to a flask with the Injection
aid of 50 ml of water, add 20 ml of strong ammonia solution, Sulphobromophthalein Sodium Injection is a sterile solution
dilute to 100 ml with water, add 0.3 ml of sodium sulphide of Sulphobromophthalein Sodium in Water for Injections.
solution, 1 ml of potassium cyanide solution and 75 ml of
ethanol (95 per cent) and titrate with 0.01 M disodium edetate Sulphobromophthalein Sodium Injection contains not less than
using 0.1 g of a mixture of 99 parts of potassium nitrate and 95.0 per cent and not more than 105.0 per cent of the stated
1 part of methyl thymol blue as indicator until a faint grey amount of sulphobromophthalein sodium, C20H8Br4Na2O10S2.
colour is produced. Not more than 6.25 ml of 0.01 M disodium
Identification
edetate is required.
Sulphates. To 10 ml of a 0.2 per cent w/v solution add 0.25 ml A. To 5 ml add 0.15 ml of 1 M sodium hydroxide; an intense
of dilute hydrochloric acid, boil and add 1 ml of barium purple colour is produced which disappears on the addition
chloride solution; the hot solution remains clear for 2 minutes. of an acid.

Loss on drying (2.4.19). Not more than 5.0 per cent determined B. To a volume containing 50 mg of Sulphobromophthalein
on 1.0 g by drying in an oven at 105º. Sodium add 0.25 g of sodium carbonate, evaporate to dryness
and ignite. Cool the residue, add 2.5 ml of hot water, heat for
Assay. For sulphur — Determine by the oxygen-flask method 5 minutes on a water-bath and filter; the filtrate gives the
(2.3.34), burning 0.2 g in a 1000-ml glass-stoppered flask and a reactions of bromides (2.3.1).
mixture of 30 ml of water and 0.5 ml of hydrogen peroxide
solution (100 vol) as the absorbing liquid. When the process Tests
is complete, add 2 ml of hydrochloric acid, dilute to 250 ml
with water, heat to boiling and slowly add 10 ml of barium pH (2.4.24). 5.0 to 6.5, determined by using a suitable agar-
chloride solution. Heat on a water-bath for 1 hour, filter, wash potassium nitrate salt bridge.
the precipitate with water, dry and ignite at a temperature of Bacterial endotoxins (2.2.3). Not more than 1.0 Endotoxin Unit
about 600º until, after further ignition, two successive per mg of Sulphobromophthalein Sodium.
weighings do not differ by more than 0.2 per cent.
Other tests. Complies with the tests stated under Parenteral
1 g of residue is equivalent to 0.1374 g of sulphur, S. Preparations (Injections).
For bromine — Determine by the oxygen-flask method Assay. To an accurately measured volume containing 0.1 g of
(2.3.34), burning 0.2 g in a 1000-ml glass-stoppered flask and a Sulphobromophthalein Sodium add sufficient water to
mixture of 10 ml of 0.1 M sodium hydroxide, 0.5 ml of hydrogen produce 500.0 ml and mix. To 5.0 ml of this solution add
peroxide solution (100 vol) and 10 ml of water as the sufficient of a 1.0 per cent w/v solution of sodium carbonate
absorbing liquid. When the process is complete, boil the to produce 200.0 ml and mix. Measure the absorbance of the
solution for 5 minutes, cool, acidify with 2 M nitric acid, add resulting solution at the maximum at about 580 nm (2.4.7),
20.0 ml of 0.1 M silver nitrate, shake, and titrate the excess of using as the blank the sodium carbonate solution.
silver nitrate with 0.1 M ammonium thiocyanate using ferric
Calculate the content of C 20 H 8 Br 4 Na 2 O 10 S 2 from the
ammonium sulphate solution as indicator.
absorbance obtained by carrying out the determination
1 ml of 0.1 M silver nitrate is equivalent to 0.007991 g of simultaneously on 0.1 g, accurately weighed, of
bromine, Br. sulphobromophthalein sodium RS.
Storage. Store protected from moisture.

1148
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

T
Talc ....
Tamoxifen Citrate ....
Tamoxifen Tablets ....
Tartaric Acid ....
Tenofovir Disoproxil Fumartea ....
Tenofovir Disoproxil Fumarate Tablets ....
Tenofovir and Emtricitabine Tablets ....
Terbutaline Sulphate ....
Terbutaline Inhalation ....
Terbutaline Injection ....
Terbutaline Tablets ....
Testosterone Propionate ....
Testosterone Propionate Injection ....
Tetracycline ....
Tetracycline Hydrochloride ....
Tetracycline Capsules ....
Tetracycline Ointment ....
Theophylline ....
Theophylline Injection ....
Thiabendazole ....
Thiabendazole Tablets ....
Thiacetazone ....
Thiacetazone And Isoniazid Tablets ....
Thiamine Hydrochloride ....
Thiamine Injection ....
Thiamine Tablets ....
Thiamine Mononitrate ....
Thiomersal ....
Thiopentone Sodium ....
Thiopentone Injection ....

1149
MONOGRAPHS INDIAN PHARMACOPOEIA 2007

Thiotepa ....
Thiotepa Injection ....
Thymol ....
Thyroxine Sodium ....
Thyroxine Tablets ....
Timolol Maleate ....
Timolol Eye Drops ....
Timolol Tablets ....
Tinidazole ....
Tinidazole Tablets ....
Tiotropium Bromide Monohydrate ....
Tiotropium powder for Inhalation ....
Titanium Dioxide ....
Tizanidine Hydrochloride ....
Tizanidine Tablets ....
Tobramycin ....
Tobramycin Injection ....
Tocopheryl Acetate ....
Tolbutamide ....
Tolbutamide Tablets ....
Topotecan Hydrochloride ....
Topotecan Injection ....
Triamcinolone ....
Triamcinolone Tablets ....
Triamcinolone Acetonide ....
Triamcinolone Acetonide Injection ....
Triamterene ....
Triamterene Capsules ....
Trifluoperazine Hydrochloride ....
Trifluoperazine Injection ....
Trifluoperazine Tablets ....
Triflupromazine Hydrochloride ....
Triflupromazine Injection ....

1150
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

Triflupromazine Tablets ....


Trimethoprim ....
Trimethoprim and Sulphamethoxazole Oral Suspension ....
Trimethoprim And Sulphamethoxazole Tablets ....
Trimethoprim Tablets ....
Triprolidine Hydrochloride ....
Triprolidine Tablets ....
Trisodium Edetate Concentrate for Injection ....
Tropicamide ....
Tropicamide Eye Drops ....
Troxidone ....
Troxidone Capsules ....
Tubocurarine Chloride ....
Tubocurarine Injection ....

1151
IP 2007 TAMOXIFEN CITRATE

Talc Organic compounds. The residue obtained in the test for Loss
on drying is not more than slightly yellow or grey.
Purified Talc; Talcum
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Talc is a powdered, selected natural hydrated magnesium on 1.0 g by drying in an oven at 180º for 1 hour.
silicate. It may contain varying amounts of aluminium and
Storage. Store protected from moisture.
iron in forms insoluble in 1M sulphuric acid.
Description. A white or almost white powder, free from
grittiness; readily adheres to the skin; unctuous to the touch;
odourless. Tamoxifen Citrate
Identification O CH3
N
A. When examined microscopically, shows irregular plates, CH3 HO COOH
the majority less than 50 µm in length. The particles are not ,
H3C HOOC COOH
notably stained by a 0.1 per cent w/v solution of methylene
blue in ethanol (95 per cent).
B. Melt 0.5 g in a metal crucible with 1 g of potassium nitrate
and 3 g of anhydrous sodium carbonate, add 20 ml of boiling C26H29NO,C6H8O7 Mol. Wt. 563.7
water, mix and filter. Wash the residue with 50 ml of water. Mix
the residue with a mixture of 0.5 ml of hydrochloric acid and Tamoxifen Citrate is (Z)-2-[4-(1,2-diphenylbut-1-enyl)-1-
5 ml of water and filter. To the filtrate add 1 ml of 9 M ammonia phenoxy]ethyldimethylamine citrate.
and 1 ml of ammonium chloride solution and filter. To the Tamoxifen Citrate contains not less than 99.0 per cent and not
filtrate add 1 ml of disodium hydrogen phosphate solution; a more than 101.0 per cent of C26H29NO, C6H8O7, calculated on
white, crystalline precipitate is produced. the dried basis.
C. Gives the reaction of silicates (2.3.1). Description. A white or almost white, crystalline powder.

Tests Identification
Acidity or alkalinity. Shake 5.0 g with 25 ml of carbon dioxide- Test A may be omitted if tests B, C and D are carried out. Tests
free water for 1 minute, filter and add to the filtrate 0.5 ml of C and D may be omitted if tests A and B are carried out.
bromothymol blue solution; the solution is not acid and
requires not more than 0.3 ml of 0.1 M hydrochloric acid to A. Determine by infrared absorption spectrophotometry (2.4.6).
make it acid. Compare the spectrum with that obtained with tamoxifen
citrate RS or with the reference spectrum of tamoxifen citrate.
Iron (2.3.14). Boil 4.0 g with 25 ml of water for 30 minutes,
replacing the water lost by evaporation, and filter. The filtrate, B. When examined in the range 220 nm to 360 nm (2.4.7), a
after the addition of 5 ml of nitric acid, complies with the limit 0.002 per cent w/v solution in methanol shows absorption
test for iron (10 ppm). maxima at about 237 nm and 275 nm.

Acid-soluble substances. Not more than 2.0 per cent, C. Determine by thin-layer chromatography (2.4.17), coating
determined by the following method. Digest 2.0 g with 40 ml of the plate with silica gel GF254.
dilute hydrochloric acid for 15 minutes, filter, evaporate the Mobile phase. A mixture of 90 volumes of toluene and
filtrate; to the residue add 0.1 ml of sulphuric acid and ignite 10 volumes of triethylamine.
to constant weight.
Test solution. Dissolve 0.1 g of the substance under
Water-soluble substances. Shake 5.0 g with 25 ml of water for examination in 10 ml of methanol.
1 minute, filter, evaporate the filtrate and dry to constant weight;
Reference solution. A 1 per cent w/v solution of tamoxifen
the residue weighs not more than 10 mg.
citrate RS in methanol.
Carbonates. To 1 g add 20 ml of dilute hydrochloric acid; no
Apply to the plate 5 µl of each solution. After development,
effervescence is produced.
dry the plate in air and examine in ultraviolet light at 254 nm.
Chlorides (2.3.12). Suspend 2.0 g in 10 ml of water, add 10 ml The principal spot in the chromatogram obtained with the test
of 2 M nitric acid, shake for 15 minutes and filter. 10 ml of the solution corresponds to that in the chromatogram obtained
filtrate complies with the limit test for chlorides (250 ppm). with the reference solution.

1153
TAMOXIFEN TABLETS IP 2007

D. To 10 mg add 4 ml of pyridine and 2 ml of acetic anhydride in the chromatogram obtained with reference solution (a) and
and shake; a yellow colour is produced. Heat in a water-bath the sum of the areas of all such peaks is not greater than the
for 2 minutes; a pink to red colour is produced. area of the peak due to tamoxifen in the chromatogram obtained
with reference solution (a). Ignore any peak with a retention
Tests time less than 2.5 minutes and any peak with an area less than
0.05 times the area of the principal peak in the chromatogram
E-Isomer and related substances. Determine by liquid obtained with reference solution (a).
chromatography (2.4.14).
Heavy metals (2.3.13). 2.0 g complies with limit the test for
Test solution. Dissolve 0.25 g of the substance under heavy metals, Method B (10 ppm).
examination in 100 ml of a mixture of 60 volumes of acetonitrile,
25 volumes of water and 15 volumes of tetrahydrofuran. Sulphated ash (2.3.18). Not more than 0.2 per cent.
Reference solution (a). A 0.0025 per cent w/v solution of the Loss on drying (2.4.19). Not more than 0.5 per cent, determined
substance under examination in a mixture of 60 volumes of on 1.0 g by drying in an oven at 105º.
acetonitrile, 25 volumes of water and 15 volumes of Assay. Weigh accurately about 1.0 g, dissolve in 150 ml of
tetrahydrofuran. anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
Reference solution (b). A 0.25 per cent w/v solution of acid, using 1-naphtholbenzein solution as indicator. Carry
tamoxifen citrate impurity standard RS in the same solvent out a blank titration.
mixture. 1 ml of 0.1 M perchloric acid is equivalent to 0.05636 g of
Chromatographic system C26H29NO, C6H8O7.
– a stainless steel column 20 cm x 5 mm, packed with Storage. Store protected from light and moisture.
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 60 volumes of acetonitrile,
25 volumes of water, 15 volumes of tetrahydrofuran
and 0.4 volume of strong ammonia solution,
– flow rate. 1.5 ml per minute, Tamoxifen Tablets
– spectrophotometer set at 240 nm,
– a 20 µl loop injector. Tamoxifen Citrate Tablets
In the chromatogram obtained with reference solution (b) a Tamoxifen Tablets contain not less than 90.0 per cent and not
peak due to E-isomer appears immediately following the peak more than 110.0 per cent of the stated amount of tamoxifen,
due to Z-tamoxifen. Adjust the sensitivity of the instrument C26H29NO.
so that the height of the peak due to E-isomer is about 15 per
cent of the full-scale deflection on the recorder. Measure the Identification
height of the peak due to E-isomer by dropping a perpendicular
A. To a quantity of the powdered tablets containing 0.1 g of
from the top of the peak to a line drawn tangentially between
tamoxifen add 20 ml of water, warm, add 2 ml of 5 M sodium
the troughs on each side of the E-isomer peak or the trough
hydroxide and cool. Extract with two quantities, each of 10 ml,
between the E- and Z-isomer peaks and the baseline, whichever
of ether, filtering after each extraction. Combine the ether
is appropriate.
extracts and evaporate to dryness in a stream of nitrogen at
The test is not valid unless the height of the trough separating room temperature. Dry the residue at a pressure not exceeding
the peaks due to E- and Z-tamoxifen in the chromatogram 0.7 kPa for 30 minutes.
obtained with reference solution (b) is less than 7 per cent of
On the residue, determine by infrared absorption
full-scale deflection on the recorder and the retention time of
spectrophotometry (2.4.6). Compare the spectrum with that
the principal peak is less than 30 minutes. (The retention time
obtained with tamoxifen citrate RS or with the reference
decreases with increasing concentration of ammonia in the
spectrum of tamoxifen citrate.
mobile phase).
B. When examined in the range 220 nm to 360 nm (2.4.7), a
The content of E-isomer in the substance under examination
0.002 per cent w/v solution in methanol of the residue obtained
is not more than 1 per cent calculated from the declared content
in test A shows absorption maxima at about 237 nm and
of E-isomer in tamoxifen citrate impurity standard RS. The
275 nm.
area of any secondary peak in the chromatogram obtained
with the test solution, other than a peak due to E-isomer, is C. Determine by thin-layer chromatography (2.4.17), coating
not greater than half that of the peak due to tamoxifen citrate the plate with silica gel GF254.

1154
IP 2007 TAMOXIFEN TABLETS

Mobile phase. A mixture of 100 volumes of ethyl acetate, cent of full-scale deflection on therecorder. Measure the height
10 volumes of methanol and 1 volume of strong ammonia of the peak due to E-isomer by dropping a perpendicular from
solution. the top of the peak to a line drawn tangentially between the
Test solution. Shake a quantity of the powdered tablets troughs on each side of the E-isomer peak or the trough
containing 0.1 g tamoxifen with 10 ml of methanol, filter, between the E- and Z-isomer peaks and the baseline, whichever
evaporate the filtrate to dryness on a water-bath, dry the is appropriate.
residue at 100º for 30 minutes and dissolve 10 mg of the residue The test is not valid unless the height of the trough separating
in 10 ml of methanol. the peaks due to E- and Z-tamoxifen in the chromatogram
Reference solution. A 0.1 per cent w/v solution of tamoxifen obtained with reference solution (b) is less than 7 per cent of
citrate RS in methanol. the full-scale deflection on the recorder and the retention time
of the principal peak is less than 30 minutes. (The retention
Apply to the plate 10 µl of each solution. After development, time decreases with increasing concentration of ammonia in
dry the plate in air and examine in ultraviolet light at 254 nm. the mobile phase).
The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained The content of E-isomer in the substance under examination
with the reference solution. is not more than 1 per cent calculated from the declared content
of E-isomer in tamoxifen citrate impurity standard RS. The
D. Dissolve 10 mg of the residue obtained in the preparation area of any secondary peak in the chromatogram obtained
of the test solution in test C in 4 ml of pyridine, add 2 ml of
with the test solution, other than a peak due to E-isomer, is
acetic anhydride and heat on a water-bath for 2 minutes; a
not greater than half that of the peak due to tamoxifen citrate
pink to red colour is produced.
in the chromatogram obtained with reference solution (a) and
the sum of the areas of all such peaks is not greater than the
Tests
area of the peak due to tamoxifen in the chromatogram obtained
E-Isomer and Related substances. Determine by liquid with reference solution (a). Ignore any peak with a retention
chromatography (2.4.14). time less than 2.5 minutes and any peak with an area less than
0.05 times the area of the principal peak in the chromatogram
Test solution. Mix a quantity of the powdered tablets obtained with reference solution (a).
containing 60 mg of tamoxifen with 60 ml of a mixture of
60 volumes of acetonitrile, 25 volumes of water and Uniformity of content. (For tablets containing equivalent of
15 volumes of tetrahydrofuran with the aid of ultrasound for 10 mg or less) — Comply with the test stated under Tablets.
5 minutes, dilute to 100 ml with the same solvent mixture and Crush one tablet and transfer to a 100-ml volumetric flask with
filter through Whatman No. 1 filter paper. the aid of 75 ml of methanol. Shake well for 5 minutes, add
Reference solution (a). Dilute 1.0 ml of the test solution to sufficient methanol to produce 100.0 ml and filter. Dilute
100 ml with the same solvent mixture. 10.0 ml of the filtrate to 100.0 ml with methanol. Measure the
absorbance of the resulting solution at the maximum at about
Reference solution (b). A 0.1 per cent w/v solution of 275 nm (2.4.7). Calculate the content of C26H29NO in the tablet
tamoxifen citrate impurity standard RS in the same solvent taking 325 as the specific absorbance at 275 nm.
mixture.
Other tests. Comply with the tests stated under Tablets.
Chromatographic system
– a stainless steel column 20 cm x 5 mm, packed with Assay. Weigh and powder 20 tablets. Weigh accurately a
octadecylsilane bonded to porous silica (5 µm), quantity of the powder containing about 25 mg of tamoxifen,
– mobile phase: a mixture of 60 volumes of acetonitrile, shake with 100 ml of methanol for 15 minutes, add sufficient
25 volumes of water, 15 volumes of tetrahydrofuran methanol to produce 250.0 ml and filter. Dilute 10.0 ml of the
and 0.4 volume of strong ammonia solution, filtrate to 100.0 ml with methanol and measure the absorbance
– flow rate. 1.5 ml per minute, of the resulting solution at the maximum at about 275 nm (2.4.7).
– spectrophotometer set at 240 nm, Calculate the content of C26H29NO taking 325 as the specific
– a 20 µl loop injector. absorbance at 275 nm.
In the chromatogram obtained with reference solution (b) a Storage. Store protected from light and moisture.
peak due to E-isomer appears immediately following the peak
due to Z-tamoxifen. Adjust the sensitivity of the instrument Labelling. The label states the strength in terms of the
so that the height of the peak due to E-isomer is about 15 per equivalent amount of tamoxifen.

1155
TARTARIC ACID IP 2007

Tartaric Acid Assay. Weigh accurately about 1.0 g, dissolve in 25 ml of


water and titrate with 1 M sodium hydroxide using 0.5 ml of
L-Tartaric Acid phenolphthalein solution as indicator.
1 ml of 1 M sodium hydroxide is equivalent to 0.07505 g of
H OH C4H6O6.
COOH Storage. Store protected from moisture.
HOOC
H OH

C4H6O6 Mol. Wt. 150.1 Tenofovir Disoproxil Fumarate


Tartaric Acid is (2R,3R)-2,3-dihydroxybutanedioic acid. NH2
Tartaric Acid contains not less than 99.5 per cent and not N N
more than 101.0 per cent of C4H6O6, calculated on the dried O CH3
N N O H COOH
basis.
O P O O O CH3 ,
Description. Colourless crystals or a white or almost white O O O CH3 HOOC H
CH3
crystalline powder.
O CH3

Identification C19H30N5O10P,C4H4O4 Mol. Wt. 635.5


A. Ignite a few mg; it gradually decomposes giving off an Tenofovir Disoproxil Fumarate salt of
odour resembling that of burnt sugar (distinction from citric bis(isopropyloxycarbonyloxymethyl ester of (R)-9-(2-
acid). phosphonomethoxypropyl)adenine with fumaric acid.
B. A 10 per cent w/v solution in distilled water (solution A) is Tenofovir Disoproxil Fumarate contain not less than 97.0 per
strongly acidic. cent and not more than 102. 0 per cent of C19H30N5O10P,C4H4O4,
calculated on the anhydrous basis.
C. Gives reactions A and B of tartrates (2.3.1).
Description. A white to off- white crystalline powder.
Tests
Identification
Appearance of solution. Solution A is clear (2.4.1), and not
more intensely coloured than reference solution YS6 (2.4.1). A. Determine by infrared absorption spectrophotometry
(2.4.6). Compare the spectrum with that obtained with tenofovir
Specific optical rotation (2.4.22). +12.0 º to +12.8º, determined disoproxil fumarate RS or with the reference spectrum of
in a 20.0 per cent w/v solution. tenofovir disoproxil fumarate.
Arsenic (2.3.10). Dissolve 5.0 g in 50 ml of water and add 10 ml B. In the Assay, the principal peak in the chromatogram
of stannated hydrochloric acid AsT. The resulting solution obtained with the test solution corresponds to the peak in the
complies with the limit test for arsenic (2 ppm). chromatogram obtained with the reference solution.
Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method A (10 ppm). Tests
Chlorides (2.3.12). 20 ml of solution A complies with the limit Related substances. Determine by liquid chromatography
test for chlorides (125 ppm). (2.4.14).
Sulphates (2.3.17). 10 ml of solution A complies with the limit NOTE — Prepare the solutions immediately before use.
test for sulphates (150 ppm). Test solution. Dissolve 100 mg of the substance under
Oxalate. Neutralise 10 ml of solution A with dilute ammonia examination in 50 ml of methanol.
solution, add 0.1 ml of dilute acetic acid and 10 ml of calcium Reference solution (a). A 0.2 per cent w/v solution of tenofovir
sulphate solution; no opalescence is produced within disoproxil fumarate RS in methanol.
20 minutes.
Reference solution (b). Dilute 1.0 ml of reference solution (a)
Sulphated ash (2.3.18). Not more than 0.1 per cent. to 100.0 ml with methanol.
Loss on drying (2.4.19). Not more than 0.2 per cent, determined Reference solution (c). Dissolve 10 mg of the fumaric acid in
on 1.0 g by drying in an oven at 105º. 50 ml of methanol.

1156
IP 2007 TENOFOVIR DISOPROXIL FUMARATE TABLETS

Chromatographic system 0.1 per cent v/v of triethylamine in 0.05M sodium


– a stainless steel column 25 cm x 4.6 mm, packed with dihydrogen phosphate with the pH adjusted to 2.3 with
octadecylsilane bonded to porous silica (5 µm) (such as orthophosphoric acid and filtered,
ODS 3V), – flow rate. 1 ml per minute,
– column. temperature 30º, – spectrophotometer set at 210 nm,
– mobile phase: A. 0.1 M ammonium acetate solution – a 10 µl loop injector.
with the pH adjusted to 3.8 with glacial acetic acid, Inject the reference solution. The test is not valid unless the
B. a mixture of 70 volumes of methanol tailing factor is not more than 2.0, the column efficiency in not
and 30 volumes of acetonitrile, less than 2000 theoretical plates and the relative standard
– flow rate. 1.5 ml per minute, deviation for replicate injections is not more than 2.0 per cent.
– a linear gradient programme using the conditions given
below, Inject the test solution and the reference solution.
– spectrophotometer set at 260 nm, Calculate the content of fumaric acid.
– a 10 µl loop injector. Water (2.3.43). Not more than 1.0 per cent, determined on
Time mobile phase A mobile phase B 1.0 g.
(in min.) (per cent v/v) (per cent v/v)
Assay. Determine by liquid chromatography (2.4.14).
0 95 5
NOTE — Prepare the solutions immediately before use and
10 50 50
carry out the test protected from light.
25 50 50
50 20 80 Test solution. Dissolve 50 mg of the substance under
examination in 50.0 ml of the mobile phase. Dilute 5.0 ml of the
60 95 5
solution to 50.0 ml with the mobile phase.
65 95 5
Reference solution. A 0.1 per cent w/v solution of tenofovir
Inject reference solution (a). The test is not valid unless the disoproxil fumarate RS in the mobile phase. Dilute 5.0 ml of
tailing factor is not more than 2.0 and the column efficiency in the solution to 50.0 ml with the mobile phase.
not less than 2000 theoretical plates.
Chromatographic system
Inject the test solution, reference solution (b) and reference – a stainless steel column 25 cm x 4.6 mm, packed with
solution (c). In the chromatogram obtained with the test octadecylsilane bonded to porous silica (5 µm),
solution, the area of any secondary peak is not more than – mobile phase: a mixture of 40 volumes of acetonitrile
the area of the peak in the chromatogram obtained with the and 60 volumes of a buffer solution prepared by adding
reference solution (b) (1.0 per cent) and the sum of all the 1 ml triethylamine to 0.05M sodium dihydrogen
secondary peaks is not more than 2.5 times the area of the phosphate with the pH adjusted to 2.3 with
peak in the chromatogram obtained with the reference solution orthophosphoric acid and filtered,
(b) (2.5 per cent). Ignore any peak corresponding to the peak – flow rate. 1 ml per minute,
obtained in the chromatogram in the reference solution (c) – spectrophotometer set at 260 nm,
and any peak having area less than 0.02 times the area of the – a 20 µl loop injector.
principal peak in the chromatogram obtained with reference
Inject the reference solution. The test is not valid unless the
solution (b) (0.02 per cent).
tailing factor is not more than 2.0, the column efficiency in not
Fumaric Acid. 17.5 per cent to 19.0 per cent. less than 2000 theoretical plates and the relative standard
Determine by liquid chromatography (2.4.14). deviation for replicate injections is not more than 2.0 per cent.
Test solution. Dissolve 25 mg of the substance under Inject the test solution and the reference solution.
examination in 100 ml of the mobile phase. Calculate the content of C19H30N5O10P,C4H4O4.
Reference solution. Dissolve 25 mg of the fumaric acid in Storage. Store protected from light in a refrigerator (2º to 8º).
50 ml of the mob.ile phase. Dilute 10 ml of the solution to 100
ml with the mobile phase.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with
Tenofovir Disoproxil Fumarate Tablets
octadecylsilane bonded to porous silica (5 µm), Tenofovir Disoproxil Fumarate Tablets contain not less than
– mobile phase: a mixture of 40 volumes of acetonitrile 90.0 per cent and not more than 110.0 per cent of the stated
and 60 volumes of a buffer solution prepared by adding amount of tenofovir disoproxil fumarate, C19H30N5O10P,C4H4O4.

1157
TENOFOVIR DISOPROXIL FUMARATE TABLETS IP 2007

Identification Time mobile phase A mobile phase B


(in min.) (per cent v/v) (per cent v/v)
A. In the Assay, the principal peak in the chromatogram
0 95 5
obtained with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. 10 50 50
25 50 50
B. When examined in the range 200 nm to 400 nm (2.4.7), a
50 20 80
0.001 per cent w/v solution in methanol shows an absorption
maximum at the same wavelength as the reference solution. 60 95 5
Inject reference solution (a). The test is not valid unless the
Tests tailing factor is not more than 2.0 and the column efficiency in
Dissolution (2.5.2). not less than 2000 theoretical plates.

Apparatus. No 1 Inject the test solution, reference solutions (b) and (c). In the
Medium. 900 ml of 0.1 M hydrochloric acid. chromatogram obtained with the test solution, the area of any
secondary peak is not more than 3.5 times the area of the peak
Speed and time. 50 rpm and 45 minutes
in the chromatogram obtained with the reference solution (b)
Withdraw a suitable volume of the medium and filter promptly. (3.5 per cent) and the sum of areas of all the secondary peaks
Dilute the filtrate, if necessary, with the same solvent. Measure is not more than 6 times the area of the peak in the
the absorbance (2.4.7) of the resulting solution at the maximum chromatogram obtained with the reference solution (b)
at about 260 nm. Calculate the content of C19H30N5O10P,C4H4O4 (6.0 per cent). Ignore the peak corresponding to the peak in
in the medium from the absorbance obtained from a solution the chromatogram obtained in the reference solution (c).
of known concentration of tenofovir disoproxil fumarate RS.
Other tests. Comply with the tests stated under Tablets.
D. Not less than 80 per cent of the stated amount of
Water (2.3.43). Not more than 5.0 per cent, determined on
C19H30N5O10P,C4H4O4.
0.5 g.
Related substances. Determine by liquid chromatography Assay: Determine by liquid chromatography (2.4.14).
(2.4.14).
NOTE — Prepare the solutions immediately before use.
NOTE — Prepare the solutions immediately before use.
Solvent mixture. Equal volumes of water and methanol.
Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing 100 mg of Tenofovir Test solution. Weigh and powder 20 tablets. Weigh accurately
Disoproxil Fumarate and disperse in 50 ml of methanol and a quantity of the powder containing 300 mg of Tenofovir
filter. Disoproxil Fumarate, dissolve in 100 ml of solvent mixture and
filter. Dilute 5.0 ml of the filtrate to 50.0 ml with the mobile
Reference solution (a). A solution of tenofovir disoproxil phase.
fumarate RS containing 0.2 per cent of tenofovir disoproxil in
methanol. Reference solution. A 0.120 per cent w/v solution of tenofovir
disoproxil fumarate RS in the solvent mixture. Dilute 5.0 ml of
Reference solution (b). Dilute 1 ml of reference solution (a) to the solution to 20.0 ml with the mobile phase.
100 ml with methanol.
Chromatographic system
Reference solution (c). Dissolve 10 mg of fumaric acid in – a stainless steel column 15 cm x 4.6 mm, packed with
50 ml of methanol. octadecylsilane bonded to porous silica (5 µm) (such as
Chromatographic system Inertsil ODS 3V),
– a stainless steel column 25 cm x 4.6 mm, packed with – mobile phase: a mixture of 60 volumes of a buffer solution
octadecylsilane bonded to porous silica (5 µm), prepared by dissolving 7.8 g of sodium dihydrogen
– mobile phase: A. dissolve 1.9 g of ammonium acetate orthophosphate in 1000 ml of water, adding 1 ml of
in 1000 ml of water and adjust the pH to 3.8 with glacial triethylamine and adjusting the pH to 2.3 with
acetic acid, orthophosphoric acid (10 per cent v/v), and 40 volumes
B. methanol, of acetonitrile,
– flow rate. 1.5 ml per minute, – flow rate. 1 ml per minute,
– a linear gradient programme using the conditions given – spectrophotometer set at 260 nm,
below, – a 10 µl loop injector.
– spectrophotometer set at 260 nm, Inject the reference solution. The test is not valid unless the
– a 10 µl loop injector. tailing factor is not more than 2.0, the column efficiency in not

1158
IP 2007 TENOFOVIR AND EMTRICITABINE TABLETS

less than 2000 theoretical plates and the relative standard disperse in 5 ml of methanol, dilute to 50 ml with mobile phase
deviation for replicate injections is not more than 2.0 per cent. A and filter.
Inject the test solution and the reference solution. Reference solution (a). A 0.1 per cent w/v solution of
emtricitabine RS in mobile phase A.
Calculate the content of C19H30N5O10P,C4H4O4 in the tablets.
Reference solution (b). Dilute 1 ml of reference solution (a) to
Storage. Store protected from moisture, at a temperature not 100 ml with mobile phase A.
exceeding 30º.
Chromatographic system,
– a stainless steel column 25 cm x 4.6 mm, packed with
octadecysilane bonded to porous silica (5µm),
Tenofovir and Emtricitabine Tablets – column temperature 35º,
– mobile phase: A. a mixture of 95 volumes of a buffer
Tenofovir Disoproxil Fumarate and Emtricitabine Tablets solution prepared by dissolving 1.9 g of ammonium
Tenofovir and Emtricitabine Tablets contain not less than acetate in 1000 ml of water, adjusted the pH to
90.0 per cent and not more than 110.0 per cent of the stated 3.8 with glacial acetic acid, and 5 volumes of methanol
amounts of tenofovir disoproxil fumarate, C19H30N5O10P,C4H4O4 and filtered,
and emtricitabine, C8H10FN3O3S. B. a filtered mixture of 30 volumes of the
buffer solution and 70 volumes of methanol,
Identification – flow rate. 1 ml per minute,
– a linear gradient programme using the conditions given
In the Assay, the principal peak in the chromatogram obtained
below,
with the test solution corresponds to the peak in the
– spectrophotometer set at 277 nm,
chromatogram obtained with the reference solution.
– a 20 µl loop injector.
Tests Time Mobile phase A Mobile phase B
(in min.) (per cent v/v) (per cent v/v)
Dissolution (2.5.2).
0.01 100 0
Apparatus. No 1 30 100 0
Medium. 900 ml of 0.1 M hydrochloric acid, 35 0 100
Speed and time. 50 rpm and 45 minutes. 55 0 100
Withdraw a suitable volume of the medium and filter. 60 100 0
Determine by liquid chromatography (2.4.14) 75 100 0

Test solution. The filtrate obtained as given above. Dilute the Inject reference solution (a). The test is not valid unless the
filtrate if necessary, with the dissolution medium. column efficiency is not less than 500 theoretical plates and
the tailing factor is not more than 2.0.
Reference solution. A solution containing 0.32 per cent w/v of
tenofovir disoproxil fumarate RS and 0.24 per cent w/v of Inject the test solution and reference solution (b). In the
emtricitabine RS in methanol. Dilute 5 ml of the solution to chromatogram obtained with the test solution, the area of any
50 ml with the dissolution medium. secondary peak is not more than the area of the peak in the
chromatogram obtained with the reference solution (b)
Use the chromatographic system given under Assay. (1.0 per cent) and the sum of areas of all the secondary peaks
Inject the test solution and the reference solution. is not more than 3 times the area of the peak in the
chromatogram obtained with the reference solution (b) (3.0
Calculate the contents of C 19 H 30 N 5 O 10 P.C 4 H 4 O 4 and per cent).
C8H10FN3O3S.
For Tenofovir Disoproxil Fumarate
D. Not less than 75 per cent of the stated amounts of
C19H30N5O10P.C4H4O4 and C8H10FN3O3S. Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing 50 mg of Tenofovir
Related substances. Determine by liquid chromatography Disoproxil Fumarate and disperse in 50 ml of methanol.
(2.4.14).
Reference solution (a). A 0.1 per cent w/v solution of tenofovir
For Emtricitabine disoproxil fumarate RS in methanol.
Test solution. Weigh and powder 20 tablets. Weigh accurately Reference solution (b). Dilute 1 ml of reference solution (a) to
a quantity of the powder containing 50 mg of Emtricitabine, 100 ml with methanol.

1159
TERBUTALINE SULPHATE IP 2007

Chromatographic system – mobile phase: A. a buffer solution prepared by


– a stainless steel column 25 cm x 4.6 mm packed with dissolving 1.35 g of monobasic potassium
octadecylsilane bonded to porous silica (5µm), phosphate in 1000 ml of water and adjusting the pH to
– mobile phase: A. a buffer solution prepared by 3.0 with orthophosphoric acid,
dissolving 1.9 g of ammonium acetate in 1000 ml of B. a mixture of 20 volumes of the buffer
water and adjusting the pH to 3.8 with glacial acetic solution and 80 volumes of acetonitrile,
acid, – flow rate. 1.5 ml per minute,
B. methanol, – a linear gradient programme using the conditions given
– flow rate. 1.5 ml per minute, below,
– a linesr gradient programme using the conditions given – spectrophotometer set at 260 nm,
below, – a 10 µl loop injector.
– spectrophotometer set at 260 nm, Time Mobile phase A Mobile phase B
– a 20 µl loop injector. (in mins) (per cent) (per cent)
Time Mobile phase A Mobile phase B 0 94 6
(in mins) (per cent) (per cent) 3 94 6
0 95 5 5 50 50
10 50 50 8 35 65
25 50 50 9 94 6
50 20 80 12 94 6
60 95 5
Inject the reference solution. The test is not valid unless the
75 95 5 tailing factor is not more than 2.0, the column efficiency in not
Inject reference solution (a). The test is not valid unless the less than 2000 theoretical plates for the peak due to tenofovir
column efficiency is not less than 2000 theoretical plates and disoproxil, 500 theoretical plates for the peak due to
the tailing factor is not more than 2.0. emtricitabine and the relative standard deviation for replicate
Inject the test solution and reference solution (b). In the injections is not more than 2.0 per cent for each component.
chromatogram obtained with the test solution, the area of any Inject the test solution and the reference solution.
secondary peak is not more than 3.5 times the area of the peak
Calculate the contents of C 19 H 30 N 5 O 10 P,C 4 H 4 O 4 and
in the chromatogram obtained with the reference solution (b)
C8H10FN3O3S in the tablets.
(3.5 per cent) and the sum of areas of all the secondary peaks
is not more than 6 times the area of the peak in the Storage. Store protected from moisture, at a temperature not
chromatogram obtained with the reference solution (b) (6.0 exceeding 30º.
per cent).
Other tests. Comply with the tests stated under the Tablets.
Assay. Determine by liquid chromatography (2.4.14). Terbutaline Sulphate
Solvent mixture. 30 volumes of water and 70 volumes of
methanol.
OH H
Test solution. Weigh and powder 20 tablets. Weigh accurately
a quantity of the powder containing 100 mg of Tenofovir HO N CH3
CH3 , H2SO4
Disoproxil Fumarate, disperse in 10 ml of water and dilute to
CH3
250.0 ml with methanol. Dilute 5.0 ml of the solution to 25.0 ml
with the solvent mixture. OH
Reference solution. A solution containing 0.025 per cent w/v 2
of emtricitabine RS and 0.04 per cent w/v of tenofovir
disoproxil fumarate RS in methanol. Dilute 5.0 ml of the (C12H19NO3)2,H2SO4 Mol. Wt. 548.7
solution to 25.0 ml with the solvent mixture. Terbutaline Sulphate is (RS)-2-(tert-butylamino)-1-(3,5-
Chromatographic system dihydroxyphenyl)ethanol sulphate.
– a stainless steel column 4.6 mm x 5 cm packed with Terbutaline Sulphate contains not less than 98.0 per cent and
octadecylsilane bonded to porous silica (3µm), not more than 101.0 per cent of (C12H19NO3)2, H2SO4, calculated
– column temperature 40º, on the dried basis.

1160
IP 2007 TERBUTALINE INHALATION

Description. A white or almost white, crystalline powder; Wash the plate with the mobile phase and apply to the plate
odourless or almost odourless. 20 µl of each solution. After development, dry the plate in air,
spray with dilute potassium permanganate solution. Any
Identification secondary spot in the chromatogram obtained with test
solution (a) is not more intense than the spot in the
Test A may be omitted if tests B, C and D are carried out. Tests
chromatogram obtained with reference solution (a).
B and C may be omitted if tests A and D are carried out.
Heavy metals (2.3.13). Mix 1.6 g with 0.6 g of anhydrous sodium
A. Determine by infrared absorption spectrophotometry (2.4.6).
sulphate and ignite without melting the sodium sulphate. Cool,
Compare the spectrum with that obtained with terbutaline
add 3 ml of 2 M hydrochloric acid, boil and dilute to 50 ml
sulphate RS or with the reference spectrum of terbutaline
with water. Cool and filter, 25 ml of the filtrate complies with
sulphate.
the limit test for heavy metals, Method A (25 ppm).
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
0.007 per cent w/v solution in 0.1 M hydrochloric acid shows
on 1.0 g by drying in an oven at 105º for 3 hours.
absorption maxima at about 276 nm and 280 nm, which may be
fused; absorbance at both 276 nm and 280 nm, 0.46 to 0.49. Assay. Weigh accurately about 0.5 g, dissolve in 60 ml of
anhydrous glacial acetic acid with the aid of heat. Titrate
C. In the test for Related substances, the principal spot in the
with 0.1 M perchloric acid, determining the end-point
chromatogram obtained with test solution (b) corresponds to
potentiometrically (2,4,25). Carry out a blank titration.
that in the chromatogram obtained with reference solution (b).
1 ml of 0.1 M perchloric acid is equivalent to 0.05486 g of
D. A 2.0 per cent w/v solution in carbon dioxide-free water
(C12H19NO3)2, H2SO4.
gives reaction A of sulphates (2.3.1).
Storage. Store protected from light and moisture.
Tests
Labelling. The label states (1) the number of metered doses in
Appearance of solution. A 2.0 per cent w/v solution in carbon the container; (2) the amount of active ingredient delivered
dioxide-free water is clear (2.4.1); absorbance of a 2-cm layer per inhalation; (3) that the container should be shaken before
at 400 nm, not more than 0.11 (2.4.7). use each time; (4) a warning that the container is pressurised
and must be kept away from heat and direct sunlight and that
Acidity. Dissolve 0.2 g in 10 ml of carbon dioxide-free water
it must not be punctured, broken or incinerated even when
and titrate with 0.01 M sodium hydroxide, using methyl red
apparently empty; (5) the warning “Keep out of reach of
solution as indicator. Not more than 1.2 ml of 0.01 M sodium
children”.
hydroxide is required to change the colour of the solution to
yellow.
tert-Butylamino-3,5-dihydroxyacetophenone. Absorbance of
a 2 per cent w/v solution in 0.01 M hydrochloric acid at Terbutaline Inhalation
about 330 nm, not more than 0.50 (2.4.7).
Terbutaline Sulphate Inhalation; Terbutaline Inhalation
Related substances. Determine by thin-layer chromatography
Aerosol; Terbutaline Sulphate Inhalation Aerosol
(2.4.17), coating the plate with silica gel G.
Terbutaline Inhalation is a suspension of Terbutaline Sulphate,
Mobile phase. A mixture of 90 volumes of aldehyde-free
as a superfine powder, in a suitable liquid in a suitable
methanol, 10 volumes of water and 1.5 volumes of strong
pressurised container. It may contain suitable pharmaceutical
ammonia solution.
aids such as surfactants, stabilising agents, etc.
Test solution (a). Dissolve 0.25 g of the substance under
Terbutaline Inhalation delivers not less than 75.0 per cent and
examination in 1 ml of water and add sufficient ethanol
not more than 125.0 per cent of the stated amount of terbutaline
(95 per cent) to produce 10 ml.
sulphate (C12H19NO3)2,H2SO4, per inhalation, by actuation of
Test solution (b). Dilute 1 ml of test solution (a) to 5 ml with the valve.
ethanol (95 per cent).
Identification
Reference solution (a). Dilute 1 ml of test solution (a) to
200 ml with ethanol (95 per cent). Determine by thin-layer chromatography (2.4.17), coating the
Reference solution (b). Dissolve 25 mg of terbutaline sulphate plate with silica gel GF254.
RS in 0.5 ml of water and add sufficient ethanol (95 per cent) Mobile phase. A mixture of 65 volumes of 2-propanol, 25
to produce 5 ml. volumes of cyclohexane and 5 volumes of formic acid.

1161
TERBUTALINE INJECTION IP 2007

Test solution. Remove the actuator from the pressurised Labelling. The label states the amount of active ingredient
container, shake the container for about 30 seconds and place delivered per inhalation.
it in an inverted position in a small beaker containing 5 ml of
water. Discharge a sufficient number of deliveries containing
5 mg of Terbutaline Sulphate, under the surface of the solvent.
Reference solution. A 0.1 per cent w/v solution of terbutaline Terbutaline Injection
sulphate RS in water. Terbutaline Sulphate Injection
Apply to the plate 2 µl of each solution. After development, Terbutaline Injection is a sterile solution of Terbutaline
dry the plate in air, spray with a 2 per cent w/v solution of 4- Sulphate in Water for Injections.
aminoantipyrine in methanol. Examine in daylight and in
ultraviolet light at 365 nm. The principal spot in the Terbutaline Injection contains not less than 90.0 per cent and
chromatogram obtained with the test solution corresponds to not more than 110.0 per cent of the stated amount of terbutaline
the chromatogram obtained with reference solution. sulphate (C12H19NO3)2, H2SO4.

Tests Identification
Other tests. Complies with the tests stated under Inhalation Determine by thin-layer chromatography (2.4.17), coating the
Preparations (Pressurised metered-dose Preparations). plate with silica gel G.
Follow the procedure described under Assay wherever the Mobile phase. A mixture of 65 volumes of 2-propanol,
amount of active substance is to be determined in any test. 25 volumes of cyclohexane and 5 volumes of formic acid.
Assay. Carry out the test for Content of active ingredient Test solution. Use the injection.
delivered per actuation stated under Inhalation Preparations Reference solution. A 0.1 per cent w/v solution of terbutaline
(Pressurised metered-dose Preparations). sulphate RS in saline solution.
Use 35 ml of water and finally dilute to 50.0 ml with water. Apply to the plate 2 µl of each solution. After development,
Dilute a suitable volume of this solution with water to produce dry the plate in air, spray with a 2 per cent w/v solution of
a solution containing 50 µg of Terbutaline Sulphate per ml. To 4-aminoantipyrine in methanol. Dry the plate in air and spray
10.0 ml of this solution in a 50-ml volumetric flask add 35 ml of with a freshly prepared 8.0 per cent w/v solution of potassium
buffer solution pH 9.5 and 1.0 ml of 4-aminoantipyrine ferricyanide in a mixture of 4 volumes of strong ammonia
solution. Mix, add 1.0 ml of potassium ferricyanide solution solution and 1 volume of water. The principal spot in the
with vigorous swirling of the flask, dilute to volume with water chromatogram obtained with the test solution corresponds to
and mix. Exactly 75 seconds after the addition of the potassium that in the chromatogram obtained with the reference solution.
ferricyanide solution measure the absorbance of the resulting
solution at the maximum at about 550 nm (2.4.7), using as the Tests
blank a solution prepared in the same manner using 10.0 ml of
water in place of the solution of the substance under pH (2.4.24). 3.0 to 5.0.
examination. Other tests. Complies with the tests stated under Parenteral
Calculate the content of (C12H19NO3)2,H2SO4 in the solution preparations (Injections).
from the absorbance obtained by repeating the operation using Assay. Measure accurately a volume containing about 5 mg
a solution containing 100 µg of terbutaline sulphate RS in of Terbutaline Sulphate and add sufficient water to produce
place of the solution of the substance under examination. 50.0 ml. To 5.0 ml add 35 ml of a buffer solution prepared by
Calculate the amount of (C12H19NO3)2,H2SO4 delivered per dissolving 36.3 g of tris (hydroxymethyl) aminomethane in
actuation of the valve. 900 ml of water, adjusting the pH to between 9.4 and 9.6 and
adding sufficient water to produce 1000 ml. Add 1.0 ml of a
Determine the content of active ingredient a second and third
freshly prepared 2.0 per cent w/v solution of
time by repeating the procedure on the middle ten and on the
4-aminoantipyrine, mix and add 1.0 ml of a freshly prepared
last ten successive combined actuations of the valve. For
8.0 per cent w/v solution of potassium ferricyanide with
each of the three determinations the average content of
vigorous swirling and sufficient of the buffer solution to
(C12H19NO3)2,H2SO4 delivered per actuation of the valve meets
produce 50.0 ml. Exactly 75 seconds after the addition of the
the requirements.
potassium ferricyanide solution measure the absorbance of
Storage. Store protected from moisture at a temperature not the resulting solution at the maximum at about 550 nm (2.4.7),
exceeding 30º. using water as the blank.

1162
IP 2007 TESTOSTERONE PROPIONATE

Calculate the content of (C12H 19NO 3)2, H2SO4 from the Powder one tablet, transfer to a 25-ml volumetric flask, add
absorbance obtained by repeating the operation using a 0.01 15 ml of 0.01 M hydrochloric acid and shake for 10 minutes.
per cent w/v solution of terbutaline sulphate RS and Dilute to volume with 0.01 M hydrochloric acid and filter,
beginning at the words “To 5.0 ml add 35 ml......”. rejecting the first 5 ml of the filtrate. Dilute, if necessary, a
Storage. Store protected from light, in single dose containers. suitable volume of the filtrate with 0.01 M hydrochloric acid
to produce a solution containing 0.01 per cent w/v solution of
Labelling. The label states that the injection should not be Terbutaline Sulphate. Carry out the method as described under
used if the solution is discoloured. Assay beginning at the words “To 5.0 ml add 35 ml of a buffer
solution....”. Calculate the content of (C12H19NO3)2, H2SO4 in
the tablet from the absorbance obtained by carrying out the
Terbutaline Tablets Assay simultaneously using terbutaline sulphate RS.
Terbutaline Sulphate Tablets Other tests. Comply with the tests stated under Tablets.
Terbutaline Tablets contain not less than 90.0 per cent and Assay. Weigh and powder 20 tablets. Weigh accurately a
not more than 110.0 per cent of the stated amount of terbutaline quantity of the powder containing about 5 mg of Terbutaline
sulphate (C12H19NO3)2, H2SO4. Sulphate, transfer to a 50-ml volumetric flask, add 30 ml of
0.01 M hydrochloric acid and shake for 10 minutes. Dilute to
Identification volume with 0.01 M hydrochloric acid and filter, rejecting the
first 5 ml of the filtrate. To 5.0 ml add 35 ml of a buffer solution
A. Shake a quantity of the powdered tablets containing 20 mg prepared by dissolving 36.3 g of tris (hydroxymethyl)
of Terbutaline Sulphate with 50 ml of 0.1 M sodium hydroxide aminomethane in 900 ml of water, adjusting the pH to between
for 10 minutes, dilute to 100 ml with 0.1 M sodium hydroxide 9.4 and 9.6 and adding sufficient water to produce 1000 ml.
and filter. Dilute 20 ml of the filtrate to 50 ml with 0.1 M sodium Add 1.0 ml of a freshly prepared 2.0 per cent w/v solution of
hydroxide. 4-aminoantipyrine, mix and add 1.0 ml of a freshly prepared
When examined in the range 230 nm to 360 nm (2.4.7), the 8.0 per cent w/v solution of potassium ferricyanide with
resulting solution shows an absorption maximum at about vigorous swirling and sufficient of the buffer solution to
296 nm. produce 50.0 ml. Exactly 75 seconds after the addition of the
B. Determine by thin-layer chromatography (2.4.17), coating potassium ferricyanide solution measure the absorbance of
the plate with silica gel G. the resulting solution at the maximum at about 550 nm (2.4.7),
using water as the blank.
Mobile phase. A mixture of 65 volumes of 2-propanol,
25 volumes of cyclohexane and 5 volumes of formic acid. Calculate the content of (C12H19NO 3)2, H2SO 4 from the
absorbance obtained by carrying out the Assay simultaneously
Test solution. Shake a quantity of the powdered tablets
using terbutaline sulphate RS.
containing 10 mg of Terbutaline Sulphate with 4 ml of a mixture
of equal volumes of ethanol (95 per cent) and water for Storage. Store protected from light and moisture.
10 minutes, centrifuge and use the clear supernatant liquid.
Reference solution (a). A 0.25 per cent w/v solution of
terbutaline sulphate RS in water.
Reference solution (b). A mixture of equal volumes of the test Testosterone Propionate
solution and reference solution (a).
Apply to the plate 2 µl of each solution. After development, O
dry the plate in air, allow to stand for a few minutes in an CH3
atmosphere saturated with diethylamine and spray with H 3C O
diazotised nitroaniline solution. The principal spot in the
chromatogram obtained with the test solution corresponds to H3C H
that in the chromatogram obtained with reference solution (a)
H H
and the principal spot in the chromatogram obtained with
reference solution (b) appears as a single compact spot. O

Tests C22H32O3 Mol. Wt. 344.5


Uniformity of content. Comply with the test stated under Testosterone Propionate is 3-oxoandrost-4-en-17β-yl
Tablets propionate.

1163
TESTOSTERONE PROPIONATE INJECTION IP 2007

Testosterone Propionate contains not less than 97.0 per cent The test is not valid unless in the chromatogram obtained
and not more than 103.0 per cent of C22H32O3, calculated on with reference solution (a) the resolution between the peaks
the dried basis. due to testosterone and testosterone acetate is not less than
4.0.
Description. A white or almost white powder or colourless
crystals; odourless. In the chromatogram obtained with the test solution the area
of any peak other than the principal peak is not more than
Identification 0.5 times the area of the principal peak in the chromatogram
obtained with reference solution (b) (0.5 per cent); the sum of
Test A may be omitted if tests B and C are carried out. Tests B the areas of any secondary peaks is not greater than the area
and C may be omitted if test A is carried out. of the principal peak in the chromatogram obtained with
A. Determine by infrared absorption spectrophotometry (2.4.6). reference solution (b) (1.0 per cent). Ignore any peak with an
Compare the spectrum with that obtained with testosterone area less than 0.05 times the area of the principal peak in the
propionate RS or with the reference spectrum of testosterone chromatogram obtained with reference solution (b) (0.05 per
propionate. cent).

B. In the test for Related substances the principal peak in the Sulphated ash (2.3.18). Not more than 0.1 per cent.
chromatogram obtained with the test solution corresponds to Loss on drying (2.4.19). Not more than 0.5 per cent, determined
the peak due to testosterone propionate in the chromatogram on 1.0 g by drying in an oven at 105º for 2 hours.
obtained with reference solution (a).
Assay. Weigh accurately about 25 mg, dissolve in sufficient
C. Melting range. 119º to 123º (2.4.21). ethanol to produce 250.0 ml, mix, dilute 5.0 ml to 50.0 ml with
the same solvent and measure the absorbance of the resulting
Tests solution at the maximum at about 241 nm (2.4.7).

Specific optical rotation (2.4.22). +83.0º to +90.0º, determined Calculate the content of C22H32O3 taking 490 as the specific
in a 1.0 per cent w/v solution in ethanol. absorbance at 241 nm.

Related substances. Determine by liquid chromatography Storage. Store protected from light and moisture.
(2.4.14).
Test solution. Dissolve 50 mg of the substance under
examination in methanol and dilute to 50 ml with the same
solvent.
Testosterone Propionate Injection
Reference solution (a). Dissolve 2 mg of the substance under Testosterone Propionate Injection is a sterile solution of
examination and 2 mg of testosterone acetate RS in methanol Testosterone Propionate in Ethyl Oleate or any other suitable
and dilute to 50 ml with the same solvent. ester, in a suitable fixed oil or in any mixture of these.

Reference solution (b). Dilute 1 ml of the test solution to Testosterone Propionate Injection contains not less than
100 ml with methanol. 92.5 per cent and not more than 107.5 per cent of the stated
amount of testosterone propionate, C22H32O3.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with Identification
octadecylsilane bonded to porous silica (5 µm),
Dissolve a volume containing 50 mg of Testosterone Propionate
– mobile phase: a mixture of 20 volumes of water and
in 8 ml of light petroleum (40º to 60º) and extract with three
80 volumes of methanol,
quantities, each of 8 ml, of a mixture of 7 volumes of glacial
– flow rate. 1.5 ml per minute,
acetic acid and 3 volumes of water. Wash the combined
– spectrophotometer set at 254 nm,
extracts with 10 ml of light petroleum (40º to 60º), dilute with
– a 20 µl loop injector.
water until the solution becomes turbid, allow to stand for 2
Inject the test solution and the reference solutions. Continue hours in ice and filter. The precipitate, after washing with water
the chromatography for twice the retention time for and drying at 105º, complies with the following tests.
testosterone propionate.
A. Determine by infrared absorption spectrophotometry (2.4.6).
The relative retention time of about four impurities with Compare the spectrum with that obtained with testosterone
reference to testosterone propionate (retention time, about propionate RS or with the reference spectrum of testosterone
9 minutes) range from 0.5 to about 1.4. propionate.

1164
IP 2007 TETRACYCLINE

B. Determine by thin-layer chromatography (2.4.17), coating Tetracycline


the plate with silica gel G.
Solvent mixture. A mixture of 90 volumes of light petroleum OH O HO O
(40º to 60º) and 10 volumes of liquid paraffin. HO CONH2
Mobile phase. A mixture of 30 volumes of water and
20 volumes of glacial acetic acid.
OH
Test solution. Dissolve 25 mg of the residue in 10 ml of the H
HO CH H
solvent mixture. 3 N(CH3)2
Reference solution (a). A 0.25 per cent w/v solution of C22H24N2O8 Mol. Wt. 444.5
testosterone propionate RS in the solvent mixture.
Tetracycline is (4S,4aS,5aS,6S,12aS)-4-dimethylamino-
Reference solution (b). Mix equal volumes of the test solution 1,4,4a,5,5a,6,11,12a-octahydro-3,6,10,12,12a-pentahydroxy-6-
and reference solution (a). methyl-1,11-dioxonaphthacene-2-carboxamide. It contains a
Place the dry plate in a tank containing a shallow layer of the variable quantity of water.
solvent mixture, allow the solvent mixture to ascend to the Tetracycline contains not less than 88.0 per cent of C22H24N2O8,
top, remove the plate from the tank and allow the solvent to calculated on the dried basis.
evaporate. Use within 2 hours, with the flow of the mobile
Description. A yellow, crystalline powder.
phase in the direction in which the aforementioned treatment
was done. Identification
Apply to the plate 2 µl of each solution. Allow the mobile A. In the test for Assay, the principal peak in the chromatogram
phase to rise 12 cm. Dry the plate in a current of warm air, allow obtained with the test solution corresponds to the peak due
the solvent to evaporate, heat at 120° for 15 minutes and spray to tetracycline hydrochloride in the chromatogram obtained
the hot plate with ethanolic sulphuric acid (20 per cent v/v). with reference solution (a).
Heat at 120° for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal B. To about 2 mg add 5 ml of sulphuric acid; a reddish violet
spot in the chromatogram obtained with the test solution colour develops. Add the solution to 2.5 ml of water; the
corresponds to that in the chromatogram obtained with colour changes to yellow.
reference solution (a). The principal spot in the chromatogram C. Dissolve about 10 mg in a mixture of 1 ml of 2 M nitric acid
obtained with reference solution (b) appears as a single, and 5 ml of water, shake well and add 1 ml of silver nitrate
compact spot.A. solution. Any opalescence produced is not more intense than
that in a solution prepared in the same manner omitting the
Tests substance under examination.
Other tests. Complies with the tests stated under Parenteral Tests
preparations (Injections).
pH (2.4.24). 3.5 to 6.0, determined in a 1.0 per cent w/v
Assay. To an accurately measured volume containing about suspension in carbon dioxide-free water.
0.1 g of Testosterone Propionate add sufficient chloroform to
produce 100.0 ml and mix. Dilute 3.0 ml to 50.0 ml with Specific optical rotation (2.4.22).–260º to –280º, determined at
chloroform and to 5.0 ml of the solution add 10 ml of isoniazid 20º in a 1.0 per cent w/v solution in 0.1 M hydrochloric acid.
solution and sufficient methanol to produce 20.0 ml. Allow to Related substances. Determine by liquid chromatography
stand for 45 minutes and measure the absorbance of the (2.4.14)
resulting solution at the maximum at about 380 nm (2.4.7), Test solution. Dissolve 0.1 g of the substance under
using as the blank a solution prepared by treating 5 ml of examination in 100 ml of a mixture of 68 ml of 0.1 M ammonium
chloroform in the same manner. oxalate and 27 ml of dimethylformamide (diluting solvent).
Calculate the content of C 22H 32O3 from the absorbance Reference solution (a). A 0.0025 per cent w/v solution of
obtained by repeating the operation using a 0.006 per cent 4-epitetracycline hydrochloride RS in the diluting solvent.
w/v solution of testosterone propionate RS in chloroform
and beginning at the words “to 5.0 ml of the solution.....”. Reference solution (b). A solution containing 0.0025 per cent
w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent
Storage. Store protected from light. w/v of tetracycline hydrochloride RS in the diluting solvent.

1165
TETRACYCLINE HYDROCHLORIDE IP 2007

Chromatographic system Calculate the content of C22H24N2O8.


– a stainless steel column 25 cm x 4.6 mm, packed with 1 mg of C22H24N2O8, HCl is equivalent to 0.92 mg of C22H24N2O8.
octylsilane bonded to porous silica (5 to 10 µm),
– mobile phase: a mixture of 680 ml of 0.1 M ammonium Storage. Store protected from light and moisture.
oxalate, 270 ml of dimethylformamide and 50 ml of
0.2 M dibasic ammonium phosphate, the pH of the
mixture being adjusted, if necessary, to 7.6 to 7.7 with
3 M ammonia or 3 M phosphoric acid,
Tetracycline Hydrochloride
– flow rate. 2 ml per minute,
– spectrophotometer set at 280 nm, OH O OH O
HO
– a 20 µl loop injector. CONH2
, HCl
The test is not valid unless the resolution between the two
principal peaks in the chromatogram obtained with reference OH
H H
solution (b) is not less than 1.5. In the chromatogram obtained HO CH3 N(CH3)2
with the test solution, the area of any peak corresponding to
4-epitetracycline is not greater than the area of the principal C22H24N2O8,HCl Mol. Wt. 480.9
peak in the chromatogram obtained with reference solution Tetracycline Hydrochloride is (4S,4aS,5aS,6S,12aS)-4-
(a) and the total area of all the peaks other than the principal dimethylamino-1,4,4a,5,5a,6,11,12a-octahydro-3,6,10,12,12a-
peak is not greater than 5.0 per cent. pentahydroxy-6-methyl-1,11-dioxonaphthacene-2-
Heavy metals (2.3.13). 0.5 g complies with the limit test for carboxamide hydrochloride.
heavy metals, Method B (50 ppm). Use 2.5 ml of lead standard Tetracycline Hydrochloride contains not less than 95.0 per
solution (10 ppm Pb) to prepare the standard. cent and not more than 100.5 per cent of C22H24N2O8, HCl,
Sulphated ash (2.3.18). Not more than 0.5 per cent. calculated on the dried basis.
Loss on drying (2.4.19). Not more than 13.0 per cent, Description. A yellow, crystalline powder.
determined on 0.5 g by drying in an oven at 105º.
Identification
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh accurately about 50 mg of the substance A. In the test for Assay, the principal peak in the chromatogram
under examination and dissolve in 100.0 ml of a mixture of obtained with the test solution corresponds to the peak due
68 ml of 0.1 M ammonium oxalate and 27 ml of to tetracycline hydrochloride in the chromatogram obtained
dimethylformamide (diluting solvent). with reference solution (a).
Reference solution (a). A 0.05 per cent w/v solution of B. To about 2 mg add 5 ml of sulphuric acid; a reddish violet
tetracycline hydrochloride RS in the diluting solvent. colour develops. Add the solution to 2.5 ml of water; the
colour changes to yellow.
Reference solution (b). A solution containing 0.0025 per cent
w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent C. Gives reaction A of chlorides (2.3.1).
w/v of tetracycline hydrochloride RS in the diluting solvent.
Tests
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with pH (2.4.24). 1.8 to 3.0, determined in a 1.0 per cent w/v
octylsilane bonded to porous silica (5 to 10 µm), suspension in carbon dioxide-free water.
– mobile phase: a mixture of 680 ml of 0.1 M ammonium Specific optical rotation (2.4.22). –239º to –255º, determined
oxalate, 270 ml of dimethylformamide and 50 ml of at 20º in a 1.0 per cent w/v solution in 0.1 M hydrochloric
0.2 M dibasic ammonium phosphate, the pH of the acid.
mixture being adjusted, if necessary, to 7.6 to 7.7 with
Related substances. Determine by liquid chromatography
3 M ammonia or 3 M phosphoric acid,
(2.4.14).
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 280 nm, Test solution. Dissolve 0.1 g of the substance under
– a 20 µl loop injector. examination in 100 ml of a mixture of 68 ml of 0.1 M ammonium
oxalate and 27 ml of dimethylformamide (diluting solvent).
The test is not valid unless the resolution between the two
principal peaks in the chromatogram obtained with reference Reference solution (a). A 0.0025 per cent w/v solution of
solution (b) is not less than 1.5. 4-epitetracycline hydrochloride RS in the diluting solvent.

1166
IP 2007 TETRACYCLINE CAPSULES

Reference solution (b). A solution containing 0.0025 per cent The test is not valid unless the resolution between the two
w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent principal peaks in the chromatogram obtained with reference
w/v of tetracycline hydrochloride RS in the diluting solvent. solution (b) is not less than 1.5.
Chromatographic system Calculate the content of C22H24N2O8.
– a stainless steel column 25 cm x 4.6 mm, packed with
Tetracycline Hydrochloride intended for use in the
octylsilane bonded to porous silica (5 to 10 µm),
manufacture of parenteral preparations without a further
– mobile phase: 680 ml of 0.1 M ammonium oxalate,
appropriate procedure for removal of bacterial endotoxins
270 ml of dimethylformamide and 50 ml of 0.2 M dibasic
complies with the following additional requirement.
ammonium phosphate, the pH of the mixture being
adjusted, if necessary, to 7.6 to 7.7 with 3 M ammonia or Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
3 M phosphoric acid, per mg.
– flow rate. 2 ml per minute, Tetracycline hydrochloride intended for use in the
– spectrophotometer set at 280 nm, manufacture of parenteral preparations without a further
– a 20 µl loop injector. appropriate sterilisation procedure complies with the
The test is not valid unless the resolution between the two following additional requirement.
principal peaks in the chromatogram obtained with reference
Sterility (2.2.11). Complies with the test for sterility.
solution (b) is not less than 1.5. In the chromatogram obtained
with the test solution the area of any peak corresponding to Storage. Store protected from light and moisture. If it is
4-epitetracycline is not greater than the area of the principal intended for use in the manufacture of parenteral preparations,
peak in the chromatogram obtained with reference solution the container should be sterile, tamper-evident and sealed so
(a) and the total area of all peaks other than the principal peak as to exclude microorganisms.
is not greater than 5.0 per cent. Labelling. The label states whether or not the material is
Heavy metals (2.3.13). 0.5 g complies with the limit test for intended for use in the manufacture of parenteral preparations.
heavy metals, Method B (50 ppm). Use 2.5 ml of lead standard
solution (10 ppm Pb) to prepare the standard.
Sulphated ash (2.3.18). Not more than 0.5 per cent.
Tetracycline Capsules
Loss on drying (2.4.19). Not more than 2.0 per cent, determined
on 1.0 g by drying in an oven at 60º over phosphorus pentoxide Tetracycline Hydrochloride Capsules
at a pressure not exceeding 0.7 kPa for 3 hours. Tetracycline Capsules contain not less than 90.0 per cent and
Assay. Determine by liquid chromatography (2.4.14). not more than 110.0 per cent of the stated amount of
Test solution. Weigh accurately about 50 mg of the substance tetracycline hydrochloride, C22H24N2O8, HCl.
under examination and dissolve in about 50 ml of the diluting Identification
solvent described in the test for related substances and further
add sufficient diluting solvent to produce 100.0 ml. A. In the test for Assay, the principal peak in the chromatogram
Reference solution (a). A 0.05 per cent w/v solution of obtained with the test solution corresponds to the peak due
tetracycline hydrochloride RS in the diluting solvent. to tetracycline hydrochloride in the chromatogram obtained
with reference solution (a).
Reference solution (b). A solution containing 0.0025 per cent
w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent B. To a quantity of the mixed contents of 20 capsules
w/v of tetracycline hydrochloride RS in the diluting solvent. containing about 10 mg of Tetracycline Hydrochloride add
20 ml of warm ethanol (95 per cent), allow to stand for 20
Chromatographic system
minutes, filter and evaporate the filtrate to dryness on a water-
– a stainless steel column 25 cm x 4.6 mm, packed with
bath. To 0.5 mg of the residue add 2 ml of sulphuric acid; a
octylsilane bonded to porous silica (5 to 10 µm),
reddish violet colour develops. Add the solution to 2.5 ml of
– mobile phase: a mixture of 680 volumes of 0.1 M
water; the colour changes to yellow.
ammonium oxalate, 270 volumes of dimethylformamide
and 50 volumes of 0.2 M dibasic ammonium phosphate, C. The residue obtained in test B gives reaction A of chlorides
the pH of the mixture being adjusted, if necessary, to 7.6 (2.3.1).
to 7.7 with 3 M ammonia or 3 M phosphoric acid,
– flow rate. 1.5 ml per minute, Tests
– spectrophotometer set at 280 nm, Related substances. Determine by liquid chromatography
– a 20 µl loop injector. (2.4.14).

1167
TETRACYCLINE ONITMENT IP 2007

Test solution. Shake a quantity of the mixed contents of Assay. Determine by liquid chromatography (2.4.14).
20 capsules containing about 25 mg of Tetracycline Test solution. Weigh accurately a quantity of the mixed
Hydrochloride with 80 ml of 0.01 M methanolic hydrochloric contents of 20 capsules containing about 50 mg of
acid for 10 minutes, dilute to 100 ml with the same solvent, mix Tetracycline Hydrochloride, shake with about 50 ml of 0.01 M
and filter if necessary. methanolic hydrochloric acid and dilute with the same solvent
Reference solution (a). A 0.002 per cent w/v solution of to produce 100.0 ml, mix and filter. Discard the first few ml of
4-epitetracycline hydrochloride RS in 0.01 M methanolic the filtrate.
hydrochloric acid. Reference solution (a). A 0.05 per cent w/v solution of
Reference solution (b). A solution containing 0.0015 per cent tetracycline hydrochloride RS in 0.01 M methanolic
w/v each of 4-epitetracycline hydrochloride RS and hydrochloric acid.
tetracycline hydrochloride RS in 0.01 M methanolic Reference solution (b). A solution containing 0.0025 per cent
hydrochloric acid. w/v of 4-epitetracycline hydrochloride RS and 0.01 per cent
Chromatographic system w/v of tetracycline hydrochloride RS in 0.01 M methanolic
– a stainless steel column 25 cm x 4.6 mm, packed with hydrochloric acid.
octadecylsilane bonded to porous silica (10 µm), Chromatographic system
– column. temperature 40º, – a stainless steel column 25 cm x 4.6 mm, packed with
– mobile phase: a mixture of 5 volumes of octylsilane bonded to porous silica (5 to10 µm),
dimethylformamide and 95 volumes of 0.1 M oxalic – mobile phase: a mixture of 68 volumes of 0.1 M
acid, the pH of the mixture being adjusted to 3.9 with ammonium oxalate and 27 volumes of
triethylamine, dimethylformamide and 5 volumes of 0.2 M dibasic
– flow rate. 2 ml per minute, ammonium phosphate, the pH of the mixture being
– spectrophotometer set at 280 nm, adjusted, if necessary, to 7.6 to 7.7 with 3 M ammonia or
– a 20 µl loop injector. 3 M phosphoric acid,
The test is not valid unless the resolution between the two – flow rate. 1.5 ml per minute,
principal peaks in the chromatogram obtained with reference – spectrophotometer set at 280 nm,
solution (b) is not less than 2.0. In the chromatogram obtained – a 20 µl loop injector.
with the test solution the area of any peak corresponding to The test is not valid unless the resolution between the two
4-epitetracycline hydrochloride is not greater than the area of principal peaks in the chromatogram obtained with reference
the principal peak in the chromatogram obtained with reference solution (b) is not less than 1.5.
solution (a) and the total area of all the peaks other than the
principal peak is not greater than 10.0 per cent. Calculate the content of C22H24N2O8, HCl in the capsules.

Dissolution (2.5.2). Storage. Store protected from light and moisture.


Apparatus. No 1
Medium. 900 ml of water freshly prepared by distillation.
Speed and time. 75 rpm and 60 minutes. Tetracycline Ointment
Withdraw a suitable volume of the medium and filter through Tetracycline Hydrochloride Eye Ointment
a membrane filter disc having an average pore diameter not
greater than 1.0 µm, rejecting the first few ml of the filtrate. Tetracycline Ointment contains not less than 90.0 per cent
Measure the absorbance of the resulting solution, suitably and not more than 115.0 per cent of the stated amount of
diluted if necessary, at the maximum at about 276 nm (2.4.7). tetracycline hydrochloride, C22H24N2O8, HCl.
Calculate the content of C22H24N2O8, HCl in the medium from
Identification
the absorbance obtained from a solution of known
concentration of tetracycline hydrochloride. In the test for Assay, the principal peak in the chromatogram
D. Not less than 70 per cent of the stated amount of obtained with the test solution corresponds to the peak due
C22H24N2O8, HCl. to tetracycline hydrochloride in the chromatogram obtained
with the reference solution.
Loss on drying (2.4.19). Not more than 3.0 per cent, determined
on 1.0 g of the contents of the capsules by drying in an oven Tests
at 60º at a pressure not exceeding 0.7 kPa for 3 hours.
Water (2.3.43). Not more than 0.5 per cent, determined on
Other tests. Comply with the tests stated under Capsules. 2.0 g dissolved in a mixture of 2 volumes of carbon

1168
IP 2007 THEOPHYLLINE

tetrachloride, 2 volumes of chloroform and 1 volume of Description. A white, crystalline powder; odourless.
methanol.
Identification
Other tests. Complies with the tests stated under Eye
Ointments. Test A may be omitted if tests B, C and D are carried out. Tests
Assay. Determine by liquid chromatography (2.4.14). B and D may be omitted if tests A and C are carried out.

Test solution. Weigh accurately a quantity containing about A. Determine by infrared absorption spectrophotometry (2.4.6).
25 mg of Tetracycline Hydrochloride and transfer to a Compare the spectrum with that obtained with theophylline
separating funnel with the aid of 15 ml of cyclohexane. Add RS or with the reference spectrum of theophylline.
15 ml of a mixture of 68 volumes of 0.1 M ammonium oxalate B. Dissolve about 10 mg in 10 ml of water, add 0.5 ml of a 5 per
and 27 volumes of dimethylformamide (diluting solvent) and cent w/v solution of mercuric acetate and allow to stand; a
shake well. Collect the lower layer in a 50-ml volumetric flask. white, crystalline precipitate is produced.
Repeat the extraction with two further quantities, each of
C. The melting range, after drying at 105º, 270º to 274º (2.4.21).
15 ml, of the diluting solvent, combining the extracts in the
same 50-ml volumetric flask. Add sufficient diluting solvent to D. Gives the reaction of xanthines (2.3.1).
produce 50.0 ml, mix and filter.
Tests
Reference solution. A 0.05 per cent w/v solution of tetracycline
hydrochloride RS in the diluting solvent. Appearance of solution. Dissolve 0.5 g in 75 ml of carbon
Chromatographic system dioxide-free water with heating; the resulting solution
– a stainless steel column 25 cm x 4.6 mm, packed with (solution A), is clear (2.4.1), and colourless (2.4.1).
octylsilane bonded to porous silica (5 to10 µm), Acidity. To 50 ml of solution A add 0.1 ml of methyl red solution.
– mobile phase: a mixture of 68 volumes of 0.1 M The solution is red and not more than 1.0 ml of 0.01 M sodium
ammonium oxalate and 27 volumes of hydroxide is required to change the colour of the solution to
dimethylformamide and 5 volumes of 0.2 M dibasic yellow.
ammonium phosphate, the pH of the mixture being
Light absorption (2.4.7). Absorbance of a 0.001 per cent w/v
adjusted, if necessary, to 7.6 to 7.7 with 3 M ammonia or
solution in 0.1 M hydrochloric acid at the maximum at about
3 M phosphoric acid,
270 nm, not less than 0.53.
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 280 nm, Related substances. Determine by thin-layer chromatography
– a 20 µl loop injector. (2.4.17), coating the plate with silica gel GF254.
Calculate the content of C22H24N2O8, HCl in the eye ointment. Mobile phase. A mixture of 40 volumes of 1-butanol,
30 volumes of acetone, 30 volumes of chloroform and
Storage. Store protected from moisture.
10 volumes of strong ammonia solution.
Test solution. Dissolve 0.2 g of the substance under
examination in 10 ml of a mixture of 60 volumes of chloroform
Theophylline and 40 volumes of methanol.
O Reference solution. Dissolve 10 mg of the substance under
H examination in 100 ml of a mixture of 60 volumes of chloroform
H3C N
N and 40 volumes of methanol.

O N N Apply to the plate 10 µl of each solution. After development,


dry the plate in air and examine in ultraviolet light at 254 nm.
CH3
Any secondary spot in the chromatogram obtained with the
C7H8N4O2 Mol. Wt. 180.2 (anhydrous) test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
C7H8N4O2,H2O Mol. Wt. 198.2 (hydrate)
Heavy metals (2.3.13).1.0 g complies with the limit test for
Theophylline is 1,3-dimethyl-3,7-dihydro-1H-purine-2,6-
heavy metals, Method B (20 ppm).
dione. It contains one molecule of water or is anhydrous.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Theophylline contains not less than 98.0 per cent and not
more than 101.0 per cent of C7H8N4O2, calculated on the dried Loss on drying (2.4.19). Not more than 0.5 per cent (for the
basis. anhydrous form) and 8.0 per cent to 9.5 per cent (for the

1169
THEOPHYLLINE INJECTION IP 2007

hydrated form), determined on 1.0 g by drying in an oven at at a flow rate of about 3.5 ml per minute, discarding the first
105º. 50 ml of the eluate. Measure the absorbance of the eluate at
Assay. Weigh accurately about 0.25 g, add 50 ml of water and 284 nm, using water as the blank; the absorbance is not more
gently warm the mixture on a water-bath until complete solution than 0.25 (2.4.7).
is effected. Cool, add 20.0 ml of 0.1 M silver nitrate and 1.0 ml Other tests. Complies with the tests stated under Parenteral
of bromothymol solution and titrate with 0.1 M sodium Preparations (Injections).
hydroxide until a blue colour is obtained.
Assay. For theophylline — Dilute a suitable volume of the
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01802 g of injection with sufficient 0.1 M sodium hydroxide to produce a
C7H8N4O2. solution containing 0.008 per cent w/v of anhydrous
theophylline. Measure the absorbance of the resulting
Storage. Store protected from moisture.
solution at the maximum at about 274 nm (2.4.7), using as the
blank a solution prepared in the same manner omitting the
substance under examination.
Theophylline Injection Calculate the content of C7H8N4O2 from the absorbance
obtained by repeating the operation using theophylline RS in
Theophylline in Dextrose Injection place of the substance under examination.
Theophylline Injection is a sterile solution of Theophylline For dextrose — Transfer an accurately measured volume of
and Dextrose in Water for Injections. the injection containing about 5 mg of Dextrose to a 100-ml
Theophylline Injection contains not less than 92.5 per cent volumetric flask, add 0.2 ml of 6 M ammonia, dilute to volume
and not more than 107.5 per cent of the stated amount of with water and mix. Determine the optical rotation of the
anhydrous theophylline, C7H8N4O2, and not less than 95.0 per resulting solution in a suitable polarimeter tube at 25º (2.4.22).
cent and not more than 105.0 per cent of the stated amount of The observed rotation, in degrees multiplied by 1.0425A,
dextrose, C6H12O6, H2O. represents the weight, in g, of C6H12O6,H2O in the volume of
the injection taken for the assay, where A is the ratio 200 divided
Identification by the length, in mm, of the polarimeter tube employed.
A. When examined in the range 230 nm to 360 nm (2.4.7), the Storage. Store protected from light, in single dose containers.
solution obtained in the Assay for theophylline shows an Labelling. The label states the strength in terms of the amounts
absorption maximum at about 274 nm. of anhydrous theophylline and Dextrose.
B. Add 0.2 ml of the injection to 5 ml of potassium cupri-
tartrate solution and heat to boiling; a red to orange precipitate
is formed.
Thiabendazole
Tests Tiabendazole
pH (2.4.24). 3.5 to 6.5.
H
5-Hydroxymethylfurfural and Related substances. Use a glass N S
chromatographic column (66 cm ´ 11 mm) with a sealed-in,
coarse-porosity sintered disc or a glass wool plug and fitted N N
with a stopcock. Mix 8 g of a 20- to 50-mesh
styrenedivinylbenzene anion-exchange resin in the hydroxide C10H7N3S Mol. Wt. 201.3
form with 25 ml of water, allow to settle and decant the Thiabendazole is 2-(1,3-thiazol-4-yl)-1H-benzimidazole.
supernatant liquid until a slurry of resin remains. Pour the
Thiabendazole contains not less than 98.0 per cent and not
slurry into the column and allow to settle as a homogeneous
more than 101.0 per cent of C10H7N3S, calculated on the
bed having a bed volume of about 15 ml. Wash the resin bed
anhydrous basis.
at a flow rate of about 3 ml per minute with 100 ml of a 5.7 per
cent w/v solution of ammonium carbonate followed by Description. A white or almost white, crystalline powder.
washing with water until the eluate has a pH of 7.
Identification
Dilute an accurately measured volume of the injection
containing 1.0 g of Dextrose, C6H12O6,H2O, to 250.0 ml with Test A may be omitted if tests B, C and D are carried out. Tests
water. Pass this solution through the resin bed in the column B, C and D may be omitted if test A is carried out.

1170
IP 2007 THIABENDAZOLE TABLETS

A. Determine by infrared absorption spectrophotometry (2.4.6). Sulphated ash (2.3.18). Not more than 0.2 per cent.
Compare the spectrum with that obtained with thiabendazole Water (2.3.43). Not more than 0.5 per cent, determined on
RS or with the reference spectrum of thiabendazole. 1.0 g.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Assay. Weigh accurately about 0.15 g, dissolve in 30 ml of
0.0005 per cent w/v solution in 0.1 M hydrochloric acid shows
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
absorption maxima at about 243 nm and 302 nm; ratio of the
acid, determining the end-point potentiometrically (2.4.25).
absorbance at the maximum at about 302 nm to that at about
Carry out a blank titration.
243 nm, 1.8 to 2.1.
C. In the test for Related substances, the principal spot in the 1 ml of 0.1 M perchloric acid is equivalent to 0.02013 g of
chromatogram obtained with the test solution (b) corresponds C10H7N3S.
to that in the chromatogram obtained with reference solution Storage. Store protected from light and moisture.
(c).
D. Dissolve 5 mg in 5 ml of 0.1 M hydrochloric acid, add 3 mg
of 4-phenylenediamine dihydrochloride and shake until Thiabendazole Tablets
dissolved. Add 0.1 g of zinc powder, mix, allow to stand for
2 minutes and add 10 ml of ferric ammonium sulphate solution; Tiabendazole Tablets
a deep blue or bluish violet colour is produced. Thiabendazole Tablets contain not less than 95.0 per cent and
Tests not more than 105.0 per cent of the stated amount of
thiabendazole, C10H7N3S. The tablets may contain permitted
Appearance of solution. Add 5 ml of methanol to 0.5 g in a colours.
flask fitted with a ground-glass stopper, stir for 5 minutes,
with a magnetic stirrer and filter through a sintered-glass filter Identification
(1.6 µm to 4 µm). The solution is not more intensely coloured
A. When examined in the range 230 nm to 360 nm (2.4.7), the
than reference solution BS6 (2.4.1).
final solution obtained in the Assay shows an absorption
Related substances. Determine by thin-layer chromatography maximum at about 302 nm.
(2.4.17), coating the plate with silica gel HF254. B. Dissolve a quantity of the powdered tablets containing
Mobile phase. A mixture of 62.5 volumes of toluene, 30 mg of Thiabendazole in 5 ml of 0.1 M hydrochloric acid,
25 volumes of glacial acetic acid, 10 volumes of acetone and add 3 mg of 4-phenylenediamine dihydrochloride and shake
2.5 volumes of water. until dissolved. Add 0.1 g of zinc powder, mix, allow to stand
for 2 minutes and add 10 ml of ferric ammonium sulphate
Test solution (a). Dissolve 0.1 g of the substance under
solution; a deep bluish violet colour is produced.
examination in 10 ml of methanol.
Test solution (b). Dilute 2 ml of test solution (a) to 20 ml with Tests
methanol.
Disintegration. The test does not apply.
Reference solution (a). Dilute 1 ml of test solution (b) to 10 ml
with methanol. Other tests. Comply with the tests stated under Tablets.

Reference solution (b). Dilute 1 ml of test solution (b) to 25 ml Assay. Weigh and powder 20 tablets. Weigh accurately a
with methanol. quantity of the powder containing about 0.1 g of
Thiabendazole, add 75 ml of 0.1 M hydrochloric acid, warm
Reference solution (c). Dissolve 20 mg of thiabendazole RS on a water-bath for 15 minutes, shaking occasionally, cool,
in 20 ml of methanol. dilute to 100.0 ml with 0.1 M hydrochloric acid and filter.
Apply to the plate 20 µl of each solution. After development, Dilute 5.0 ml of the filtrate to 1000.0 ml with 0.1 M hydrochloric
dry the plate in air and examine in ultraviolet light at 254 nm. acid and measure the absorbance of the resulting solution at
Any secondary spot in the chromatogram obtained with test the maximum at about 302 nm (2.4.7).
solution (a) is not more intense than the spot in the Calculate the content of C10H7N3S taking 1230 as the specific
chromatogram obtained with reference solution (a) and not absorbance at 302 nm.
more than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b) Storage. Store protected from light and moisture.
Heavy metals (2.3.13). 2.0 g complies with the limit test for Labelling. The label states that the tablets should be chewed
heavy metals, Method B (10 ppm). before swallowing.

1171
THIACETAZONE IP 2007

Thiacetazone Reference solution. Dissolve with the aid of heat 0.016 g of


4-acetamidobenzalazine RS in 150 ml of methanol, cool and
dilute to 200 ml with methanol and further dilute 5 ml of this
H
N NH2 solution to 50 ml with methanol.
O N
Apply to the plate 5 µl of each solution. After development,
S dry the plate in air, spray with 2 M nitric acid and within
H3 C N
H 2 minutes examine in ultraviolet light at 254 nm. Any secondary
spot in the chromatogram obtained with the test solution is
C10H12N4OS Mol. Wt. 236.3 not more intense than the spot in the chromatogram obtained
Thiacetazone is 4-acetamidobenzaldehyde with the reference solution.
thiosemicarbazone. Heavy metals (2.3.13). 2.0 g complies with the limit test for
heavy metals, Method B ( 10 ppm).
Thiacetazone contains not less than 98.0 per cent and not
more than 102.0 per cent of C10H12N4OS, calculated on the Sulphated ash (2.3.18). Not more than 0.2 per cent.
dried basis. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Description. Pale yellow crystals or a crystalline powder; on 1.0 g by drying in an oven at 105º.
almost odourless. Assay. Weigh accurately about 0.1 g, dissolve in 60 ml of
methanol by heating at 60º in a water-bath, add slowly 20 ml
Identification of hot methanolic silver nitrate solution, maintain the solution
Test A may be omitted if tests B and C are carried out. Tests B at 60º until the precipitate coagulates and leaves a clear
and C may be omitted if test A is carried out. supernatant liquid. Cool, filter through a sintered-glass crucible
(porosity No. 4), wash the residue with methanol until the
A. Determine by infrared absorption spectrophotometry (2.4.6). washings are free from silver nitrate and dry to constant weight
Compare the spectrum with that obtained with thiacetazone at 105º.
RS or with the reference spectrum of thiacetazone.
1 g of residue is equivalent to 0.4606 g of C10H12N4OS.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Storage. Store protected from light and moisture.
0.0003 per cent w/v solution in ethanol (95 per cent) shows
an absorption maximum at about 328 nm absorbance at about
328 nm, about 0.55.
C. Boil 10 mg with 5 ml of 1 M hydrochloric acid for 3 minutes,
Thiacetazone And Isoniazid Tablets
cool and add sufficient water to produce 200 ml. Mix 5 ml of Thiacetazone and Isoniazid Tablets contain one part by weight
this solution with 0.25 ml of sodium nitrite solution and add of Thiacetazone and two parts by weight of Isoniazid.
the mixture to 0.5 ml of 2-naphthol solution; a red colour is
Thiacetazone and Isoniazid Tablets contain not less than
produced.
92.5 per cent and not more than 107.5 per cent of the stated
Tests amounts of thiacetazone, C10H12N4OS, and isoniazid, C6H7N3O2.

Thiosemicarbazide. To 2.0 g, finely powdered, add sufficient Identification


water to produce 50 ml, shake, allow to stand for 1 hour with
A. Extract a quantity of the powdered tablets containing about
occasional shaking and filter, discarding the first few ml of the
30 mg of Thiacetazone with 70 ml of ethanol (95 per cent)
filtrate. Acidify 25 ml of the clear filtrate with dilute sulphuric
with the aid of heat for 15 minutes with occasional shaking.
acid, add 0.1 ml of o-phenanthroline-ferrous complex solution
Cool, dilute to 100 ml with ethanol (95 per cent) and filter.
and titrate with 0.1 M ceric ammonium sulphate to a blue
Dilute 1 ml to 100 ml with ethanol (95 per cent). The solution
end-point which persists for 1 minute; not more than 0.8 ml is
complies with the following test.
required.
When examined in the range 230 nm to 360 nm (2.4.7), a
4-acetamidobenzalazine. Determine by thin-layer
0.0005 per cent w/v solution in 0.1 M hydrochloric acid shows
chromatography (2.4.17), coating the plate with silica gel
absorption maxima at about 243 nm and 302 nm; ratio of the
GF254.
absorbance at the maximum at about 302 nm to that at about
Mobile phase. Ethyl acetate. 243 nm, 1.8 to 2.1.
Test solution. Dissolve 0.4 g of the substance under B. Shake a quantity of the powdered tablets containing 1 mg
examination in 100 ml of methanol. of Isoniazid with 50 ml of ethanol (95 per cent) and filter. To

1172
IP 2007 THIAMINE HYDROCHLORIDE

5 ml of the filtrate add 0.1 g of borax and 5 ml of a 5 per cent Thiamine Hydrochloride contains not less than 98.5 per cent
w/v solution of 1-chloro-2,4-dinitrobenzene in ethanol and not more than 101.5 per cent of C12H17ClN4OS, HCl,
(95 per cent), evaporate to dryness on a water-bath and calculated on the dried basis.
continue heating for a further 10 minutes. To the residue add Description. A white or almost white, crystalline powder or
10 ml of methanol and mix; a reddish purple colour is produced. small colourless crystals; odour, slight and characteristic.
Tests Identification
Disintegration (2.5.1). 30 minutes. Test A may be omitted if tests B and C are carried out. Test B
Other tests. Comply with the tests stated under Tablets. may be omitted if tests A and C are carried out.
Assay. For thiacetazone — Weigh and finely powder A. Determine by infrared absorption spectrophotometry
20 tablets. Weigh accurately a quantity of the powder (2.4.6). Compare the spectrum with that obtained with thiamine
containing about 30 mg of Thiacetazone, add 70 ml of ethanol hydrochloride RS or with the reference spectrum of thiamine
(95 per cent) and heat on a water-bath for 15 minutes with hydrochloride.
intermittent shaking. Cool, add sufficient ethanol (95 per cent) B. Dissolve about 20 mg in 10 ml of water, add 1 ml of 2 M
to produce 100.0 ml and filter. To 1.0 ml of the filtrate add acetic acid and 1.6 ml of 1 M sodium hydroxide, heat on a
sufficient ethanol (95 per cent) to produce 100.0 ml and water-bath for 30 minutes and allow to cool. Add 5 ml of 2 M
measure the absorbance of the resulting solution at the sodium hydroxide, 10 ml of potassium ferricyanide solution
maximum at about 328 nm (2.4.7), using ethanol (95 per cent) and 10 ml of 1-butanol and shake vigorously for 2 minutes.
as the blank. The upper layer exhibits an intense light blue fluorescence,
Calculate the content of C10H12N4OS from the absorbance particularly in ultraviolet light at 365 nm. Repeat the test but
obtained by repeating the operation using thiacetazone RS in adding 0.9 ml of 1 M sodium hydroxide and 0.2 g of sodium
place of the tablets under examination. sulphite in place of the 1.6 ml of 1 M sodium hydroxide;
practically no fluorescence is produced.
For isoniazid — Weigh accurately a quantity of the powdered
tablets containing about 0.2 g of Isoniazid, dissolve as C. Gives reaction A of chlorides (2.3.1).
completely as possible in 100 ml of water and filter. Wash the
residue with water, combine the filtrate and washings and
Tests
dilute to 250.0 ml with water. To 50.0 ml of the resulting solution Appearance of solution. A 5.0 per cent w/v solution is clear
add 50 ml of water, 20 ml of hydrochloric acid and 0.2 g of (2.4.1), and not more intensely coloured than reference solution
potassium bromide. Titrate with 0.0167 M potassium bromate, YS7 or GYS6 (2.4.1).
determining the end-point potentiometrically (2.4.25).
pH (2.4.24). 2.7 to 3.3, determined in a 2.5 per cent w/v solution.
1 ml of 0.0167 M potassium bromate is equivalent to 0.003429
Heavy metals (2.3.13). 1.0 g complies with the limit test for
g of C6H7N3O2.
heavy metals, Method A (20 ppm).
Storage. Store protected from light and moisture. Nitrates. To 2 ml of a 2.0 per cent w/v solution add 2 ml of
sulphuric acid, cool and superimpose 2 ml of ferrous sulphate
solution; no brown ring is produced at the junction of the two
layers.
Thiamine Hydrochloride Sulphates (2.3.17). 0.5 g complies with the limit test for
Aneurine Hydrochloride; Vitamin B1 sulphates (300 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
NH2 CH3 Loss on drying (2.4.19). Not more than 5.0 per cent, determined
OH Cl, HCl on 1.0 g by drying in an oven at 105º.
N N
S Assay. Weigh accurately about 0.15 g, dissolve in 5 ml of
H3C N anhydrous formic acid, add 65 ml of anhydrous glacial acetic
acid and 10 ml of mercuric acetate solution, with stirring.
C12H17ClN4OS, HCl Mol. Wt. 337.3 Titrate with 0.1 M perchloric acid, determining the end-point
Thiamine Hydrochloride is 3-[(4-amino-2-methylpyrimidin-5- potentiometrically (2.4.25). Carry out a blank titration.
yl)methyl]-5-(2-hydroxyethyl)-4-methylthiazolium chloride 1 ml of 0.1 M perchloric acid is equivalent to 0.01686 g of
hydrochloride hydrate. C12H17ClN4OS, HCl.

1173
THIAMINE INJECTION IP 2007

Storage. Store protected from light and moisture, non-metallic – mobile phase: a solution prepared by dissolving 1 g of
containers. sodium heptanesulphonate in a mixture of 180 ml of
methanol and 10 ml of triethylamine, diluting to 1000 ml
with water and adjusting the pH to 3.2 with phosphoric
acid,
– flow rate. 2 ml per minute,
Thiamine Injection – spectrophotometer set at 244 nm,
Thiamine Hydrochloride Injection; Aneurine – a 20 µl loop injector.
Hydrochloride Injection; Vitamin B1 Injection Calculate the content of C12H17ClN4OS.
Thiamine Injection is a sterile solution of Thiamine 1 mg of C12H17N5O4S is equivalent to 1.030 mg of C12H17ClN4OS,
Hydrochloride in Water for Injection. HCl.
Thiamine Injection contains not less than 95.0 per cent and Storage. Store protected from light.
not more than 110.0 per cent of the stated amount of thiamine
hydrochloride, C12H17ClN4OS, HCl.

Identification Thiamine Tablets


A. To a volume containing 20 mg of Thiamine Hydrochloride Thiamine Hydrochloride Tablets; Aneurine Hydrochloride
in 10 ml of water, add 1 ml of 2 M acetic acid and 1.6 ml of 1 M Tablets; Vitamin B1 Tablets
sodium hydroxide, heat on a water-bath for 30 minutes and
Thiamine Tablets contain not less than 92.5 per cent and not
allow to cool. Add 5 ml of 2 M sodium hydroxide, 10 ml of
more than 110.0 per cent of the stated amount of thiamine
potassium ferricyanide solution and 10 ml of 1-butanol and
hydrochloride, C12H17ClN4OS, HCl.
shake vigorously for 2 minutes. The upper layer exhibits an
intense light blue fluorescence, particularly in ultraviolet light Identification
at 365 nm. Repeat the test but adding 0.9 ml of 1 M sodium
hydroxide and 0.2 g of sodium sulphite in place of the 1.6 ml A. Dissolve a quantity of the powdered tablets containing
of 1 M sodium hydroxide; practically no fluorescence is 20 mg of Thiamine Hydrochloride as completely as possible in
produced. 10 ml of water and 2 ml of 1 M acetic acid and filter. Add 5 ml
of 2 M sodium hydroxide, 10 ml of potassium ferricyanide
B. To a mixture of 0.1 ml of nitrobenzene and 0.2 ml of sulphuric
solution and 10 ml of 1-butanol and shake vigorously for
acid add a volume containing 5 mg of Thiamine Hydrochloride.
2 minutes. The upper layer exhibits an intense light blue
Allow to stand for 10 minutes, cool in ice and add slowly with
fluorescence, particularly in ultraviolet light at 365 nm. Repeat
stirring 5 ml of water followed by 5 ml of 10 M sodium
the test but adding 0.9 ml of 1 M sodium hydroxide and 0.2 g
hydroxide. Add 5 ml of acetone and allow to stand; no violet
of sodium sulphite in place of the 1.6 ml of 1 M sodium
colour is produced in the upper layer.
hydroxide; practically no fluorescence is produced.
Tests B. To a mixture of 0.1 ml of nitrobenzene and 0.2 ml of sulphuric
acid add the powdered tablets containing 5 mg of Thiamine
pH (2.4.24). 2.5 to 4.5. Hydrochloride. Allow to stand for 10 minutes, cool in ice and
Other tests. Complies with the tests stated under Parenteral add slowly with stirring 5 ml of water followed by 5 ml of 10 M
Preparations (Injections). sodium hydroxide. Add 5 ml of acetone and allow to stand;
no violet colour is produced in the upper layer.
Assay. Determine by liquid chromatography (2.4.14).
C. The powdered tablets give the reactions of chlorides (2.3.1).
Test solution. Dilute a volume of the injection containing about
0.1 g of Thiamine Hydrochloride to 100.0 ml with 0.1 M Tests
hydrochloric acid and further dilute 5.0 ml to 100.0 ml with
water. Uniformity of content. (For tablets containing 10 mg or less)
— Comply with the test stated under Tablets.
Reference solution. A 0.005 per cent w/v solution of thiamine
mononitrate RS in 0.005 M hydrochloric acid. Finely crush one tablet, add 20 ml of ethanol (95 per cent),
stir the mixture for 30 minutes and centrifuge. Repeat the
Chromatographic system extraction with three further quantities, each of 15 ml, of
– a stainless steel column 10 cm x 4.6 mm, packed with ethanol (95 per cent). Combine the extracts, add sufficient
octadecylsilane bonded to porous silica (5 µm), ethanol (95 per cent) to produce 100.0 ml and mix. Dilute a

1174
IP 2007 THIAMINE MONONITRATE

suitable volume of the resulting solution containing 1 mg of Thiamine Mononitrate is 3-[(4-amino-2-methylpyrimidin-5-


Thiamine Hydrochloride with sufficient ethanol (95 per cent) yl)methyl]-5-(2-hydroxyethyl)-4-methylthiazolium nitrate.
to produce 100.0 ml. Measure the absorbance of the resulting Thiamine Mononitrate contains not less than 98.0 per cent
solution at the maximum at about 233 nm (2.4.7). and not more than 102.0 per cent of C12H17N5O4S, calculated
Calculate the content of C12H17ClN4OS, HCl in the tablet taking on the dried basis.
380 as the specific absorbance at 233 nm.
Description. A white or almost white, crystalline powder or
Other tests. Comply with the tests stated under Tablets. small, colourless crystals.
Assay. Determine by liquid chromatography (2.4.14). Identification
Test solution. For tablets containing less than 10 mg of Test A may be omitted if tests B and C are carried out. Test B
Thiamine Hydrochloride, add 5 ml of 0.1 M hydrochloric acid may be omitted if tests A and C are carried out.
and 50 ml of water to a quantity of the powdered tablets
containing 6 mg of Thiamine Hydrochloride, shake for A. Determine by infrared absorption spectrophotometry (2.4.6).
20 minutes, dilute to 100.0 ml with water and filter. For tablets Compare the spectrum with that obtained with thiamine
containing 10 mg or more of Thiamine Hydrochloride, add mononitrate RS or with the reference spectrum of thiamine
50 ml of 0.1 M hydrochloric acid and 500 ml of water to a mononitrate.
quantity of the powdered tablets containing 60 mg of Thiamine B. Dissolve about 20 mg in 10 ml of water, add 1 ml of 2 M
Hydrochloride, shake for 20 minutes, dilute to 1000.0 ml with acetic acid and 1.6 ml of 1 M sodium hydroxide, heat on a
water and filter. water-bath for 30 minutes and allow to cool. Add 5 ml of 2 M
Reference solution. A 0.006 per cent w/v solution of thiamine sodium hydroxide, 10 ml of potassium ferricyanide solution
mononitrate RS in 0.005 M hydrochloric acid. and 10 ml of 1-butanol and shake vigorously for 2 minutes.
The upper layer exhibits an intense light blue fluorescence,
Chromatographic system particularly in ultraviolet light at 365 nm. Repeat the test but
– a stainless steel column 10 cm x 4.6 mm, packed with adding 0.9 ml of 1 M sodium hydroxide and 0.2 g of sodium
octadecylsilane bonded to porous silica (5 µm), sulphite in place of the 1.6 ml of 1 M sodium hydroxide;
– mobile phase: a solution prepared by dissolving 1 g of practically no fluorescence is produced.
sodium heptanesulphonate in a mixture of 180 ml of
methanol and 10 ml of triethylamine, diluting to 1000 ml C. Gives reaction A of nitrates (2.3.1).
with water and adjusting the pH to 3.2 with phosphoric
Tests
acid,
– flow rate. 2 ml per minute, Appearance of solution. A 2.0 per cent w/v solution in carbon
– spectrophotometer set at 244 nm, dioxide-free water is clear (2.4.1), and not more intensely
– a 20 µl loop injector. coloured than reference solution YS7 (2.4.1).
Calculate the content of C12H17ClN4OS in the tablets. pH (2.4.24). 6.8 to 7.6, determined in a 2.0 per cent w/v solution.
1 mg of C12H17N5O4S is equivalent to 1.030 mg of C12H17ClN4OS, Heavy metals (2.3.13). 1.0 g complies with limit test for heavy
HCl. metals, Method A (20 ppm).
Storage. Store protected from light and moisture in non-metallic Chlorides (2.3.12). 1.0 g complies with the limit test for chlorides
containers. (250 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
Thiamine Mononitrate on 1.0 g by drying in an oven at 105º.
Thiamine Nitrate Assay. Weigh accurately about 0.15 g, dissolve in 5 ml of
anhydrous formic acid, add 70 ml of acetic anhydride. Titrate
NH2 CH3 with 0.1 M perchloric acid, determining the end-point
potentiometrically (2.4.25). Carry out a blank titration.
OH . NO
N N 3
1 ml of 0.1 M perchloric acid is equivalent to 0.01637 g of
S C12H17N5O4S.
H3 C N
Storage. Store protected from light and moisture, non-metallic
C12H17N5O4S Mol. Wt. 327.4 containers.

1175
THIOMERSAL IP 2007

Thiomersal expression 0.7019 (Ab - Aa - Ae)/(Aa + Ad - Ab - Ac), where


the figure 0.7019 is a constant obtained from the formula
Thimerosal
atomic weight of mercury concentrat ion of HgCl 2
×
SHgCH2CH3 molecular weight of HgCl 2 100
COONa
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying over phosphorus pentoxide at a pressure
not exceeding 0.7 kPa for 24 hours.
C9H9HgNaO2S Mol. Wt. 404.8 Assay. Weigh accurately about 0.5 g, transfer to a 100-ml long-
Thiomerosal is the sodium salt of [(2-carboxyphenyl)thio] necked flask, add 5 ml of sulphuric acid and heat gently until
ethylmercury. charring occurs; continue to heat and add hydrogen peroxide
solution (100 vol) dropwise, until the mixture is colourless.
Thiomersal contains not less than 97.0 per cent and not more
Dilute with water, evaporate until slight fuming occurs, dilute
than 101.0 per cent of C9H9HgNaO2S, calculated on the dried
to 10 ml with water, cool and titrate with 0.1 M ammonium
basis.
thiocyanate using ferric ammonium sulphate solution as
Description. A light cream, crystalline powder; odour, slight indicator.
and characteristic. 1 ml of 0.1 M ammonium thiocyanate is equivalent to 0.02024
Identification g of C9H9HgNaO2S.
Storage. Store protected from light and moisture.
A. Dissolve 0.1 g in 10 ml of water and add 2 ml of silver
nitrate solution; a pale yellow precipitate is produced.
B. Dissolve 0.5 g in 10 ml of water and add 2 ml of 2 M
hydrochloric acid; a white precipitate is produced which, Thiopentone Sodium
after washing with water and drying over phosphorus Thiopental Sodium
pentoxide at a pressure not exceeding 0.7 kPa, melts at about
110º (2.4.21).
H
Tests O N SNa

pH (2.4.24). 6.0 to 8.0, determined in a 1.0 per cent w/v solution. H3C N + Na2CO3
Ether-soluble matter. Shake 0.5 g with 20 ml of ether for O
H3 C CH3
10 minutes, filter and evaporate to dryness. The residue, after
drying over phosphorus pentoxide at a pressure not exceeding
0.7 kPa for 24 hours, weighs not more than 3 mg. C11H17N2NaO2S Mol. Wt. 264.3
Inorganic mercury compounds. Not more than 0.7 per cent, Thiopentone Sodium is a mixture of sodium (RS)-5-ethyl-5-
determined by the following method. (1-methylbutyl)-2-thiobarbiturate and anhydrous sodium
Protect the solutions from light throughout the procedure. carbonate.
Thiopentone Sodium contains not less than 84.0 per cent and
Label five 10-ml volumetric flasks A, B, C, D and E. Place 5 ml
not more than 87.0 per cent of C11H18N2O2S and not less than
of a 0.1 per cent w/v solution of the substance under
10.2 per cent and not more than 11.2 per cent of Na, both
examination in each of flasks A, B, C and D. To each of flasks
calculated on the dried basis.
C and D add 0.5 ml of a 0.0095 per cent w/v solution of mercuric
chloride. Add sufficient water to flasks A and C to produce Description. A yellowish white powder; odour, faintly
10.0 ml and add sufficient of a freshly prepared 33.2 per cent resembling garlic; hygroscopic.
w/v solution of potassium iodide to flasks B and D to produce
10.0 ml. Place 5 ml of the potassium iodide solution in flask E Identification
and add sufficient water to produce 10.0 ml. Measure the Test A may be omitted if tests B, C, D and E are carried out.
absorbances of each of the solutions (Aa, Ab, Ac, Ad, Ae) at Tests B and D may be omitted if tests A, C and E are carried
about 323 nm (2.4.7), using water as the blank. out.
Calculate the content of inorganic mercury compounds, A. Acidify 10 ml of a 10 per cent w/v solution in carbon dioxide-
expressed as Hg, in the substance under examination from the free water with 2 M hydrochloric acid; the solution

1176
IP 2007 THIOPENTONE INJECTION

effervesces. Shake the solution with 20 ml of ether, separate methoxide. Titrate immediately with 0.1 M lithium methoxide,
the ether layer, wash with 10 ml of water and dry over using 1 drop of a 0.2 per cent w/v solution of thymol blue in
anhydrous sodium sulphate. Filter, evaporate the filtrate to methanol as indicator, until a blue colour is obtained. Protect
dryness and dry the residue at 105º. the solution from absorption of carbon dioxide during the
On the residue, determine by infrared absorption titration.
spectrophotometry (2.4.6). Compare the spectrum with that 1 ml of 0.1 M lithium methoxide is equivalent to 0.02423 g of
obtained with thiopentone RS or with the reference spectrum C11H18N2O2S.
of thiopentone. For sodium — Weigh accurately about 0.4 g, dissolve in 30 ml
B. Complies with the test for identification of barbiturates of water, add 1 drop of methyl red solution and titrate with
(2.3.2), but using the following solutions. 0.05 M sulphuric acid until the yellow colour changes to red.
Boil gently for 2 minutes, cool and, if necessary, continue the
Test solution. A 0.1 per cent w/v solution of the substance titration with 0.05 M sulphuric acid until the red colour is
under examination in water. restored.
Reference solution. Dissolve 85 mg of thiopentone RS in 1 ml of 0.05 M sulphuric acid is equivalent to 0.002299 g of
10 ml of 2 M sodium hydroxide and dilute to 100 ml with water. Na.
C. Acidify 5 ml of a 5 per cent w/v solution with dilute acetic Storage. Store protected from light and moisture.
acid and filter. Wash the precipitate with water, recrystallise
from water and dry at 70º; the crystals melt at about 160º
(2.4.21).
D. Dissolve 1 mg of the crystals obtained in test A in 1 ml of
Thiopentone Injection
0.1 M sodium hydroxide. Add about 1 mg of sodium Thiopentone Sodium Injection; Thiopental Injection
nitroprusside and, after 15 minutes, 1 ml of dilute hydrochloric
Thiopentone Injection is a sterile material consisting of
acid; a reddish violet colour is produced.
Thiopentone Sodium with or without auxiliary agents. It is
E. Gives the reactions of sodium salts (2.3.1). filled in a sealed container.

Tests The injection is constituted by dissolving the contents of the


sealed container in the requisite amount of sterile Water for
Appearance of solution. A 10.0 per cent w/v solution in carbon Injections, immediately before use.
dioxide-free water (solution A) is clear (2.4.1), and not more
The constituted solution complies with the requirements for
intensely coloured than reference solution GYS3 (2.4.1).
Clarity of solution and Particulate matter stated under
Related substances (2.3.4). Complies with the test, but using Parenteral Preparations (Injections).
water as the solvent for the test solution and the reference
Storage. The constituted solution should be used immediately
solution. After development, examine the plate in ultraviolet
after preparation but, in any case, within the period
light at 254 nm but do not spray it with the diphenylcarbazone-
recommended by the manufacturer.
mercury reagent.
Thiopentone Injection contains thiopentone, C11H18N2O2S that
Chlorides (2.3.12). To 10 ml of solution A add 30 ml of water is not less than 77.0 per cent and not more than 92.0 per cent
and 10 ml of 2 M nitric acid. Shake successively with three and sodium, Na that is not less than 9.4 per cent and not more
quantities, each of 25 ml, of ether, discard the ether layers and than 11.8 per cent of the stated amount of thiopentone sodium.
heat the aqueous solution on water-bath to remove any
residual ether. 30 ml of the aqueous layer complies with the The contents of the sealed container comply with the
limit test for chlorides (330 ppm). requirements stated under Parenteral Preparations
(Powders for Injection) and with the following requirements.
Loss on drying (2.4.19). Not more than 2.5 per cent, determined
on 0.5 g by drying in an oven at 100º at a pressure of 1.5 to 2.5 Description. A yellowish white powder; hygroscopic.
kPa for 4 hours.
Identification
Assay. For thiopentone — Weigh accurately about 0.15 g,
dissolve in 5 ml of water, add 2 ml of 1 M sulphuric acid and Test A may be omitted if tests B, C, D and E are carried out.
extract with four quantities, each of 10 ml, of chloroform. Filter Tests B and D may be omitted if tests A, C and E are carried
the combined chloroform extracts, evaporate the filtrate to out.
dryness on a water-bath and dissolve the residue in 30 ml of A. Acidify 10 ml of a 10 per cent w/v solution in carbon dioxide-
dimethylformamide, previously neutralised with 0.1 M lithium free water with 2 M hydrochloric acid; the solution

1177
THIOTEPA IP 2007

effervesces. Shake the solution with 20 ml of ether, separate 1 ml of 0.1 M lithium methoxide is equivalent to 0.02423 g of
the ether layer, wash with 10 ml of water and dry over C11H18N2O2S.
anhydrous sodium sulphate. Filter, evaporate the filtrate to For sodium — Mixed the contents of 10 containers and weigh
dryness and dry the residue at 105º. accurately about 0.4 g of the contents, dissolve in 30 ml of
On the residue, determine by infrared absorption water, add 1 drop of methyl red solution and titrate with
spectrophotometry (2.4.6). Compare the spectrum with that 0.05 M sulphuric acid until the yellow colour changes to red.
obtained with thiopentone RS or with the reference spectrum Boil gently for 2 minutes, cool and, if necessary, continue the
of thiopentone. titration with 0.05 M sulphuric acid until the red colour is
restored
B. Complies with the test for identification of barbiturates
(2.3.2), but using the following solutions. 1 ml of 0.05 M sulphuric acid is equivalent to 0.002299 g of
Na.
Test solution. 0.1 per cent w/v solution of the substance
under examination in water. Storage. Store in single dose containers.
Reference solution. Dissolve 85 mg of thiopentone RS in Labelling. The label states the amount of active ingredient in
10 ml of 2 M sodium hydroxide and dilute to 100 ml with water. terms of Thiopentone Sodium.

C. Acidify 5 ml of a 5 per cent w/v solution with dilute acetic


acid and filter. Wash the precipitate with water, recrystallise
from water and dry at 70º; the crystals melt at about 160º
Thiotepa
(2.4.21). S
D. Dissolve 1 mg of the crystals obtained in test A in 1 ml of N P N
0.1 M sodium hydroxide. Add about 1 mg of sodium N
nitroprusside and, after 15 minutes, 1 ml of dilute hydrochloric
acid; a reddish violet colour is produced. C6H12N3PS Mol. Wt. 189.2
E. Gives the reactions of sodium salts (2.3.1). Thiotepa is tris(1-aziridinyl)phosphine sulphide.
Tests Thiotepa contains not less than 97.0 per cent and not more
than 102.0 per cent of C6H12N3PS, calculated on the anhydrous
Appearance of solution. A 10.0 per cent w/v solution in carbon basis.
dioxide-free water (solution A) is clear (2.4.1), and not more
intensely coloured than reference solution GYS3 (2.4.1). Description. Fine, white, crystalline flakes; odourless or almost
odourless.
Related substances (2.3.4). Complies with the test, but using
water as the solvent for the test solution and the reference Identification
solution. After development, examine the plate in ultraviolet A. Determine by infrared absorption spectrophotometry (2.4.6).
light at 254 nm but do not spray it with the diphenylcarbazone- Compare the spectrum with that obtained with thiotepa RS or
mercury reagent. with the reference spectrum of thiotepa.
Loss on drying (2.4.19). Not more than 2.5 per cent, determined B. Determine on 20 mg by the oxygen-flask method (2.3.34),
on 0.5 g by drying in an oven at 100º at a pressure of 1.5 to 2.5 using 5 ml of 1.25 M sodium hydroxide as the absorbing
kPa for 4 hours. liquid. When the process is complete, dilute to 25 ml with
Assay. For thiopentone — Mixed the contents of 10 containers water (solution A). To 5 ml of solution A add 0.1 ml of hydrogen
and weigh accurately about 0.15 g, of the contents, dissolve peroxide solution (100 vol) and 1 ml of 1 M hydrochloric
in 5 ml of water, add 2 ml of 1 M sulphuric acid and extract acid, mix and add 0.05 ml of barium chloride solution; a
with four quantities, each of 10 ml, of chloroform. Filter the turbidity is produced.
combined chloroform extracts, evaporate the filtrate to dryness C. To 2 ml of solution A add 40 ml of water and 4 ml of ammonium
on a water-bath and dissolve the residue in 30 ml of molybdate-sulphuric acid solution, mix, add 0.1 g of
dimethylformamide, previously neutralised with 0.1 M lithium L-ascorbic acid and boil for 1 minute; a blue colour is
methoxide. Titrate immediately with 0.1 M lithium methoxide, produced.
using 1 drop of a 0.2 per cent w/v solution of thymol blue in
methanol as indicator, until a blue colour is obtained. Protect Tests
the solution from absorption of carbon dioxide during the Appearance of solution. A 2.0 per cent w/v solution is clear
titration. (2.4.1).

1178
IP 2007 THYMOL

Water (2.3.43). Not more than 2.0 per cent, determined on B. To 2 ml of solution A add 40 ml of water and 4 ml of ammonium
1.2 g. molybdate-sulphuric acid solution, mix, add 0.1 g of
Assay. Weigh accurately about 0.2 g into a stoppered flask, L-ascorbic acid and boil for 1 minute; a blue colour is
add 50 ml of a 20 per cent w/v solution of sodium thiosulphate produced.
and titrate immediately with 0.1 M hydrochloric acid, using Tests
methyl orange solution as indicator, until a faint red colour
persists for 10 seconds. Stopper the flask, allow to stand for Appearance of solution. Dissolve a quantity containing 15 mg
30 minutes and titrate with 0.1 M sodium hydroxide using of Thiotepa in 4 ml of water; the solution is clear (2.4.1).
phenolphthalein solution as indicator. Subtract the volume
pH (2.4.24). 5.5 to 7.5, determined in a 1.0 per cent w/v solution
of 0.1 M sodium hydroxide used from the volume of 0.1 M
in carbon dioxide-free water.
hydrochloric acid used. Repeat the operation without the
substance under examination. The difference between the Bacterial endotoxins (2.2.3). Not more than 6.25 Endotoxin
titrations represents the amount of hydrochloric acid required. Units per mg of Thiotepa.
1 ml of 0.1 M hydrochloric acid is equivalent to 0.006307 g of Assay. Determine by liquid chromatography (2.4.14).
C6H12N3PS. Test solution. Dilute a suitable quantity of the injection
Storage. Store protected from light and moisture. At higher containing 0.15 g of Thiotepa in 100 ml of water.
temperatures it tends to polymerise with loss of activity. Reference solution. A 0.15 per cent w/v solution of thiotepa
RS in water.
Chromatographic system
– a stainless steel column 20 cm x 4.6 mm, packed with
Thiotepa Injection octadecylsilane bonded to porous silica (5 µm),
Thiotepa Injection is a sterile material consisting of Thiotepa – mobile phase: a mixture of 70 volumes of water and 30
with or without auxiliary agents. It is filled in a sealed container. volumes of acetonitrile,
– flow rate. 1 ml per minute,
The injection is constituted by dissolving the contents of the
– spectrophotometer set at 215 nm,
sealed container in the requisite amount of sterile Water for
– a 20 µl loop injector.
Injections, immediately before use.
Calculate the content of C6H12N3PS in the injection.
The constituted solution complies with the requirements for
Clarity of solution and Particulate matter stated under Storage. Store protected from light. If solid matter separates
Parenteral Preparations (Injections). from the constituted injection, the solution should not be
used.
Storage. The constituted solution should be used immediately
after preparation but, in any case, within the period
recommended by the manufacturer.
Thiotepa Injection contains not less than 90.0 per cent and
Thymol
not more than 110.0 per cent of the stated amount of thiotepa, OH CH3
C6H12N3PS.
The contents of the sealed container comply with the CH3
requirements stated under Parenteral Preparations
H3 C
(Powders for Injection) and with the following requirements.
Description. A white powder. C10H14O Mol. Wt. 150.2
Thymol is 2-isopropyl-5-methylphenol.
Identification
Thymol contains not less than 99.0 per cent and not more
A. Determine on 20 mg by the oxygen-flask method (2.3.34), than 101.0 per cent of C10H14O.
using 5 ml of 1.25 M sodium hydroxide as the absorbing
Description. Colourless crystals; odour, characteristic.
liquid. When the process is complete, dilute to 25 ml with
water (solution A). To 5 ml of solution A add 0.1 ml of hydrogen Identification
peroxide solution (100 vol) and 1 ml of 1 M hydrochloric
acid, mix and add 0.05 ml of barium chloride solution; a Test A may be omitted if tests B, C and D are carried out. Tests
turbidity is produced. B, C and D may be omitted if test A is carried out.

1179
THYROXINE SODIUM IP 2007

A. Determine by infrared absorption spectrophotometry (2.4.6). area of the principal peak in the chromatogram obtained with
Compare the spectrum with that obtained with thymol RS or reference solution (a). Ignore any peak with an area less than
with the reference spectrum of thymol. that of the principal peak in the chromatogram obtained with
B. Dissolve 0.2 g with heating in 2 ml of 2 M sodium hydroxide, reference solution (c).
add 0.2 ml of chloroform and heat on a water-bath; a violet Residue on evaporation. Evaporate 2.0 g on a water-bath and
colour develops. heat at 105º for 1 hour. The residue weighs not more than
1.0 mg (0.05 per cent).
C. Dissolve about 2 mg in 1 ml of anhydrous acetic acid, add
0.15 ml of sulphuric acid and 0.05 ml of nitric acid; a bluish Assay. Weigh accurately about 0.1 g, transfer to an iodine
green colour develops. flask and dissolve in 25 ml of 1 M sodium hydroxide. Add 20
ml of hot dilute hydrochloric acid and immediately titrate
D. Melting range (2.4.21). 48º to 52º.
with 0.05 M bromine to within 1 to 2 ml of the calculated end-
Tests point. Warm the solution to about 75º, add 0.1 ml of methyl
orange solution and shake vigorously. If the solution is red,
Appearance of solution. Dissolve 1.0 g in 10 ml of 2 M sodium continue the titration, dropwise and with shaking until the red
hydroxide. The solution is not more opalescent than colour is discharged. Repeat the alternate addition of 0.05 M
opalescence standard OS4 (2.4.1), and not more intensely bromine and methyl orange solution until the red colour is
coloured than reference solution RS6 (2.4.1). discharged after the addition of the methyl orange solution.
Acidity. To 1.0 g in a glass-stoppered flask add 20 ml of water, 1 ml of 0.05 M bromine is equivalent to 0.003755 g of C10H14O.
boil until dissolution is complete, cool, stopper the flask and Storage. Store protected from light and moisture.
shake vigorously for 1 minute. Add a few crystals of the
substance under examination to induce crystallisation, shake
vigorously for 1 minute and filter. To 5 ml of the filtrate add
0.05 ml of methyl red solution and 0.05 ml of 0.01 M sodium
hydroxide; the solution is yellow. Thyroxine Sodium
Related substances. Determine by gas chromatography Levothyroxine Sodium; L-Thyroxine Sodium
(2.4.13).
Test solution. Dissolve 0.1 g in sufficient ethanol (95 per I COONa
cent) to produce 10 ml.
H NH2
Reference solution (a). Dilute 1 ml of the test solution to O , xH2O
100 ml with ethanol (95 per cent). I
Reference solution (b). Dilute 1 ml of reference solution (a) to
10 ml with ethanol (95 per cent). I I
Reference solution (c). Dilute 5 ml of reference solution (b) to OH
10 ml with ethanol (95 per cent).
Chromatographic system C15H10I4NNaO4,xH2O Mol. Wt. 798.9 (anhydrous)
– a glass column 4 m x 2 mm, packed with diatomaceous
Thyroxine Sodium is sodium O4-(4-hydroxy-3,5-
support (125 to 180 mesh) impregnated with a mixture
diiodophenyl)-3,5-diiodo-L-tyrosinate and contains a
suitable for the separation of free fatty acids (such as
variable quantity of water of crystallisation.
FFAP),
– temperature: Thyroxine Sodium contains not less than 97.0 per cent and
column 80º, not more than 101.0 per cent of C15H10I4NNaO4, calculated on
inlet port at 250º and detector at 300º, the dried basis.
– flow rate. 30 ml per minute of the carrier gas. Description. A white or slightly brownish yellow powder,
Inject separately 1 µl of each solution and, after 2 minutes, slightly coloured, crystalline powder.
increase the temperature of the column to 240º at a rate of 8º
per minute and maintain at this temperature for 15 minutes. Identification
In the chromatogram obtained with the test solution the sum A. When examined in the range 230 nm to 360 nm (2.4.7), a
of the areas of any secondary peaks is not greater than the 0.01 per cent w/v solution in 0.1 M sodium hydroxide shows

1180
IP 2007 THYROXINE TABLETS

an absorption maximum at about 325 nm; absorbance at about the chromatogram obtained with reference solution (a) shows
325 nm, 0.73 to 0.79. two clearly separated spots.
B. In the test for Liothyronine, the principal spot in the Loss on drying (2.4.19). 6.0 to 12.0 per cent, determined on
chromatogram obtained with the test solution corresponds to 0.5 g by drying in an oven at 105º.
that in the chromatogram obtained with reference solution
Assay. Determine by liquid chromatography (2.4.14).
(b).
Test solution. A 0.001 per cent w/v solution of the substance
C. To about 50 mg in a porcelain dish add a few drops of
under examination in 0.1 M methanolic sodium hydroxide.
sulphuric acid (96 per cent w/w); violet vapors are evolved.
Reference solution. A 0.001 per cent w/v solution of
D. To 20 mg add 2 ml of 1 M sulphuric acid. Heat on a water- levothyroxine sodium RS in 0.1 M methanolic sodium
bath followed by heating carefully over a naked flame, hydroxide.
increasing the temperature to about 600º. Continue ignition
until most of the black particles have disappeared. Dissolve Chromatographic system
the residue in 2 ml of water; the solution gives reaction A of – a stainless steel column 25 cm x 4.6 mm, packed with
sodium salts (2.3.1). silicagel consisting of porous spherical particles with
chemically bonded nitrile groups (5 µm),
Tests – mobile phase: a mixture of 65 volumes of water, 0.2
volume of orthophosphoric acid and
Appearance of solution. Dissolve 0.5 g in 23 ml of a gently 35 volumes of acetonitrile,
boiling mixture of 4 volumes of ethanol (95 per cent) and – flow rate. 1 ml per minute,
1 volume of 1 M hydrochloric acid. Cool and dilute to 25 ml – spectrophotometer set at 225 nm,
with the same mixture of solvents (solution A). The freshly – a 20 µl loop injector.
prepared solution is not more intensely coloured than reference
solution BYS3 (2.4.1). Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
Specific optical rotation (2.4.22).+16.0º to +20.0º, determined than 2.0 per cent.
in solution A.
Inject alternately the test solution and the reference solution.
Liothyronine. Determine by thin-layer chromatography
(2.4.17), coating the plate with a slurry of 30 g of silica gel H in Calculate the content of C15H10I4NNaO4.
60 ml of a 0.75 per cent w/v solution of soluble starch. Storage. Store protected from light and moisture.
Solvent mixture. 70 volumes of methanol and 5 volumes of
strong ammonia solution.
Mobile phase. A mixture of 55 volumes of ethyl acetate, Thyroxine Tablets
35 volumes of 2-propanol and 20 volumes of strong ammonia
solution. Thyroxine Sodium Tablets; Levothyroxine Tablets;
Levothyroxine Sodium Tablets; L-Thyroxine Sodium
Test solution. Dissolve 0.1 g of the substance under
Tablets
examination in 10 ml of the solvent mixture.
Thyroxine Tablets contain not less than 90.0 per cent and not
Reference solution (a). A solution containing 1.0 per cent
more than 110.0 per cent of the stated amount of anhydrous
w/v of the substance under examination and 0.01 per cent
thyroxine sodium, C15H10I4NNaO4.
w/v of the liothyronine RS in the solvent mixture.
Reference solution (b). A 1 per cent w/v solution of Identification
levothyroxine sodium RS in the solvent mixture.
A. To a quantity of the powdered tablets containing 500 µg of
Reference solution (c). A 0.01 per cent w/v solution of anhydrous thyroxine sodium add a mixture of 3 ml of ethanol
liothyronine RS in the solvent mixture. (50 per cent) and 0.2 ml of hydrochloric acid, boil gently for
30 seconds, cool, filter, add 0.1 ml of a 10 per cent w/v solution
Apply to the plate 5 µl of each solution. After development,
of sodium nitrite and boil; a yellow colour is produced. Cool
dry the plate in air, spray lightly with ferric chloride-
and make alkaline with 5 M ammonia; the solution becomes
ferricyanide-arsenite solution. Any spot corresponding to
orange.
liothyronine in the chromatogram obtained with the test
solution is not more intense than the spot in the chromatogram B. In the Assay, the principal peak in the chromatogram
obtained with reference solution (c). The test is not valid unless obtained with the test solution corresponds to the peak due

1181
TIMOLOL MALEATE IP 2007

to thyroxine sodium in the chromatogram obtained with the Timolol Maleate


reference solution.

Tests S
N N H3 C COOH
CH3
Uniformity of content. Comply with the test stated under ,
N O N CH3
Tablets. using the following solutions. H COOH
O OH
Test solution. Place one tablet in a 25-ml volumetric flask, add
10 ml of 0.1 M methanolic sodium hydroxide and shake for
C13H24N4O3S,C4H4O4 Mol. Wt. 432.5
about 30 minutes. Dilute to volume with 0.1 M methanolic
sodium hydroxide. Dilute further, if necessary, with 0.1 M Timolol Maleate is (S)-1-tert-butylamino-3-(4-morpholino-
methanolic sodium hydroxide to produce a solution 1,2,5-thiadiazol-3-yloxy)propan-2-ol hydrogen maleate.
containing 0.0002 per cent w/v solution of Thyroxine Sodium. Timolol Maleate contains not less than 98.5 per cent and not
Reference solution. A 0.0002 per cent w/v solution of more than 101.0 per cent of C13H24N4O3S,C4H4O4, calculated
levothyroxine sodium RS in 0.1 M methanolic sodium on the dried basis.
hydroxide. Description. A white or almost white, crystalline powder or
Follow the procedure described under Assay. colourless crystals.
Calculate the content of C15H10I4NNaO4 in the tablet. Identification
Other tests. Comply with the tests stated under Tablets.
Test A may be omitted if tests B and C are carried out. Tests B
Assay. Determine by liquid chromatography (2.4.14). and C may be omitted if test A is carried out.
Test solution. Weigh accurately a quantity of the powdered A. Determine by infrared absorption spectrophotometry (2.4.6).
tablets containing about 1000 µg of Thyroxine Sodium, transfer Compare the spectrum with that obtained with timolol maleate
to a 100-ml volumetric flask and add 40 ml of 0.1 M methanolic RS or with the reference spectrum of timolol maleate.
sodium hydroxide. Shake for about 30 minutes, mix well and
dilute to volume with the same solvent, mix well and filter. B. In the test for Related substances, after exposure to iodine
vapour, the principal spot in the chromatogram obtained with
Reference solution. A 0.001 per cent w/v solution of test solution (b) corresponds to that in the chromatogram
levothyroxine sodium RS in 0.1 M methanolic sodium obtained with reference solution (b).
hydroxide.
C. Triturate 0.1 g with a mixture of 1 ml of 2 M sodium hydroxide
Chromatographic system and 3 ml of water and shake with three quantities, each of 5 ml,
– a stainless steel column 25 cm x 4.6 mm, packed with a of ether. To 0.1 ml of the aqueous layer add a solution of 10 mg
stationary phase consisting of porous silica particles of resorcinol in 3 ml of sulphuric acid and heat on a water-
(5 to 10 µm) to which nitrile groups are chemically bath for 15 minutes; no violet-red colour is produced. Neutralise
bonded, the remainder of the aqueous layer with 1 M sulphuric acid,
– mobile phase: a mixture of 65 volumes of water, 0.2 add 1 ml of bromine water, heat on a water-bath for 15 minutes,
volume of orthophosphoric acid and then heat to boiling and cool. To 0.2 ml of this solution add a
35 volumes of acetonitrile, solution of 10 mg of resorcinol in 3 ml of sulphuric acid and
– flow rate. 1 ml per minute, heat on a water-bath for 15 minutes; a violet-red colour is
– spectrophotometer set at 225 nm, produced. Add 0.2 ml of a 10 per cent w/v solution of potassium
– a 20 µl loop injector. bromide and heat on a water-bath for 5 minutes; the colour
Inject the reference solution. The test is not valid unless the changes to violet-blue.
relative standard deviation for replicate injections is not more
than 2.0 per cent.
Tests

Inject alternately the test solution and the reference solution. Appearance of solution. A 2.0 per cent w/v solution in carbon
dioxide-free water is clear (2.4.1), and not more intensely
Calculate the content of C15H10I4NNaO4 in the tablets. coloured than reference solution BS8 (2.4.1).
Storage. Store protected from light and moisture. pH (2.4.24). 3.8 to 4.3, determined in a 2.0 per cent w/v solution.
Labelling. The label states the strength in terms of the Specific optical rotation (2.4.22). –5.7º to –6.2º, determined in
equivalent amount of anhydrous thyroxine sodium. a 10.0 per cent w/v solution in M hydrochloric acid.

1182
IP 2007 TIMOLOL EYE DROPS

Related substances. Determine by thin-layer chromatography quantities, each of 40 ml, of dichloromethane. Reserve the
(2.4.17), coating the plate with silica gel GF254. aqueous layer for test B. Dry the combined dichloromethane
Mobile phase. A mixture of 80 volumes of dichloromethane, extracts with anhydrous sodium sulphate and evaporate to
20 volumes of methanol and 1 volume of strong ammonia dryness. Dry the residue at 60º at a pressure of 2 kPa for
solution. 15 minutes.

Test solution (a). Dissolve 0.5 g of the substance under On the residue, determine by infrared absorption
examination in 10 ml of methanol. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with timolol RS or with the reference spectrum of
Test solution (b). Dissolve 0.1 g of the substance under timolol.
examination in 100 ml of methanol.
B. Filter the aqueous layer reserved in test A and evaporate to
Reference solution (a). A 0.02 per cent w/v solution of the about 1 ml. Add 1 ml of bromine solution, heat in a water-bath
substance under examination in methanol. for 10 minutes, boil, cool and add 0.1 ml of the solution to a
Reference solution (b). A 0.1 per cent w/v solution of timolol solution of 10 mg resorcinol in 3 ml of sulphuric acid; a bluish
maleate RS in methanol. black colour is produced on heating in a water-bath for
15 minutes.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm
and then expose to iodine vapour for 2 hours and examine in
Tests
daylight. By both methods of visualisation, any secondary pH (2.4.24). 6.5 to 7.5.
spot in the chromatogram obtained with test solution (a) is
not more intense than the spot in the chromatogram obtained Related substances. Determine by liquid chromatography
with reference solution (a). (2.4.14).

Sulphated ash (2.3.18). Not more than 0.1 per cent. Test solution. Use the undiluted eye drops.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Reference solution (a). Dilute 1 volume of the eye drops to
on 1.0 g by drying in an oven at 105º. 250 volumes with the mobile phase.
Assay. Weigh accurately about 0.35 g, dissolve in 60 ml of Reference solution (b). Dilute 1 volume of the eye drops to
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric 500 volumes with the mobile phase.
acid, determining the end-point potentiometrically (2.4.25).
Reference solution (c). A 0.3 per cent w/v solution of maleic
Carry out a blank titration.
acid in the mobile phase.
1 ml of 0.1 M perchloric acid is equivalent to 0.04325 g of
Chromatographic system
C13H24N4O3S, C4H4O4.
– a stainless steel column 20 cm x 4 mm, packed with
Storage. Store protected from light and moisture. octadecylsilane bonded to porous silica (10 µm),
– mobile phase: a mixture of 57.5 volumes of methanol
and 42.5 volumes of 0.02 M sodium octanesulphonate,
adjusted to pH 3.0 with glacial acetic acid,
Timolol Eye Drops – flow rate. 2 ml per minute,
– spectrophotometer set at 295 nm,
Timolol Maleate Eye Drops – a 20 µl loop injector.
Timolol Eye Drops are a sterile solution of Timolol Maleate in Record the chromatogram of the test solution for 4 times the
Purified Water. They may contain suitable antimicrobial retention time of the principal peak. In the chromatogram
preservatives, buffering agents, stabilisers and suitable obtained with the test solution the area of any secondary
substances to increase the viscosity of the solution. peak, other than the peak corresponding to maleic acid, is not
Timolol Eye Drops contain not less than 90.0 per cent and not greater than the area of the peak in the chromatogram obtained
more than 110.0 per cent of the stated amount of timolol, with reference solution (a) and not more than two such peaks
C13H24N4O3S. have an area greater than that of the peak obtained with
reference solution (b).
Identification Other tests. Comply with the tests stated under Eye Drops.
A. Add a volume containing 50 mg of timolol to an equal Assay. To a volume containing about 25 mg of timolol add
volume of carbonate buffer pH 9.7 and extract with two water to produce 50.0 ml and mix. To 5.0 ml add 15 ml of

1183
TIMOLOL TABLETS IP 2007

carbonate buffer pH 9.7 and extract with three quantities, Test solution. Shake a quantity of the powdered tablets
each of 20 ml, and one quantity of 10 ml of toluene. Wash each containing 0.1 g of Timolol Maleate with 10 ml of the mobile
extract successively with the same 10-ml volume of carbonate phase for 10 minutes and filter.
buffer pH 9.7. Combine the toluene extracts and extract with
Reference solution (a). Dilute 1 volume of the test solution to
four quantities, each of 20 ml, of 0.05 M sulphuric acid.
250 volumes with the mobile phase.
Combine the extracts, dilute to 100.0 ml, filter and measure the
absorbance of the filtrate at the maximum at about 295 nm Reference solution (b). Dilute 1 volume of the test solution to
(2.4.7), using as the blank a solution prepared by treating 500 volumes with the mobile phase.
5.0 ml of water in the same manner beginning at the words
Chromatographic system
“add 15 ml of carbonate buffer pH 9.7...”.
– a stainless steel column 20 cm x 4 mm, packed with
Calculate the content of C13H24N4O3S taking 279 as the specific octadecylsilane bonded to porous silica (10 µm),
absorbance at 295 nm. – mobile phase: a mixture of 57.5 volumes of methanol
and 42.5 volumes of 0.02 M sodium octanesulphonate,
Storage. Store protected from light and moisture.
adjusted to pH 3.0 with glacial acetic acid,
Labelling. The label states (1) the strength in terms of the – flow rate. 2 ml per minute,
equivalent amount of timolol; (2) the names and concentration – spectrophotometer set at 295 nm,
of any added antimicrobial preservatives. – a 20 µl loop injector.
Record the chromatogram of the test solution for 4 times the
retention time of the principal peak. In the chromatogram
obtained with the test solution the area of any secondary
peak, other than the peak corresponding to maleic acid, is not
Timolol Tablets greater than the area of the peak in the chromatogram obtained
Timolol Maleate Tablets with reference solution (a) and not more than two such peaks
have an area greater than that of the peak obtained with
Timolol Tablets contain not less than 90.0 per cent and not reference solution (b).
more than 110.0 per cent of the stated amount of timolol maleate,
C13H24N4O3S,C4H4O4. Uniformity of content. Comply with the test stated under
Tablets.
Identification To one tablet add 25.0 ml of 0.05 M sulphuric acid, shake for
20 minutes and complete the Assay beginning at the words
A.To a quantity of the powdered tablets containing 70 mg of
“and centrifuge....”.
Timolol Maleate add 20 ml of carbonate buffer pH 9.7 and
extract with two quantities, each of 40 ml, of dichloromethane. Other tests. Comply with the tests stated under Tablets.
Reserve the aqueous layer for test B. Dry the extracts with
Assay. Weigh and powder 20 tablets. Weigh accurately a
anhydrous sodium sulphate, evaporate to dryness using a
quantity of the powder containing about 15 mg of Timolol
rotary evaporator, dry the residue at 60º at a pressure of 2 kPa
Maleate, add 25.0 ml of 0.05 M sulphuric acid, shake for
for 15 minutes.
20 minutes and centrifuge until clear. Add 5.0 ml of the resulting
On the residue, determine by infrared absorption supernatant liquid to 15 ml of carbonate buffer pH 9.7 and
spectrophotometry (2.4.6). Compare the spectrum with that extract with three quantities, each of 20 ml, and one quantity
obtained with timolol RS or with the reference spectrum of of 10 ml of toluene. Wash each extract successively with the
timolol. same 10-ml volume of carbonate buffer pH 9.7, combine the
toluene extracts and extract with four quantities, each of 20 ml,
B. Filter the aqueous layer reserved in test A and evaporate to
of 0.05 M sulphuric acid. Combine the acid extracts, dilute to
about 1 ml. Add 1 ml of bromine water, heat on a water-bath
100.0 ml, filter and measure the absorbance of the filtrate at the
for 15 minutes, then heat to boiling and cool. Add 0.1 ml of this
maximum at about 295 nm (2.4.7), using as the blank a solution
solution to a solution of 10 mg of resorcinol in 3 ml of sulphuric
prepared by treating a mixture of 5 ml of water and 15 ml of
acid and heat on a water-bath for 15 minutes; a bluish black
carbonate buffer pH 9.7 in the same manner beginning at the
colour is produced.
words “and extract with three quantities,...”
Tests Calculate the content of C13H24N4O3S,C4H4O4 taking 204 as
the specific absorbance at 295 nm.
Related substances. Determine by liquid chromatography
(2.4.14). Storage. Store protected from moisture.

1184
IP 2007 TINIDAZOLE TABLETS

Tinidazole test solution is not more intense than the spot in the
chromatogram obtained with the reference solution.
NO2 O O Sulphated ash (2.3.18). Not more than 0.2 per cent.
S CH3 Loss on drying (2.4.19). Not more than 2.0 per cent, determined
N
on 1.0 g by drying in an oven at 105º for 4 hours.
N
CH3 Assay. Weigh accurately about 0.5 g, dissolve in 30 ml of
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
C8H13N3O4S Mol. Wt. 247.3 acid, using 0.15 ml of nile blue A solution as indicator. Carry
Tinidazole is 1-[2-(ethylsulphonyl)ethyl]-2-methyl-5- out a blank titration.
nitroimidazole. 1 ml of 0.1 M perchloric acid is equivalent to 0.02473 g of
Tinidazole contains not less than 98.0 per cent and not more C8H13N3O4S.
than 100.5 per cent of C8H13N3O4S, calculated on the dried Storage. Store protected from light and moisture.
basis.
Description. Pale yellow crystals or a crystalline powder;
odour, slight and characteristic.
Tinidazole Tablets
Identification Tinidazole Tablets contain not less than 95.0 per cent and not
more than 105.0 per cent of the stated amount of tinidazole,
Test A may be omitted if tests B and C are carried out. Tests B
C8H13N3O4S. The tablets may be coated.
and C may be omitted if test A is carried out.
A. Determine by infrared absorption spectrophotometry (2.4.6). Identification
Compare the spectrum with that obtained with tinidazole RS
A. When examined in the range 230 nm to 360 nm (2.4.7), the
or with the reference spectrum of tinidazole.
final solution obtained in the Assay shows an absorption
B. When examined in the range 230 nm to 360 nm (2.4.7), a maximum at about 310 nm.
0.001 per cent w/v solution in methanol shows an absorption
Extract a quantity of the powdered tablets containing 0.1 g of
maximum at about 310 nm; absorbance at about 310 nm, about
Tinidazole with 10 ml of methanol, filter and evaporate the
0.35.
filtrate to dryness. The residue complies with the following
C. To about 5 mg add 5 ml of 0.1 M hydrochloric acid, 50 mg tests.
of zinc powder, 4 ml of hydrochloric acid and allow to stand
B. To about 5 mg add 5 ml of 0.1 M hydrochloric acid, 50 mg
for 30 minutes. Add 4 ml of a 1 per cent w/v solution of vanillin,
of zinc powder, 4 ml of hydrochloric acid and allow to stand
heat on a boiling water-bath for 20 minutes, allow to cool to
for 30 minutes. Add 4 ml of a 1 per cent w/v solution of vanillin,
room temperature and dilute to 20 ml with water; a greenish
heat on a boiling water-bath for 20 minutes, allow to cool to
yellow colour is produced.
room temperature and dilute to 20 ml with water; a greenish
Tests yellow colour is produced.
C. Melting range (2.4.21). 125º to 128º.
Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254. Tests
Mobile phase. A mixture of 95 volumes of ethyl acetate,
Other tests. Comply with the tests stated under Tablets.
5 volumes of methanol and 5 volumes of diethylamine.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Test solution. Dissolve 0.2 g of the substance under
quantity of the powder containing about 0.15 g of Tinidazole,
examination in 10 ml of a mixture of equal volumes of chloroform
add 20 ml of methanol, shake well and add sufficient methanol
and methanol.
to produce 100.0 ml. Mix well and filter. Dilute 10.0 ml of the
Reference solution. A 0.02 per cent w/v solution of the solution to 100.0 ml with methanol and further dilute 10.0 ml of
substance under examination in a mixture of equal volumes of this solution to 100.0 ml with methanol. Measure the
chloroform and methanol. absorbance of the resulting solution at the maximum at about
Apply to the plate 10 µl of each solution. After development, 310 nm (2.4.7).
dry the plate in air and examine in ultraviolet light at 254 nm. Calculate the content of C8H13N3O4S taking 356 as the specific
Any secondary spot in the chromatogram obtained with the absorbance at 310 nm.

1185
TITANIUM DIOXIDE IP 2007

Storage. Store protected from light and moisture. Barium. Shake 20.0 g with 30 ml of hydrochloric acid, add
100 ml of distilled water and boil. Filter while hot through a
hardened filter paper until a clear filtrate is obtained. Wash the
Titanium Dioxide filter with 60 ml of distilled water and dilute the combined
filtrate and washings to 200 ml with distilled water. To 10 ml of
TiO2 Mol. Wt. 79.9 this solution add 1 ml of 1 M sulphuric acid. After 30 minutes
Titanium Dioxide contains not less than 98.0 per cent and not any opalescence is not more intense than that of a mixture of
more than 100.5 per cent of TiO2. 10 ml of the test solution and 1 ml of distilled water.
Description. A white or almost white, infusible powder; Heavy metals (2.3.13). Dilute 10 ml of the solution prepared in
odourless. the test for Barium to 20 ml with water. 12 ml of the solution
complies with the limit test for heavy metals, Method D
Identification (20 ppm).
A. When strongly heated it becomes pale yellow; the colour Iron. To 8 ml of solution A add 4 ml of water, mix and add
is discharged on cooling. 0.05 ml of bromine water, allow to stand for 5 minutes, remove
the excess of bromine with a current of air and add 3 ml of
B. To 0.5 g add 5 g of anhydrous sodium sulphate and 10 ml of potassium thiocyanate solution. Any colour in the solution
water and mix. Add 10 ml of sulphuric acid and boil gently is not more intense than that in a standard prepared at the
until clear; cool, add slowly 30 ml of a 25 per cent v/v solution same time and in the same manner using a mixture of 4 ml of
of sulphuric acid and dilute with water to 100 ml (solution A). iron standard solution (2 ppm Fe) and 8 ml of a 20 per cent
To 5 ml of solution A add 0.1 ml of strong hydrogen peroxide w/v solution of sulphuric acid (200 ppm).
solution; an orange-red colour is produced.
Assay. Weigh accurately about 0.5 g, transfer to a 300-ml
C. To 5 ml of solution A add 0.5 g of zinc, in granules; after Kjeldahl flask, add 5 g of anhydrous sodium sulphate and
45 minutes a violet-blue colour is produced. 10 ml of water, mix and add 10 ml of sulphuric acid. Boil gently
until clear, cool, add slowly 40 ml of cooled sulphuric acid
Tests (25 per cent), cool again and dilute with water to 100.0 ml
Appearance of solution. Solution A is not more opalescent (solution B). To 300 g of zinc, in granules, add 300 ml of a 2 per
than opalescence standard OS2 (2.4.1), and colourless (2.4.1). cent w/v solution of mercuric nitrate and 2 ml of nitric acid,
shake for 10 minutes and wash with water. Pack the zinc
Acidity or alkalinity. Shake 5.0 g with 50 ml of carbon dioxide-
amalgam into a glass tube (400 mm x 20 mm) fitted with a tap
free water for 5 minutes and centrifuge until a clear solution is
and a filter plate. Pass through the column, at a rate of about
obtained. To 10 ml of the solution add 0.1 ml of bromothymol
3 ml per minute, 100 ml of 1 M sulphuric acid followed by
blue solution. Not more than 1.0 ml of either 0.01 M
100 ml of water, ensuring that the amalgam is covered with
hydrochloric acid or 0.01 M sodium hydroxide is required to
liquid throughout. Pass slowly through the column, at a rate
change the colour of the solution.
of about 3 ml per minute, 200 ml of 0.5 M sulphuric acid
Water-soluble substances. Not more than 0.5 per cent, followed by 100 ml of water. Collect the combined eluates in a
determined by the following method. Boil 10.0 g for 5 minutes 500-ml conical flask containing 50 ml of a 15 per cent w/v
with 150 ml of water containing 0.5 g of ammonium sulphate. solution of ferric ammonium sulphate in sulphuric acid
Cool, dilute to 200 ml with water and filter until a clear solution (25 per cent) and titrate immediately with 0.1 M ceric
is obtained. Evaporate 100 ml of the filtrate to dryness ignite ammonium nitrate using ferroin solution as indicator until a
and weigh. greenish colour is obtained (n1 ml). Pass slowly through the
Arsenic (2.3.10). To 0.2 g in a 100-ml Kjeldahl flask add 2 g of column 100 ml of 0.5 M sulphuric acid followed by 20.0 ml of
anhydrous sodium sulphate, 7 ml of sulphuric acid and 5 ml solution B, wash with 100 ml of 0.5 M sulphuric acid followed
of nitric acid. Heat gently until a clear solution is obtained, by 100 ml of water. Collect the combined eluates in a 500-ml
cool, add 10 ml of water, cool again and add 5 g of hydrazine conical flask containing 50 ml of a 15 per cent w/v solution of
reducing mixture and 10 ml of hydrochloric acid. Immediately ferric ammonium sulphate in sulphuric acid (25 per cent),
attach an air condenser and distil into 15 ml of cooled water rinse the lower end of the column with water and titrate
until a total volume of 30 ml is obtained. Rinse the condenser immediately with 0.1 M ceric ammonium nitrate using ferroin
with 5 ml of water and dilute the combined distillate and solution as indicator until a greenish colour is obtained
rinsings to 40 ml with water. 20 ml of the resulting solution (n2 ml). Calculate the percentage content of TiO2 from the
complies with the limit test for arsenic. Use a mixture of 0.5 ml expression 3.99(n2 - n1)/w
of arsenic standard solution (1 ppm As) and 24.5 ml of water Where, w is the weight, in g, of the substance under
to prepare the standard (5 ppm). examination used in the preparation of solution A.

1186
IP 2007 TIOTROPIUM POWDER FOR INHALATION

Storage. Store protected from moisture. Avoid contact with adjusting the pH to 5.5 with dilute acetic acid (10 per
aluminium. cent v/v),
– flow rate. 1 ml per minute,
– spectrophotometer set at 235 nm,
– a 20 µl loop injector.
Tiotropium Bromide Monohydrate Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0 and the column efficiency in
H3C not less than 3000 theoretical plates.
N CH3 Inject the test solution. Any individual impurity is not more
than 0.5 per cent and the sum of all the impurities is not more
O than 1.0 per cent.
S
Br, HCl Heavy metals (2.3.13). 1.0 g complies with the limit test for
O S heavy metals, Method B (20 ppm).
OH Sulphated ash (2.3.18). Not more than 0.3 per cent.
O
Water (2.3.43). 3.5 to 4.5 per cent, determined on 0.5 g.
C19H22BrNO4S2, HCl Mol. Wt. 508.9 Assay. Determine by liquid chromatography (2.4.14).
Tiotropium Bromide Monohydrochloride is 6β,7β-epoxy-3β- Test solution. Dissolve 20 mg of the substance under
hydroxy-8-methyl-1αH-5αH-tropanium bromide examination in 100.0 ml of the mobile phase. Dilute 5.0 ml of
hydrochloride. the resulting solution to 50.0 ml with the mobile phase.
Tiotropium Bromide Monohydrate contains not less than 98.0 Reference solution. A 0.002 per cent w/v solution of tiotropium
per cent and not more than 102.0 per cent of tiotropium bromide monohydrate RS in the mobile phase.
bromide, C19H22NO4S2Br, calculated on the anhydrous basis.
Chromatographic system
Description. A white to off-white powder.
– a stainless steel column 25 cm ´ 4.6 mm, packed with
Identification octadecylsilane bonded to porous silica
(5 µm),
A. Determine by infrared absorption spectrophotometry ( – mobile phase: a mixture of 45 volumes of methanol and
2.4.6). Compare the spectrum with that obtained with 55 volumes of a buffer solution prepared by dissolving
tiotropium bromide monohydrate RS or with the reference 3.85 g of ammonium acetate in 1000 ml of water and
spectrum of tiotropium bromide monohydrate. adjusting the pH to 5.5 with dilute acetic acid ( 10 per
B. In the Assay, the principal peak in the chromatogram cent v/v),
obtained with test solution corresponds to the principal peak – flow rate. 1 ml per minute,
in the chromatogram obtained with the reference solution. – spectrophotometer set at 235 nm,
– a 20 µl loop injector.
Tests Inject the reference solution. The test is not valid unless the
tailing factor is not more than 2.0, the column efficiency in not
Related substances. Determine by liquid chromatography
less than 3000 theoretical plates and the relative standard
(2.4.14).
deviation for replicate injections is not more than 2.0 per cent.
Test solution. Dissolve 10 mg of the substance under
Inject the test solution and the reference solution.
examination in 100 ml of the mobile phase.
Calculate the content of C19H22NO4S2,Br.
Reference solution. A 0.002 per cent w/v solution of tiotropium
bromide monohydrate RS in the mobile phase. Storage. Store protected from light and moisture.
Chromatographic system
– a stainless steel column 25 cm ´ 4.6 mm, packed with
octadecylsilane bonded to porous silica Tiotropium Powder for Inhalation
(5 µm), Tiotropium Powder for Inhalation consists of Tiotropium
– mobile phase: a mixture of 45 volumes of methanol and Bromide in microfine powder either alone or admixed with
55 volumes of a buffer solution prepared by dissolving Lactose in a pre-metered unit for use in a suitable powder
3.85 g of ammonium acetate in 1000 ml of water and inhaler.

1187
TIZANIDINE HYDROCHLORIDE IP 2007

Tiotropium Powder for Inhalation contains not less than 90.0 Tizanidine Hydrochloride
per cent and not more than 125.0 per cent of the stated amount
of tiotropium, C19H22NO4S2 per unit dose.
N
S
Identification
Cl N
In the Assay, the principal peak in the chromatogram obtained NH , HCl
N
with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution. NH

Tests C9H8ClN5S.HCl Mol. Wt. 290.2


Other tests. Complies with the tests stated under Inhalation Tizanidine Hydrochloride is 5-chloro-N-(2-imidazolin-2-yl)-
Preparations (Powders for Inhalation). 2,1,3-benzothiadiazol-4-yl-amine.
Follow the procedure described under Assay with suitable Tizanidine Hydrochloride contains not less than 98.0 per cent
dilution of the reference solution wherever the amount of and not more than 102.0 per cent of C9H8ClN5S.HCl, calculated
active substance is to be determined in any test. on the dried basis.
Assay. Description. A white to yellowish white, crystalline powder.
Determine by liquid chromatography (2.4.14). Identification
Solvent mixture. 90 volumes of 0.05 per cent v/v
A. Determine by infrared absorption spectrophotometry (2.4.6).
orthophosphoric acid and 10 volumes of acetonitrile.
Compare the spectrum with that obtained with tizanidine
Test solution. To 10 intact capsules add about 70 ml of the hydrochloride RS.
solvent mixture and disperse with the aid of ultrasound for
B. In the Assay, the principal peak in the chromatogram
about 5 minutes with intermittent shaking. Add sufficient of
obtained with the test solution corresponds to the peak in the
the solvent mixture to produce 100.0 ml and dilute suitably, if
chromatogram obtained with the reference solution.
required, to get a solution containing 1.8 µg per ml of
Tiotropium per ml. C. Gives reaction A for chlorides (2.3.1).
Reference solution. A solution containing 1.8 µg per ml of
tiotropium bromide RS per ml in the solvent mixture.
Tests

Chromatographic system pH (2.4.24). 3.5 to 5.3, determined on 5.0 per cent w/v solution
– a stainless steel column 15 cm x 4.6 mm, packed with in water.
octadecylsilyl silica gel (5 mm), Related substances. Determine by liquid chromatography
– mobile phase: a mixture of 80 volumes of a buffer solution (2.4.14).
prepared by dissolving 2 ml triethylamine in 1000 ml of
water and adjusting the pH to 2.5 with orthophosphoric Test solution. Dissolve 50 mg of the substance under
acid, and 20 volumes of acetonitrile, examination in 50.0 ml of the mobile phase.
– flow rate. 2 ml per minute, Reference solution (a). A 0.1 per cent w/v solution of
– spectrophotometer set at 237 nm, tizanidine hydrochloride RS in the mobile phase.
– inject 200 µl.
Reference solution (b). Dilute 1 ml of reference solution (a) to
Inject the reference solution. The test is not valid unless the 100 ml with mobile phase.
column efficiency is not less than 4500 theoretical plates, the
Chromatographic system
tailing factor is not more than 1.7 and the relative standard
– a stainless steel column 25 cm x 4.6 mm packed with
deviation for replicate injections is not more than 2.0 per cent.
octadecylsilane bonded to porous silica (5 µm),
Inject the test solution and the reference solution.
– column temperature 50°,
Calculate the content of C19H22NO4S2 per unit. – mobile phase: a mixture of 80 volumes of buffer solution
Storage. Store protected from moisture, at temperature not prepared by dissolving 3.5 g of
exceeding 30º. sodium-1-pentane sulphonate in 1000 ml of water, adjust
the pH to 3.0 with 12 per cent
Labelling. The label states the quantity of active ingredient orthophosphoric acid solution or 1 M sodium hydroxide
per pre-metered unit. and 20 volumes of acetonitrile,

1188
IP 2007 TIZANIDINE TABLETS

– flow rate. 1 ml per minute, Storage. Store protected from light.


– spectrophotometer set at 230 nm,
– a 10 µl loop injector.
Inject reference solution (a). The test is not valid unless the Tizanidine Tablets
tailing factor is not more than 2.0 and the column efficiency in
not less than 5000 theoretical plates.
Tizanidine Hydrochloride Tablets
Tizanidine Tablets contain not less than 90.0 per cent and not
Inject the test solution and reference solution (b). In the
more than 110.0 per cent of the stated amount of tizanidine,
chromatogram obtained with the test solution, the area of any
C9H8ClN5S.
secondary peak is not more than 0.5 times the area of the peak
in the chromatogram obtained with reference solution (b) (0.5 Identification
per cent) and the sum of areas of all the secondary peaks is
not more than the area of the peak in the chromatogram In the Assay, the principal peak in the chromatogram obtained
obtained with the reference solution (b) (1.0 per cent). with the test solution corresponds to the peak in the
chromatogram obtained with the reference solution.
Heavy metals (2.3.13). Dissolve 1.0 g in 20 ml of water. 12 ml of
this solution complies with limit test for heavy metals, Method Tests
D (20 ppm).
Dissolution (2.5.2).
Total Chloride. 11.9 per cent to 12.5 per cent.
Apparatus. No 1
Weigh accurately about 0.5 g and dissolve in 50 ml of water. Medium: 900 ml of 0.1 M hydrochloric acid.
Titrate with 0.1 M silver nitrate. Determine the end point Speed and time. 50 rpm for 45 minutes.
potentiometrically (2.4.25). Carry out a blank titration.
Withdraw a suitable volume of the medium and filter. Measure
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of the absorbance of the filtrate, suitably diluted with dissolution
chloride. medium if necessary, at the maximum at about 320 nm (2.4.7).
Sulphated ash (2.3.18). Not more than 0.1 per cent. Calculate the content of C9H8ClN5S in the medium from the
Loss on drying (2.4.19). Not more than 0.5 per cent, determined absorbance obtained from a solution of known concentration
on 1 g by drying in an oven at 105° for 3 hours. of tizanidine Hydrochloride RS in the same medium.

Assay. Determine by liquid chromatography (2.4.14). D. Not less than 70 per cent of the stated amount of C9H8ClN5S.

Test solution. Dissolve 25 mg of the substance under Uniformity of content. Comply with the tests stated under
examination in 50.0 ml of the mobile phase. Dilute 10.0 ml of Tablets.
the solution to 50.0 ml with the same solvent. Determine by liquid chromatography (2.4.14), as described
Reference solution. A 0.01 per cent w/v solution of tizanidine under Assay.
hydrochloride RS in the mobile phase. Test solution. Crush one tablet and disperse in 50 ml phosphate
Chromatographic system buffer pH 6.6, dilute to 100.0 ml with acetonitrile and filter.
– a stainless steel column 15 cm x 4.6 mm packed with Calculate the content of C9H8ClN5S.
octylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 50 volumes of a buffer solution
prepared by dissolving 6.8 g of Other tests. Comply with the tests stated under Tablets.
monobasic potassium phosphate in 1000 ml of water Assay. Determine by liquid chromatography (2.4.14).
adjusting the pH to 7.5 with 5.3 M
Test solution. Weigh and powder 20 tablets. Weigh accurately
potassium hydroxide, and 50 volumes of acetonitrile,
a quantity of the powdered tablet containing about 20 mg of
– flow rate. 1 ml per minute,
Tizanidine, disperse in 50 ml phosphate buffer pH 6.6 and
– spectrophotometer set at 230 nm,
dilute to 100.0 ml with acetonitrile and filter.
– a 10 µl loop injector.
Reference solution. Weigh accurately 10 mg of
Inject the reference solution. The test is not valid unless the
tizanidine hydrochloride RS, dissolve in 25 ml phosphate
tailing factor is not more than 2.0 and the relative standard
buffer pH 6.6 and dilute to 50.0 ml with acetonitrile.
deviation for replicate injections is not more than 2.0 per cent.
Chromatographic system
Inject the test solution and the reference solution.
– a stainless steel column 25 cm x 4.6 mm packed with
Calculate the content of C9H8ClN5S.HCl. octadecylsilane bonded to porous silica (5 µm),

1189
TOBRAMYCIN IP 2007

– mobile phase: a mixture of 80 volumes of phosphate Test solution. Dissolve 0.4 g of the substance under
buffer pH 6.6 and 20 volumes of acetonitrile, examination in 100 ml of water.
– flow rate. 1 ml per minute,
Reference solution (a). A 0.4 per cent w/v solution of
– spectrophotometer set at 320 nm,
tobramycin RS in water.
– a 20 µl loop Injector.
Inject the reference solution. The test is not valid unless the Reference solution (b). A solution containing 0.4 per cent
relative standard deviation for replicate injections is not more w/v each of kanamycin sulphate RS, neomycin sulphate RS
than 2.0 per cent. and tobramycin RS in water.

Inject the test solution and the reference solution. Apply to the plate 5 µl of each solution. After development,
dry the plate in warm air, spray with a mixture of equal volumes
Calculate the content of C9H8ClN5S. of a 46 per cent w/v solution of sulphuric acid and a 0.2 per
Storage. Store protected from light and moisture. cent w/v solution of 1,3-naphthalenediol in ethanol (95 per
cent) and heat at 105º for 10 minutes. The principal spot in the
Labelling. The label states the strength in terms of the
chromatogram obtained with the test solution corresponds to
equivalent amount of Tizanidine.
that in the chromatogram obtained with reference solution (a).
The test is not valid unless the chromatogram obtained with
reference solution (b) shows three clearly separated principal
Tobramycin spots.
B. Dissolve about 5 mg in 5 ml of water, add 5 ml of a 0.1 per
OH cent w/v solution of ninhydrin in ethanol (95 per cent) and
heat in a water-bath for 3 minutes; a violet-blue colour
develops.
O
NH2 HO
H2N
Tests
O OH pH (2.4.24). 9.0 to 11.0, determined in a 10.0 per cent w/v
HO solution.
O
Specific optical rotation (2.4.22). +138º to +148º, determined
NH2 HO in a 4.0 per cent w/v solution.
NH2
O 2-Methyl-1-propanol. Not more than 1.0 per cent w/w,
H2N determined by gas chromatography (2.4.13).
Test solution (a). A 10 per cent w/v solution of the substance
C18H37N5O9 Mol. Wt. 467.3 under examination in water.
Tobramycin is 6-O-(3-amino-3-deoxy-α-D-glucopyranosyl)- Test solution (b). A solution containing 10 per cent w/v of the
2-deoxy-4-O-(2,6-diamino-2,3,6-trideoxy-α -D-ribo- substance under examination and 0.2 per cent v/v of
hexopyranosyl)-D-streptamine, an antimicrobial substance 2-propanol (internal standard).
produced by Streptomyces tenebrarius or by any other
means. Reference solution. A solution containing 0.1 per cent w/v of
2-methyl-1-propanol and 0.2 per cent v/v of the internal
Tobramycin has potency not less than 930 Units per mg, standard.
calculated on the anhydrous and 2-methyl-1-propanol-free
basis. Chromatographic system
– a glass column 1.5 m x 4 mm, packed with porous polymer
Description. A white or almost white powder.
beads (80 to 100 mesh) (such as Porapak Q),
Identification – temperature. column 165º,
Calculate the percentage w/w of 2-methyl-1-propanol.
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel H. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel H.
Mobile phase. A mixture of 60 volumes of methanol,
40 volumes of strong ammonia solution and 20 volumes of Mobile phase. A mixture of equal volumes of strong ammonia
chloroform. solution, 2-butanone and ethanol (95 per cent).

1190
IP 2007 TOBRAMYCIN INJECTION

Test solution. Dissolve 0.8 g of the substance under Tobramycin Injection contains not less than 90.0 per cent and
examination in 100 ml of 0.02 M ammonia. not more than 120.0 per cent of the stated amount of
Reference solution. A 0.008 per cent w/v solution of the tobramycin, C18H37N5O9.
substance under examination in 0.02 M ammonia. Description. A colourless solution.
Apply to the plate 5 µl of each solution. After development, Identification
dry the plate in warm air, heat at 110º for 10 minutes and spray
the hot plate with a solution prepared immediately before use A. Determine by thin-layer chromatography (2.4.17), coating
by diluting sodium hypochlorite solution (3 per cent Cl) the plate with silica gel H.
with water to contain 0.5 per cent w/v of available chlorine.
Mobile phase. A mixture of 60 volumes of methanol,
Dry in a current of cold air until a sprayed area of the plate
40 volumes of strong ammonia solution and 20 volumes of
below the line of application gives at most a very faint blue
chloroform.
colour with a drop of potassium iodide and starch solution;
avoid prolonged exposure to cold air. Spray the plate with Test solution. Dilute a suitable volume of the injection with
potassium iodide and starch solution. Any secondary spot water to produce a solution containing 0.4 per cent w/v
in the chromatogram obtained with the test solution is not solution of tobramycin.
more intense than the spot in the chromatogram obtained Reference solution (a). A 0.4 per cent w/v solution of
with the reference solution. tobramycin RS in water.
Sulphated ash (2.3.18). Not more than 0.3 per cent. Reference solution (b). A solution containing 0.4 per cent
Water (2.3.43). Not more than 8.0 per cent, determined on w/v each of kanamycin sulphate RS, neomycin sulphate RS
0.3 g. and tobramycin RS in water.
Assay. Determine by the microbiological assay of antibiotics, Apply to the plate 5 µl of each solution. After development,
Method B (2.2.10). dry the plate in warm air, spray with a mixture of equal volumes
of a 46 per cent w/v solution of sulphuric acid and a 0.2 per
Tobramycin intended for use in the manufacture of parenteral
cent w/v solution of 1,3-naphthalenediol in ethanol (95 per
preparations without a further appropriate procedure for
cent) and heat at 105º for 10 minutes. The principal spot in the
the removal of bacterial endotoxins complies with the
chromatogram obtained with the test solution corresponds to
following additional requirement.
that in the chromatogram obtained with reference solution (a).
Bacterial endotoxins (2.2.3). Not more than 2.0 Endotoxin Units The test is not valid unless the chromatogram obtained with
per mg of tobramycin. reference solution (b) shows three clearly separated principal
Tobramycin intended for use in the manufacture of parenteral spots.
preparations or eye drops without a further appropriate B. Gives the reactions of sulphates (2.3.1).
sterilisation procedure complies with the following
additional requirements. Tests
Sterility (2.2.11). Complies with the test for sterility. pH (2.4.24). 3.5 to 6.0.
Storage. Store protected from moisture. If the material is sterile, Related substances. Determine by thin-layer chromatography
the container should be sterile, tamper-evident and sealed so (2.4.17), coating the plate with silica gel H.
as to exclude micro-organisms.
Mobile phase. A mixture of equal volumes of strong ammonia
Labelling. The label states (1) the number of Units per mg; (2) solution, 2-butanone and ethanol (95 per cent).
where applicable, that it is sterile; (3) where applicable, that it
Test solution. Dilute a suitable volume of the injection with
is free from bacterial endotoxins and depressor substances.
0.01 M ammonia to obtain a solution containing 40 mg of
Tobramycin in 4 ml. Shake with 10 ml of ether and use the
aqueous layer.

Tobramycin Injection Reference solution. A 0.008 per cent w/v solution of the
substance under examination in 0.02 M ammonia.
Tobramycin Sulphate Injection Apply to the plate 5 µl of each solution. After development,
Tobramycin Injection is a sterile solution of Tobramycin in dry the plate in warm air, heat at 110º for 10 minutes and spray
Water for Injections containing sufficient Sulphuric Acid to the hot plate with a solution prepared immediately before use
adjust the pH to 3.5 to 6.0. by diluting sodium hypochlorite solution (3 per cent Cl)

1191
TOCOPHERYL ACETATE IP 2007

with water to contain 0.5 per cent w/v of available chlorine. B. When examined in the range 230 nm to 360 nm (2.4.7), a
Dry in a current of cold air until a sprayed area of the plate 0.01 per cent w/v solution in ethanol exhibits a maximum at
below the line of application gives at most a very faint blue about 284 nm, a shoulder at about 278 nm and a minimum at
colour with a drop of potassium iodide and starch solution; about 254 nm.
avoid prolonged exposure to cold air. Spray the plate with C. Determine by thin-layer chromatography (2.4.17), coating
potassium iodide and starch solution. Any secondary spot the plate with silica gel HF254.
in the chromatogram obtained with the test solution is not
more intense than the spot in the chromatogram obtained Mobile phase. A mixture of 80 volumes of cyclohexane and
with the reference solution. 20 volumes of ether.
Test solution (a). Dissolve 0.5 g of the substance under
Bacterial endotoxins (2.2.3). Not more than 2.0 Endotoxin
examination in 100 ml of cyclohexane.
Units per mg of tobramycin.
Test solution (b). Dissolve 10 mg of the substance under
Other tests. Complies with the tests stated under Parenteral examination in 2 ml of 5 M ethanolic sulphuric acid, heat on
Preparations (Injections). a water-bath for 5 minutes, cool, add 2 ml of water and 2 ml of
Assay. Determine by the microbiological assay of antibiotics, cyclohexane and shake for 1 minute; use the upper layer.
Method B (2.2.10). Reference solution (a). A 0.5 per cent w/v solution of
Calculate the content of tobramycin in the injection, taking a-tocopheryl acetate RS in cyclohexane.
each 1000 Units found to be equivalent to 1 mg of tobramycin. Reference solution (b). Prepare in the same manner as test
The upper fiducial limits of error is not less than 97.0 per cent solution (b) but using 10 mg of a -tocopheryl acetate RS in
and not more than 110.0 per cent of the stated potency. place of the substance under examination.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with test
Tocopheryl Acetate solution (a) corresponds to that in the chromatogram obtained
α-Tocopheryl Acetate; α-Tocopherol Acetate; Vitamin with reference solution (a). There are two spots in each of the
E Acetate chromatograms obtained with test solution (b) and reference
solution (b). The spots of higher Rf value are due to a-
tocopheryl acetate and correspond to that in the
CH3
CH3 chromatogram obtained with reference solution (a). The spots
H3C O CH3
of lower Rf value are due to a-tocopheryl. Spray the plate with
CH3 CH3 CH3 a mixture of 1 volume of hydrochloric acid, 4 volumes of a
O 0.25 per cent w/v solution of ferric chloride in ethanol
CH3 (95 per cent) and 4 volumes of a 1.0 per cent w/v solution of
H3C O
1,10-phenanthroline hydrochloride in ethanol (95 per cent).
In the chromatograms obtained with test solution (b) and
C31H52O3 Mol. Wt. 472.8 reference solution (b) the spot of lower Rf value a-tocopheryl
Tocopheryl Acetate is (2RS,4'RS,8'RS)-6-acetoxy-2,5,7,8- is orange.
tetramethyl-2-(4', 8', 12'-trimethyltridecyl)chroman(all-rac-α-
tocopherol acetate).
Tests

Tocopheryl Acetate contains not less than 96.0 per cent and Refractive index (2.4.27). 1.494 to 1.498, determined at 20º.
not more than 102.0 per cent of C31H52O3. Acid value (2.3.23). Not more than 2.0, determined on 2.0 g.
Description. A clear, slightly greenish yellow, viscous, oily Free tocopherol. Not more than 2.0 per cent, determined by
liquid. the following method. Weigh accurately about 0.5 g, dissolve
in 100 ml of 0.25 M ethanolic sulphuric acid, add 20 ml of
Identification water and 0.1 ml of a 0.25 per cent w/v solution of
diphenylamine in sulphuric acid and titrate with 0.01 M ceric
Test A may be omitted if tests B and C are carried out. Tests B
ammonium nitrate until a blue colour is produced that persists
and C may be omitted if test A is carried out.
for at least 5 seconds. Repeat the operation without the
A. Determine by infrared absorption spectrophotometry (2.4.6). substance under examination. The difference between the
Compare the spectrum with that obtained with a-tocopheryl titrations represents the amount of ceric ammonium nitrate
acetate RS. required.

1192
IP 2007 TOLBUTAMIDE

1 ml of 0.01 M ceric ammonium nitrate is equivalent to 0.002154 263 nm and 275 nm and a shoulder at about 268 nm. Dilute the
g of tocopherol. solution with methanol to produce a 0.001 per cent w/v
Heavy metals (2.3.13). 1.0 g complies with the limit test for solution. When examined in the range 220 nm to 235 nm, the
heavy metals, Method B (20 ppm). resulting solution shows an absorption maximum at about
228 nm; absorbance at about 228 nm, about 0.50.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
C. Boil 0.1 g with 8 ml of a 50 per cent v/v solution of sulphuric
Assay. Weigh accurately about 0.3 g, dissolve in 25 ml of acid under a reflux condenser for 30 minutes. Make the solution
ethanol (95 per cent), add 20 ml of 2.5 M ethanolic sulphuric strongly alkaline with sodium hydroxide solution and steam
acid and heat on a water-bath under a reflux condenser for distil for 30 minutes, receiving the distillate in 20 ml of 0.1 M
3 hours. Cool, transfer the solution quantitatively to a 200-ml hydrochloric acid. To 1 ml of the solution containing the
volumetric flask, rinse the apparatus with ethanol (95 per distillate add 0.1 g of sodium acetate and 10 ml of buffer
cent) and add the rinsings to the flask. Make up to volume solution pH 9.4. Cool in an ice-bath for 10 minutes, add 1 ml of
with ethanol (95 per cent) and mix. To 25.0 ml of the resulting diazotised nitroaniline solution, set aside for 20 minutes and
solution in a flask add 25 ml of 0.25 M ethanolic sulphuric add dropwise 1 ml of sodium hydroxide solution; an orange-
acid, 10 ml of water and titrate with 0.01 M ceric ammonium red colour is produced.
nitrate using 0.1 ml of diphenylamine as indicator, until a blue
D. Boil 0.1 g with 8 ml of a 50 per cent v/v solution of sulphuric
colour persisting for at least 5 seconds is obtained. Repeat
acid under a reflux condenser for 30 minutes. Cool in an ice-
the operation without the substance under examination. The
bath; a crystalline precipitate of 4-toluenesulphonylamide is
difference between the titrations represents the amount of
formed, which after recrystallisation from hot water and drying
ceric ammonium nitrate required.
at 105º melts at 135º to 140º (2.4.21).
1 ml of 0.01 M ceric ammonium nitrate is equivalent to 0.002364
g of C31H52O3. Tests
Storage. Store protected from light and moisture. Appearance of solution. A 2.0 per cent w/v solution in 1 M
sodium hydroxide is clear (2.4.1), and colourless (2.4.1).
pH (2.4.24). 4.5 to 5.5, determined in a solution prepared by
Tolbutamide dissolving 2.0 g in 50 ml of carbon dioxide-free water by
heating at 70º for 5 minutes, cooling rapidly and filtering.
O O O Non-sulphonyl urea. Dissolve 0.5 g in 1 ml of dilute ammonia
S solution and 9 ml of water; not more than a faint opalescence
N N CH3
H H is produced.
H3C Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel G.
C12H18N2O3S Mol. Wt. 270.4
Mobile phase. A mixture of 90 volumes of chloroform,
Tolbutamide is 3-butyl-1-[(4-methylphenyl)sulphonyl]urea. 8 volumes of methanol and 2 volumes of anhydrous formic
Tolbutamide contains not less than 99.0 per cent and not more acid.
than 101.0 per cent of C12H18N2O3S, calculated on the dried Test solution. Dissolve 0.5 g of the substance under
basis. examination in 10 ml of acetone.
Description. A white, crystalline powder; almost odourless. Reference solution (a). A 0.015 per cent w/v solution of
4-toluenesulphonamide in acetone.
Identification
Reference solution (b). A mixture of equal volumes of the
Test A may be omitted if tests B, C and D are carried out. Tests test solution and reference solution (a).
B, C and D may be omitted if test A is carried out.
Apply to the plate 5 µl of each of test solution and reference
A. Determine by infrared absorption spectrophotometry (2.4.6). solution (a) and 10 µl of reference solution (b). After
Compare the spectrum with that obtained with tolbutamide development, dry the plate in a current of warm air and heat at
RS or with the reference spectrum of tolbutamide. 110º for 10 minutes. While still hot, place the plate in a
B. Dissolve 25 mg in sufficient methanol to produce 100.0 ml. chromatographic tank with an evaporating dish containing a
5 per cent w/v solution of potassium permanganate, add an
When examined in the range 245 nm to 360 nm (2.4.7), the equal volume of hydrochloric acid and close the tank. Leave
resulting solution shows absorption maxima at about 258 nm, the plate in the tank for 2 minutes, then place it in a current of

1193
TOLBUTAMIDE TABLETS IP 2007

cold air until the excess of chlorine is removed and an area of Mobile phase. A mixture of 90 volumes of chloroform,
coating below the line of application gives only a very faint 8 volumes of methanol and 2 volumes of anhydrous formic
blue colour with potassium iodide and starch solution; avoid acid.
prolonged exposure to cold air. Spray the plate with potassium
Test solution. Shake a quantity of the powdered tablets
iodide and starch solution and allow to stand for 5 minutes.
containing 0.5 g of Tolbutamide with 10 ml of acetone and
Any secondary spot in the chromatogram obtained with the
filter.
test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a). The Reference solution (a). A 0.015 per cent w/v solution of
chromatogram obtained with reference solution (b) shows two 4-toluenesulphonamide in acetone.
clearly separated spots.
Reference solution (b). A mixture of equal volumes of the
Heavy metals (2.3.13). 1.0 g complies with the limit test for test solution and reference solution (a).
heavy metals, Method B (20 ppm).
Apply to the plate 5 µl of each of test solution and reference
Sulphated ash (2.3.18). Not more than 0.1 per cent. solution (a) and 10 µl of reference solution (b). After
Loss on drying (2.4.19). Not more than 0.5 per cent, determined development, dry the plate in a current of warm air and heat at
on 1.0 g by drying in an oven at 105º for 3 hours. 110º for 10 minutes. While still hot, place the plate in a
Assay. Weigh accurately about 0.5 g and dissolve in a mixture chromatographic tank with an evaporating dish containing a
of 40 ml of ethanol (95 per cent) and 20 ml of water. Titrate 5 per cent w/v solution of potassium permanganate, add an
with 0.1 M sodium hydroxide using phenolphthalein solution equal volume of hydrochloric acid and close the tank. Leave
as indicator. the plate in the tank for 2 minutes, then place it in a current of
cold air until the excess of chlorine is removed and an area of
1 ml of 0.1 M sodium hydroxide is equivalent to 0.02704 g of coating below the line of application gives only a very faint
C12H18N2O3S. blue colour with potassium iodide and starch solution; avoid
Storage. Store protected from moisture. prolonged exposure to cold air. Spray the plate with potassium
iodide and starch solution and allow to stand for 5 minutes.
Any secondary spot in the chromatogram obtained with the
Tolbutamide Tablets test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a). The
Tolbutamide Tablets contain not less than 95.0 per cent and chromatogram obtained with reference solution (b) shows two
not more than 105.0 per cent of the stated amount of clearly separated spots.
tolbutamide, C12H18N2O3S.
Dissolution (2.5.2).
Identification Apparatus. No 1
Extract a quantity of the powdered tablets containing 1 g of Medium. 900 ml of a solution containing 2.04 per cent w/v of
Tolbutamide with 10 ml of chloroform, filter, evaporate the disodium hydrogen phosphate and 0.135 per cent w/v of
filtrate to dryness, scratching the sides of the vessel, if potassium dihydrogen phosphate.
necessary, to induce crystallisation, and dry the residue at Speed and time. 50 rpm and 30 minutes.
105º for 30 minutes. The residue complies with the following
Withdraw a suitable volume of the medium and filter. Measure
tests.
the absorbance of the filtrate suitably diluted if necessary, at
A. Determine by infrared absorption spectrophotometry (2.4.6). the maximum at about 228 nm (2.4.7). Calculate the content of
Compare the spectrum with that obtained with tolbutamide C12H 18N 2O 3S in the medium taking 417 as the specific
RS or with the reference spectrum of tolbutamide. absorbance at 228 nm.
B. Boil 0.1 g of residue with 8 ml of a 50 per cent v/v solution D. Not less than 70 per cent of the stated amount of
of sulphuric acid under a reflux condenser for 30 minutes. C12H18N2O3S.
Cool in an ice-bath; a crystalline precipitate of
4-toluenesulphonylamide is formed, which after Other tests. Comply with the tests stated under Tablets.
recrystallisation from hot water and drying at 105º melts at Assay. Weigh and powder 20 tablets. Weigh accurately a
135º to 140º. quantity of the powder containing about 0.5 g of Tolbutamide,
Tests add 50 ml of ethanol (95 per cent), previously neutralised to
phenolphthalein solution, warm to dissolve, add 25 ml of
Related substances. Determine by thin-layer chromatography water and titrate with 0.1 M sodium hydroxide using
(2.4.17), coating the plate with silica gel G. phenolphthalein solution as indicator.

1194
IP 2007 TOPOTECAN HYDROCHLORIDE

1 ml of 0.1 M sodium hydroxide is equivalent to 0.02704 g of Weigh accurately about 0.5 g, dissolve in 10 ml of methanol,
C12H18N2O3S. add 20 ml of water and 20 ml of glacial acetic acid. Titrate
Storage. Store protected from moisture. with 0.1 M silver nitrate solution using eosin yellow solution
as indicator. Colour changes from orange to dark pink.
1 ml of 0.1 M silver nitrate is equivalent to 0.003545 g of Cl.

Topotecan Hydrochloride Related substances. Determine by liquid chromatography


(2.4.14).
Solvent mixture. A mixture of 30 volumes of acetonitrile and
CH3 70 volumes of buffer solution prepared by diluting 1 ml of
N trifluoroacetic acid in 1000 ml of water.
CH3
HO Test solution. Dissolve 40 mg of the substance under
O
N examination in 100 ml of solvent mixture.
N , HCl Reference solution (a). A 0.04 per cent w/v solution of
topotecan hydrochloride RS in solvent mixture.
H3C O
Reference solution (b). Dilute 1 ml of the reference solution
HO O (a) to 100 ml with solvent mixture.
Chromatographic system as described under Assay.
C23H23N3O5.HCl Mol. Wt. 457.9 Inject reference solution (a). The test is not valid unless the
Topotecan Hydrochloride is (4S)-10- column efficiency is not less than 2000 theoretical plates and
[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1H- the tailing factor is not more than 2.0.
pyrano[3′,4′:6,7]indolozino[1,2-b]quinoline- Inject the test solution and reference solution (b). In the
3,14(4H,12H)dione hydrochloride. chromatogram obtained with the test solution, the area of any
Topotecan Hydrochloride contains not less than 98.0 per cent secondary peak is not more than 0.15 times the area of the
and not more than 102.0 per cent of C23H23N3O5.HCl, calculated peak in the chromatogram obtained with the reference solution
on the anhydrous basis. (b) (0.15 per cent) and the sum of areas of all the secondary
peaks is not more than the area of the peak in the chromatogram
Description. A light yellow to greenish yellow powder.
obtained with the reference solution (b) (1.0 per cent).
CAUTION – Topotecan Hydrochloride is cytotoxic; extra
Heavy metals (2.3.13). 1.0 g complies with limit test for heavy
care required to prevent inhaling particles and exposing
metals, Method B (20 ppm).
the skin to it.
Test solution. Weigh accurately about 0.5 g of the substance
Identification under examination in 5 ml of dimethylsulphoxide. Pippet a 2
ml of this solution to a 10 ml head space vial,caped cup the
A. Determine by infrared absorption spectrophotometry (2.4.6). vial.
Compare the spectrum with that obtained with topotecan
hydrochloride RS. Reference solution (a). To 30 ml of dimethylsulphoxide, add
300 mg of methanol, 500 mg ethyl acetate, 500 mg isopropyl
B. In the Assay, the principal peak in the chromatogram of the alchohol, 200 mg of acetone, 500 mg of ethanol and 200 mg of
test solution corresponds to the peak in the chromatogram N,N’-dimethyl formamide, diluted to 100 ml with
obtained with the reference solution. dimethylsulphoxide.
Tests Reference solution (b). To 15 ml of dimethylsulphoxide, add
60 mg of chloroform, diluted to 100 ml with dimethylsulphoxide.
Appearance of solution. A 1.0 per cent w/v solution is clear
(2.4.1) and not more intensely coloured than the reference Reference solution (c). Dilute 10 ml each of reference solutions
solution GYS3 (2.4.1). (a) and (b) to 100 ml with dimethylsulphoxide.Pippet 2 ml of
this solution to a head space vial.
pH (2.4.24). 3.5 to 4.5, determined in a 1.0 per cent w/v solution.
Chromatographic system
Specific optical rotation (2.4.22). + 30° to 38°, determined on – a capillary column 30 m x 0.53 mm, packed with mega
1.0 per cent w/v solution in methanol. bore coated with a mixture of 6 per cent cyanopropyl-
Total Chloride. 7.6 per cent to 8.1 per cent. phenyl and 94 per cent dimethylpolysiloxane (3 µm ),

1195
TOPOTECAN INJECTION IP 2007

– temperature: Time Mobile phase A Mobile phase B


column. 35º for 5 minutes increase @ 5º per minute to ( in min) (per cent v/v) (per cent v/v)
150º @ 20º per minute to 220º hold for 2 minutes, 0 85 15
inlet port 180ºand detector. 260º, 28 70 30
Head space conditions
38 70 30
Vial equilibrium temperature 90º, loop temperature 100º , Transfer
line 115º, vial equilibrium 15 minutes, vial pressurisation 0.02 43 85 15
minute, sample loop fill 0.02 minute, loop equilibrium 0.05 minute 48 85 15
sample injection 1 minute, vial pressure 10 psi. Inject the reference solution. The test is not valid unless the
– a flame ionisation detector, relative standard deviation for replicate injections is not more
– nitrogen as carrier gas. than 2.0 per cent.
Inject 1 ml of the reference solution (c). The test is not valid Inject the test solution and reference solution.
unless the resolution between two adjacent peaks is not less
than 1.0. Calculate the content of C23H23N3O5.HCl.

Inject 1 ml of the test solution and reference solution (c). In Storage. Store protected from light, at a temperature between
the chromatogram obtained with test solution, the area of 2° to 8°.
peaks due to methanol, ethyl acetate, isopropyl alcohol,
acetone, ethanol, chloroform and N,N’-dimethyl formamide is
not more than the area of peaks obtained in the chromatogram Topotecan Injection
due to the reference solution (c).
Topotecan Hydrochloride Injection
Loss on ignition (2.4.20). Not more than 0.1 per cent.
Topotecan Injection is a sterile, stabilised solution of
Water (2.3.43). Not more than 5.0 per cent, determined on 0.5 g. Topotecan Hydrochloride in Water for Injection.
Bacterial endotoxins (2.2.3). Not more than 16 Endotoxin Unit Topotecan Injection contains not less than 90.0 per cent and
per mg of topotecan hydrochloride. not more than 110.0 per cent of the stated amount of the
Microbial contamination (2.2.9). Total viable aerobic count, topotecan, C23H23N3O5.
not more than 100 cfu per g. It also meets the requirements of Description. A clear, light yellow solution.
the tests for the absence of Staphylococcus aureus,
Pseudomonas aeruginosa, Salmonella species, and Identification
Escherichia coli.
A. In the Assay, the principal peak in the chromatogram
Assay. Determine by liquid chromatography (2.4.14). obtained with the test solution corresponds to the peak in the
Solvent mixture. A mixture of 30 volumes of acetonitrile and chromatogram obtained with the reference solution.
70 volumes of buffer solution, prepared by diluting 1 ml of B. It gives the reaction of chlorides (2.3.1).
trifluoroacetic acid in 1000 ml of water.
Tests
Test solution. Dissolve 10 mg of the substance under
examination in 25.0 ml of solvent mixture. pH (2.4.24). 2.5 to 3.5.
Reference solution. A 0.04 per cent w/v solution of topotecan Other tests. Complies with the tests stated under Parenteral
hydrochloride RS in solvent mixture. Preparations (Injections).

Chromatographic system Bacterial endotoxins (2.2.3). Not more than 64.9 Endotoxin
– a stainless steel column 25 cm x 4.6 mm, packed with Unit per mg of topotecan.
octadecylsylane bonded to porous silica (5 µm), Sterility (2.2.11). Complies with the test for sterility.
– mobile phase: A. dissolve 1 ml of trifluoroacetic acid Assay. Determine by liquid chromatography (2.4.14).
in 1000 ml of water,
B. acetonitrile, Test solution. Accurately measure the volume of injection
– flow rate. 1.2 ml per minute, containing 2 mg of Topotecan, diluted to 50.0 ml with mobile
– a linear gradient programme using the conditions given phase.
below, Reference solution. Dissolve 10 mg of topotecan RS in 5 ml of
– spectrophotometer set at 260 nm, water and dilute to 25.0 ml with mobile phase. Dilute 5.0 ml of
– a 10 µl loop injector. the solution to 50.0 ml with mobile phase.

1196
IP 2007 TRIAMCINOLONE

Chromatographic system heat on a water-bath under a reflux condenser for 20 minutes;


– a stainless steel column 25 cm x 4.6 mm, packed with a pinkish lavender colour is produced.
octadecylsilane bonded to porous silica
(5 µm), Tests
– mobile phase: a mixture of 78 volumes of water, 22 Specific optical rotation (2.4.22). +65.0º to +72.0º, determined
volumes of acetonitrile and 1 volume of 1 M in a 1.0 per cent w/v solution in dimethylformamide.
hydrochloric acid,
– flow rate. 0.7 ml per minute, Related substances. Determine by liquid chromatography
– spectrophotometer set at 254 nm, (2.4.14).
– a 20 µl loop injector. Prepare the following solutions immediately before use and
Inject the reference solution. The test is not valid unless the protect from light.
relative standard deviation is not more than 2.0 per cent. Test solution. Dissolve 25 mg of the substance under
Inject the test solution and the reference solution. examination in a mixture of equal volumes of methanol and
water and dilute to 10 ml with the same solvent mixture.
Calculate the content of C23H23N3O5.
Reference solution. Dilute 1 ml of the test solution to 100 ml
Storage. Store protected from light, at a temperature not with a mixture of equal volumes of methanol and water.
exceeding 2º to 8º.
Chromatographic system
– a stainless steel column 25 cm x 4.6 mm, packed with
base-deactivated end-capped octadecylsilyl silica gel
Triamcinolone (5 µm),
– mobile phase: a mixture prepared by mixing 525 ml of
methanol with 400 ml of water, allowing to equilibrate,
O OH adjusting the volume to 1000.0 ml with water and mixing
H3C again,
HO OH
OH – flow rate. 1 ml per minute,
H3C H
– spectrophotometer set at 238 nm,
F H – a 20 µl loop injector.
O Inject the test solution and the reference solution. Continue
the chromatography for 4.5 times the retention time of
C21H27FO6 Mol. Wt. 394.4 triamcinolone (about 11 minutes).
Triamcinolone is 9α-fluoro-11β,16α,17α,21- In the chromatogram obtained with the test solution the area
tetrahydroxypregna-1,4-diene-3,20-dione. of any peak other than the principal peak is not more than the
area of the principal peak in the chromatogram obtained with
Triamcinolone contains not less than 97.0 per cent and not
the reference solution (1 per cent); not more than two such
more than 103.0 per cent of C21H27FO6, calculated on the dried
peaks have an area greater than half the area of the principal
basis.
peak in the chromatogram obtained with the reference solution
Description. A white or almost white, crystalline powder; (0.5 per cent); the sum of the areas of all the peaks other than
slightly hygroscopic. the principal peak is not more than twice the area of the principal
peak in the chromatogram obtained with the reference solution
Identification (2 per cent). Ignore any peak with an area less than 0.05 times
the area of the principal peak in the chromatogram obtained
A. Determine by infrared absorption spectrophotometry (2.4.6).
with the reference solution (0.05 per cent).
Compare the spectrum with that obtained with triamcinolone
RS or with the reference spectrum of triamcinolone. Heavy metals (2.3.13). 0.8 g complies with the limit test for
heavy metals, Method B (25 ppm).
B. When examined in the range 220 nm to 360 nm (2.4.7), a
0.002 per cent w/v solution in methanol shows an absorption Sulphated ash (2.3.18). Not more than 0.2 per cent.
maximum at about 238 nm; absorbance at about 238 nm, about Loss on drying (2.4.19). Not more than 2.0 per cent, determined
0.76. on 1.0 g by drying in an oven at 60º at a pressure not exceeding
C. Dissolve 1 mg in 6 ml of ethanol (95 per cent), add 5 ml of 0.7 kPa for 3 hours.
a 1 per cent w/v solution of butylated hydroxytoluene in Assay. Weigh accurately about 25 mg, dissolve in sufficient
ethanol (95 per cent) and 5 ml of 1 M sodium hydroxide and ethanol (95 per cent) to produce 100.0 ml and mix. Dilute

1197
TRIAMCINOLONE TABLETS IP 2007

2.0 ml to 50.0 ml with ethanol (95 per cent) and measure the – spectrophotometer set at 238 nm,
absorbance of the resulting solution at the maximum at about – a 20 µl loop injector.
238 nm (2.4.7). Inject reference solution (b) and record the chromatogram for
Calculate the content of C21H27FO6 taking 380 as the specific 4 times the retention time of the triamcinolone peak.
absorbance at 238 nm.
The test is not valid unless the column efficiency determined
Storage. Store protected from light and moisture. from the principal peak in the chromatogram obtained with
reference solution (a) is at least 10,000 theoretical plates per
metre.

Triamcinolone Tablets In the chromatogram obtained with the test solution the area
of any secondary peak is not greater than twice the area of the
Triamcinolone Tablets contain not less than 90.0 per cent and principal peak in the chromatogram obtained with reference
not more than 110.0 per cent of the stated amount of solution (b) (2 per cent) and not more than one such peak has
triamcinolone, C21H27FO6. an area greater than that of the principal peak in the
chromatogram obtained with reference solution (b) (1 per
Identification cent). The sum of the areas of any such peaks is not greater
A. Dissolve 1 mg of the residue obtained in the test for Related than the area of the principal peak in the chromatogram
substances in 6 ml of ethanol (95 per cent), add 5 ml of a 1 per obtained with reference solution (a) (4 per cent).
cent w/v solution of butylated hydroxytoluene in ethanol Uniformity of content. Comply with the test stated under
(95 per cent) and 5 ml of 1 M sodium hydroxide and heat on Tablets.
a water-bath under a reflux condenser for 20 minutes; a pinkish
Crush one tablet to a fine powder, add 100 ml of ethanol
lavender colour is produced.
(95 per cent) and shake for 10 minutes. Filter, dilute 2.0 ml of
B. In the Assay, the chromatogram obtained with the principal the filtrate with sufficient ethanol (95 per cent) to produce a
peak obtained with the test solution corresponds to the peak solution containing 0.002 per cent w/v of Triamcinolone and
due to triamcinolone in the chromatogram obtained with measure the absorbance of the resulting solution at the
reference solution (a). maximum at about 238 nm (2.4.7).
Tests Calculate the content of C21H27FO6 in the tablet taking 380 as
the specific absorbance at 238 nm.
Related substances. Determine by liquid chromatography
Other tests. Comply with the tests stated under Tablets.
(2.4.14).
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Shake a quantity of the powdered tablets
containing 15 mg of Triamcinolone with 15 ml of ethanol for Test solution. Weigh and powder 20 tablets. Weigh accurately
15 minutes, filter under reduced pressure through a fine filter a quantity of the powder containing 2.5 mg of Triamcinolone,
paper (such as Whatman No 42) and evaporate the filtrate to shake with 5 ml of methanol and 20 ml of a mixture of 5 volumes
dryness using a rotary evaporator. Reserve 1 mg of the residue of methanol and 3 volumes of water. Shake for 15 minutes, mix
for Identification test A. Dissolve the remainder of the residue with the aid of ultrasound for 10 minutes, centrifuge and use
in 15 ml of methanol. the supernatant liquid.
Reference solution (a). Dilute 4 ml of the test solution to Reference solution (a). Prepare in the same manner as the test
100 ml with methanol. solution but add 5 ml of a 0.06 per cent w/v solution of
testosterone (internal standard) in methanol (solution A) in
Reference solution (b). Dilute 5 ml of the test solution to 50 ml
place of the 5 ml of methanol.
with methanol. Dilute 5 ml of the resulting solution to 50 ml
with the same solvent. Reference solution (b). Add 5 ml of solution A to 5 ml of a
0.08 per cent w/v solution of triamcinolone RS in methanol
Chromatographic system
and add 15 ml of methanol (50 per cent).
– a stainless steel column 20 cm x 4.6 mm, packed with
octadecylsilyl silica gel (5 µm), Chromatographic system
– mobile phase: a mixture of methanol and water, adjusted – a stainless steel column 20 cm x 4 mm, packed with
so that the retention time of triamcinolone is about octadecylsilane bonded to porous silica (10 µm),
5 minutes (approximately equal volumes of methanol – mobile phase: a mixture of 70 volumes of methanol,
and water), 30 volumes of water and 0.1 volume of glacial acetic
– flow rate. 2 ml per minute, acid,

1198
IP 2007 TRIAMCINOLONE ACETONIDE

– flow rate. 2 ml per minute, evaporate. Use within 2 hours, with the flow of the mobile
– spectrophotometer set at 238 nm, phase in the direction in which the aforementioned treatment
– a 20 µl loop injector. was done.
Calculate the content of C21H27FO6 in the tablets. Apply to the plate 5 µl of each solution. Allow the mobile
phase to rise 12 cm. Dry the plate in a current of warm air, allow
Storage. Store protected from moisture.
the solvent to evaporate, heat at 120º for 15 minutes and spray
the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120º for a further 10 minutes, allow to cool and examine
Triamcinolone Acetonide in daylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution
O corresponds to that in the chromatogram obtained with
OH
H3C CH3 reference solution (a). The principal spot in the chromatogram
HO O obtained with reference solution (b) appears as a single,
O CH3
H3C H compact spot.

F H C. Heat 0.5 ml of chromic-sulphuric acid in a test-tube (5 cm x


about 6 mm) in a naked flame until white fumes are evolved;
O
the solution wets the sides of the tube readily and there is no
C24H31FO6 Mol. Wt. 434.5 greasiness. Add about 2 mg of the substance under
examination and again heat in a naked flame until white fumes
Triamcinolone Acetonide is 9α-fluoro-11β,21-dihydroxy-
appear; the solution does not wet the sides of the tube and
16α,17α-isopropylidenedioxy-1,4-pregnadiene-3,20-dione.
does not pour easily from the tube.
Triamcinolone Acetonide contains not less than 96.0 per cent
and not more than 104.0 per cent of C24H31FO6, calculated on Tests
the anhydrous basis.
Light absorption (2.4.7). When examined in the range 230 nm
Description. A white or almost white, crystalline powder; to 360 nm, a 0.001 per cent w/v solution in ethanol (95 per
odourless or almost odourless. cent) shows an absorption maximum at about 239 nm;
absorbance at about 239 nm, 0.34 to 0.37.
Identification
Specific optical rotation (2,4,22). +100º to +107º, determined
Test A may be omitted if tests B and C are carried out. Test C in a 1.0 per cent w/v solution in dioxan.
may be omitted if tests A and B are carried out. Related substances. Determine by liquid chromatography
A. Determine by infrared absorption spectrophotometry (2.4.6). (2.4.14).
Compare the spectrum with that obtained with triamcinolone Carry out the test protected from light.
acetonide RS.
Test solution. Dissolve 25 mg of the substance under
B. Determine by thin-layer chromatography (2.4.17), coating examination in 7 ml of methanol and dilute to 10 ml with water.
the plate with silica gel G.
Reference solution (a). Dissolve 2 mg of triamcinolone
Solvent mixture. A mixture of 90 volumes of acetone and acetonide RS and 2 mg of triamcinolone in the mobile phase
10 volumes of formamide. and dilute to 100 ml with the mobile phase.
Mobile phase. A mixture of 115 volumes of cyclohexane, Reference solution (b). Dilute 1 ml of the test solution to
56 volumes of chloroform and 29 volumes of toluene. 100 ml with the mobile phase.
Test solution. Dissolve 25 mg of the substance under Chromatographic system
examination in 10 ml of the solvent mixture. – a stainless steel column 25 cm x 4.6 mm, packed with
Reference solution (a). Dissolve 25 mg of triamcinolone octadecylsilyl silica gel (5 µm),
acetonide RS in 10 ml of the solvent mixture. – mobile phase: a mixture prepared by mixing 525 ml of
methanol with 400 ml of water, allowing to equilibrate,
Reference solution (b). Mix equal volumes of the test solution adjusting the volume to 1000.0 ml with water and mixing
and reference solution (a). again,
Place the dry plate in a tank containing a shallow layer of the – flow rate. 1.5 ml per minute,
solvent mixture, allow the solvent mixture to ascend to the – spectrophotometer set at 254 nm,
top, remove the plate from the tank and allow the solvent to – a 20 µl loop injector.

1199
TRIAMCINOLONE ACETONIDE INJECTION IP 2007

Equilibrate the column with the mobile phase for about 10 through a sintered-glass filter, wash the residue with four
minutes. quantities, each of 5 ml of water and dry at 105º for 1 hour. The
Inject reference solution (b). Adjust the sensitivity of the residue complies with the following tests.
system so that the height of the principal peak is at least Test A may be omitted if tests B and C are carried out. Test C
50 per cent of the full scale of the recorder. may be omitted if tests A and B are carried out.
Inject reference solution (a). The retention times are; A. Determine by infrared absorption spectrophotometry (2.4.6).
triamcinolone, about 5 minutes and triamcinolone acetonide Compare the spectrum with that obtained with triamcinolone
about 17 minutes. The test is not valid unless the resolution acetonide RS or with the reference spectrum of triamcinolone
between the peaks corresponding to triamcinolone and acetonide.
triamcinolone acetonide is at least 15; if necessary, adjust the
B. Determine by thin-layer chromatography (2.4.17), coating
concentration of methanol in the mobile phase.
the plate with silica gel G.
Inject the test solution and reference solution (b). Continue
the chromatography for 3.5 times the retention time of the Solvent mixture. A mixture of 90 volumes of acetone and
principal peak in the chromatogram obtained with the test 10 volumes of formamide.
solution. In the chromatogram obtained with the test solution Mobile phase. A mixture of 115 volumes of cyclohexane,
the area of any peak other than the principal peak is not more 56 volumes of chloroform and 29 volumes of toluene.
than 0.25 times the area of the principal peak in the
Test solution. Dissolve 25 mg of the substance under
chromatogram obtained with the reference solution (b) (0.25
examination in 10 ml of the solvent mixture.
per cent); the sum of the areas of all the peaks other than the
principal peak is not greater than half the area of the principal Reference solution (a). Dissolve 25 mg of triamcinolone
peak in the chromatogram obtained with reference solution acetonide RS in 10 ml of the solvent mixture.
(b) (0.5 per cent). Ignore any peak due to the solvent and any Reference solution (b). Mix equal volumes of the test solution
peak with an area less than 0.05 times the area of the principal and reference solution (a).
peak in the chromatogram obtained with reference solution
(b). Place the dry plate in a tank containing a shallow layer of the
solvent mixture, allow the solvent mixture to ascend to the
Sulphated ash (2.3.18). Not more than 0.2 per cent. top, remove the plate from the tank and allow the solvent to
Water (2.3.43). Not more than 2.0 per cent, determined on 0.2 g. evaporate. Use within 2 hours, with the flow of the mobile
Assay. Weigh accurately about 25 mg and dissolve in sufficient phase in the direction in which the aforementioned treatment
ethanol to produce 100.0 ml and mix. Dilute 5.0 ml to 100.0 ml was done.
with ethanol (95 per cent) and measure the absorbance of Apply to the plate 5 µl of each solution. Allow the mobile
the resulting solution at the maximum at about 239 nm (2.4.7). phase to rise 12 cm. Dry the plate in a current of warm air, allow
Calculate the content of C24H31FO6 taking 354 as the specific the solvent to evaporate, heat at 120º for 15 minutes and spray
absorbance at 239 nm. the hot plate with ethanolic sulphuric acid (20 per cent v/v).
Heat at 120º for a further 10 minutes, allow to cool and examine
Storage. Store protected from light and moisture.
in daylight and in ultraviolet light at 365 nm. The principal
spot in the chromatogram obtained with the test solution
corresponds to that in the chromatogram obtained with
Triamcinolone Acetonide Injection reference solution (a). The principal spot in the chromatogram
obtained with reference solution (b) appears as a single,
Triamcinolone Acetonide Injection is a sterile suspension of compact spot.
Triamcinolone Acetonide in very fine particles in Water for
Injections containing suitable dispersing agents. C. Heat 0.5 ml of chromic-sulphuric acid in a test-tube (5 cm x
about 6 mm) in a naked flame until white fumes are evolved;
Triamcinolone Acetonide Injection contains not less than the solution wets the sides of the tube readily and there is no
90.0 per cent and not more than 110.0 per cent of the stated greasiness. Add about 2 mg of the substance under
amount of triamcinolone acetonide, C24H31FO6. examination and again heat in a naked flame until white fumes
appear; the solution does not wet the sides of the tube and
Identification does not pour easily from the tube.
Extract a volume containing about 50 mg of Triamcinolone
Tests
Acetonide with two quantities, each of 10 ml, of peroxide-free
ether and discard the ether extracts. Filter the aqueous layer pH (2.4.24). 5.0 to 7.5.

1200
IP 2007 TRIAMTERENE

Other tests. Complies with the tests stated under Parenteral absorption maxima at about 262 nm and 360 nm, and a shoulder
Preparations (Injections). at about 285 nm.
Assay. Determine by liquid chromatography (2.4.14). B. A 0.1 per cent w/v solution in anhydrous formic acid, when
examined in ultraviolet light at 365 nm shows an intense blue
Test solution. Mix an accurately measured volume of the
fluorescence. Solutions in other acids also exhibit a blue
injection, diluted with methanol, with sufficient of a solution
fluorescence.
of prednisolone RS (internal standard) with methanol to obtain
a final concentration of 0.02 per cent w/v of triamcinolone
acetonide and 0.01 per cent w/v of prednisolone, centrifuge
Tests
and use the clear supernatant liquid. Acidity. Boil 1.0 g with 20 ml of water for 5 minutes, cool, filter
Reference solution (a). A solution containing 0.02 per cent and wash the filter with three quantities, each of 10 ml, of
w/v of triamcinolone acetonide RS and 0.01 per cent w/v of water. Combine the filtrate and washings and add 0.3 ml of
prednisolone RS in methanol. phenolphthalein solution. Not more than 1.5 ml of 0.01 M
sodium hydroxide is required to change the colour of the
Reference solution (b). Prepare in the same manner as test
solution.
solution but omitting the internal standard.
Related substances. Determine by thin-layer chromatography
Chromatographic system
(2.4.17), coating the plate with silica gel G.
– a stainless steel column 25 cm x 4 mm, packed with
octadecylsilane bonded to porous silica (10 µm), Mobile phase. A mixture of 90 volumes of ethyl acetate,
– mobile phase: a mixture of 56 volumes of methanol and 10 volumes of 18 M ammonia and 10 volumes of methanol.
44 volumes of water,
Test solution. Dissolve 0.1 g of the substance under
– flow rate. 1.4 ml per minute,
examination in 20 ml of dimethyl sulphoxide and dilute 2 ml of
– spectrophotometer set at 254 nm,
the resulting solution to 50 ml with methanol.
– a 20 µl loop injector.
Adjust the flow rate of the mobile phase such that the Reference solution. Dilute 1 volume of the test solution to
separation of the triamcinolone acetonide and internal standard 200 volumes with methanol.
is optimised with a retention time of about 15 minutes for Apply to the plate 5 µl of each solution. After development,
triamcinolone acetonide. dry the plate in air until the odour of solvent is no longer
Calculate the content of C24H31FO6 in the injection. detectable and examine in ultraviolet light at 365 nm. Any
secondary spot in the chromatogram obtained with the test
Storage. Store protected from light. solution is not more intense than the spot in the chromatogram
obtained with the reference solution.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Triamterene Loss on drying (2.4.19). Not more than 1.0 per cent, determined
H2N N N NH2 on 1.0 g by drying in an oven at 105º.
Assay. Weigh accurately about 0.15 g, dissolve in 5 ml of
N anhydrous formic acid, add 100 ml of anhydrous glacial
N
acetic acid. Titrate with 0.1 M perchloric acid, determining
NH2
the end-point potentiometrically (2.4.25). Carry out a blank
C12H11N7 Mol. Wt. 253.3 titration.
Triamterene is 2,4,7-triamino-6-phenylpteridine. 1 ml of 0.1 M perchloric acid is equivalent to 0.02533 g of
C12H11N7.
Triamterene contains not less than 99.0 per cent and not more
than 101.0 per cent of C12H11N7, calculated on the dried basis. Storage. Store protected from light and moisture.
Description. A yellow, crystalline powder; odourless.

Identification Triamterene Capsules


A. When examined in the range 250 nm to 380 nm (2.4.6), a Triamterene Capsules contain not less than 95.0 per cent and
0.001 per cent w/v solution in a mixture of 9 volumes of ethanol not more than 105.0 per cent of the stated amount of
(95 per cent) and 1 volume of 1 M hydrochloric acid shows triamterene, C12H11N7.

1201
TRIFLUOPERAZINE HYDROCHLORIDE IP 2007

Identification Trifluoperazine Hydrochloride contains not less than 99.0 per


cent and not more than 101.0 per cent of C21H24F3N3S, 2HCl,
The final solution obtained in the Assay has a bluish calculated on the dried basis.
fluorescence and when examined in the range 250 nm to
380 nm (2.4.7), shows an absorption maximum at about Description. A white to pale yellow, crystalline powder;
360 nm. odourless or almost odourless; slightly hygroscopic.
NOTE — In the following procedures, the test and standard
Tests
specimens and the solutions containing them should be
Related substances. Determine by thin-layer chromatography protected by carrying out the tests without delay and in
(2.4.17), coating the plate with silica gel G. subdued light.
Mobile phase. A mixture of 90 volumes of ethyl acetate,
10 volumes of 18 M ammonia and 10 volumes of methanol. Identification
Test solution. Dissolve a quantity of the contents of the A. Dissolve 20 mg in 10 ml of water, make the solution alkaline
capsules containing 0.1 g of Triamterene in sufficient dimethyl to litmus paper with 5 M sodium hydroxide and extract with
sulphoxide to produce 20 ml and dilute 2 ml to 50 ml with two quantities, each of 20 ml, of light petroleum (60º to 80º).
methanol. Combine the extracts, wash with 10 ml of water, shake with 5 g
Reference solution. Dilute 1 volume of the test solution to of anhydrous sodium sulphate, filter and evaporate the filtrate
200 volumes with methanol. carefully to dryness.
Apply to the plate 5 µl of each solution. After development, On the residue, determine by infrared absorption
dry the plate in air until the odour of solvent is no longer spectrophotometry (2.4.6). Compare the spectrum with that
detectable and examine in ultraviolet light at 365 nm. Any obtained with trifluoperazine hydrochloride RS or with the
secondary spot in the chromatogram obtained with the test reference spectrum of trifluoperazine hydrochloride.
solution is not more intense than the spot in the chromatogram B. Complies with the test for identification of phenothiazines
obtained with the reference solution. (2.3.3), using as reference solution a 0.2 per cent w/v solution
Other tests. Comply with the tests stated under Capsules. of trifluoperazine hydrochloride RS in chloroform.
Assay. Weigh accurately a quantity of the contents of C. When examined in the range 280 nm to 360 nm (2.4.7), a
20 capsules containing about 0.1 g of Triamterene, dissolve in 0.01 per cent w/v solution in 0.1 M hydrochloric acid
50 ml of a mixture of equal volumes of glacial acetic acid and measured immediately after preparation shows an absorption
water with the aid of gentle heat, cool and add sufficient maximum at about 305 nm. Dilute 10 ml of the solution to
water to produce 500.0 ml. Dilute 5.0 ml of this solution to 100 ml with 0.1 M hydrochloric acid. When examined in the
100.0 ml with 1 M acetic acid and measure the absorbance of range 230 nm to 280 nm, the resulting solution shows an
the resulting solution at the maximum at about 360 nm (2.4.7). absorption maximum at about 256 nm; absorbance at about
Calculate the content of C 12H11N7 from the absorbance 256 nm, about 0.65.
obtained by repeating the operation using triamterene RS in
D. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand
place of the contents of the capsules.
for 5 minutes; an orange colour is produced.
Storage. Store protected from moisture.
E. A 10 per cent w/v solution gives the reactions of chlorides
(2.3.1).
Trifluoperazine Hydrochloride Tests
pH (2.4.24). 1.7 to 2.6, determined in a 10.0 per cent w/v solution.
Related substances. Complies with the test for related
N N substances in phenothiazines (2.3.5), using mobile phase A.
S N , 2HCl
CH3 Sulphated ash (2.3.18). Not more than 0.1 per cent.

CF3 Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105º at a pressure not exceeding
C21H24F3N3S,2HCl Mol. Wt. 480.4 0.7 kPa.
Trifluoperazine hydrochloride is 10-[3-(4-methylpiperazin-1- Assay. Weigh accurately about 0.4 g, dissolve in 50 ml of
yl)propyl]-2-trifluoromethylphenothiazine dihydrochloride. anhydrous glacial acetic acid add 10 ml of mercuric acetate

1202
IP 2007 TRIFLUOPERAZINE TABLETS

solution. Titrate with 0.1 M perchloric acid, using crystal Trifluoperazine Tablets
violet solution as indicator. Carry out a blank titration.
Trifluoperazine Hydrochloride Tablets
1 ml of 0.1 M perchloric acid is equivalent to 0.02402 g of
C21H24F3N3S, 2HCl. Trifluoperazine Tablets contain not less than 92.5 per cent
and not more than 107.5 per cent of trifluoperazine,
Storage. Store protected from light and moisture.
C21H24F3N3S. The tablets are coated.
NOTE — In the following procedures, the test and standard
specimens and the solutions containing them should be
protected by carrying out the tests without delay and in
Trifluoperazine Injection subdued light.
Trifluoperazine Hydrochloride Injection Identification
Trifluoperazine Injection is a sterile solution of Trifluoperazine
A. Shake a quantity of the powdered tablets containing 20 mg
Hydrochloride in Water for Injections.
of trifluoperazine with 30 ml of 1 M hydrochloric acid for
Trifluoperazine Injection contains not less than 90.0 per cent 10 minutes, filter, make the filtrate alkaline to litmus paper with
and not more than 110.0 per cent of the stated amount of 5 M sodium hydroxide and extract with two quantities, each
trifluoperazine, C21H24F3N3S. of 20 ml, of light petroleum (60º to 80º). Combine the extracts,
Description. A clear, colourless solution. wash with 10 ml of water, shake with 5 g of anhydrous sodium
sulphate, filter and evaporate the filtrate carefully to dryness.
NOTE — In the following procedures, the test and standard
specimens and the solutions containing them should be On the residue, determine by infrared absorption
protected by carrying out the tests without delay and in spectrophotometry (2.4.6). Compare the spectrum with that
subdued light. obtained with trifluoperazine hydrochloride RS or with the
reference spectrum of trifluoperazine hydrochloride.
Identification B. Extract a quantity of the powdered tablets containing 5 mg
of trifluoperazine with 5 ml of acetone, filter and evaporate the
When examined in the range 230 nm to 360 nm (2.4.7), the final filtrate to dryness. Add 2 ml of sulphuric acid to the residue
solution obtained in Assay shows an absorption maximum at and allow to stand for 5 minutes; an orange colour is produced.
about 256 nm.
Tests
Tests
Uniformity of content. Comply with the test stated under
pH (2.4.24). 4.0 to 5.0. Tablets.
Other tests. Complies with the tests stated under Parenteral Place one tablet in a 100-ml volumetric flask, add 50 ml of a
preparations (Injections). mixture of 19 volumes of water and 1 volume of hydrochloric
acid, shake until the tablet has completely disintegrated, dilute
Assay. To an accurately measured volume of the injection
to volume with the same mixture, mix and filter, rejecting the
containing about 50 mg of trifluoperazine add 10 ml of 2 M
first few ml of the filtrate. Dilute suitably, if necessary with the
sulphuric acid and extract with three quantities, each of 25 ml,
same solvent mixture and measure the absorbance of the
of carbon tetrachloride. Discard the carbon tetrachloride
resulting solution at the maximum at about 256 nm (2.4.7).
extract after each extraction. Add 10 ml of strong ammonia
Calculate the content of C21H24F3N3S taking 743 as the specific
solution and extract with five quantities, each of 50 ml, of
absorbance at 256 nm.
cyclohexane. Extract the combined cyclohexane extracts with
five quantities, each of 50 ml, of 0.1 M hydrochloric acid. Other tests. Comply with the tests stated under Tablets.
Dilute the combined acid extracts to 500.0 ml and mix. Measure Assay. Weigh and powder 20 tablets. Weigh accurately a
the absorbance of the resulting solution at the maximum at quantity of the powder containing about 5 mg of trifluoperazine,
about 256 nm (2.4.7). shake for 15 minutes with 400 ml of a mixture of 19 volumes of
Calculate the content of C21H24F3N3S taking 743 as the specific water and 1 volume of hydrochloric acid, dilute to 500.0 ml
absorbance at 256 nm. with the same solvent mixture, mix and filter. Measure the
absorbance of the filtrate at the maximum at about 256 nm
Storage. Store protected from light. (2.4.7).
Labelling. The label states the strength in terms of the Calculate the content of C21H24F3N3S taking 743 as the specific
equivalent amount of trifluoperazine per ml. absorbance at 256 nm.

1203
TRIFLUPROMAZINE HYDROCHLORIDE IP 2007

Storage. Store protected from light and moisture. D. A 10 per cent w/v solution gives the reactions of chlorides
Labelling. The label states the strength in terms of the (2.3.1).
equivalent amount of trifluoperazine. Tests
Related substances. Complies with the test for related
substances in phenothiazines (2.3.5), using mobile phase A.
Triflupromazine Hydrochloride Sulphated ash (2.3.18). Not more than 0.1 per cent.
Flupromazine Hydrochloride Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105º for 2 hours.
Assay. Weigh accurately about 0.8 g, dissolve in 50 ml of
CH3 anhydrous glacial acetic acid add 10 ml of mercuric acetate
N N
, HCl solution. Titrate with 0.1 M perchloric acid, using crystal
S CH3 violet solution as indicator. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.03889 g of
CF3 C18H19F3N2S, HCl.

C18H19F3N2S, HCl Mol. Wt. 388.9 Storage. Store protected from light and moisture.

Triflupromazine Hydrochloride is 10-[3-


(dimethylamino)propyl)]-2-trifluoromethylphenothiazine
hydrochloride. Triflupromazine Injection
Triflupromazine Hydrochloride contains not less than Triflupromazine Hydrochloride Injection; Flupromazine
97.0 per cent and not more than 103.0 per cent of C18H19F3N2S, Hydrochloride Injection; Flupromazine Injection
HCl, calculated on the dried basis.
Triflupromazine Injection is a sterile solution of Triflupromazine
Description. A white to pale yellowish brown, crystalline Hydrochloride in Water for Injections.
powder; odour slight and characteristic.
Triflupromazine Injection contains not less than 90.0 per cent
NOTE — In the following procedures, the test and standard and not more than 110.0 per cent of the stated amount of
specimens and the solutions containing them should be triflupromazine hydrochloride, C18H19F3N2S, HCl.
protected by carrying out the tests without delay and in
subdued light. Description. A clear, almost colourless solution.
NOTE — In the following procedures, the test and standard
Identification specimens and the solutions containing them should be
A. Dissolve 20 mg in 10 ml of water, make the solution alkaline protected by carrying out the tests without delay and in
to litmus paper with 5 M sodium hydroxide and extract with subdued light.
two quantities, each of 20 ml, of light petroleum (60º to 80º).
Identification
Combine the extracts, wash with 10 ml of water, shake with 5 g
of anhydrous sodium sulphate, filter and evaporate the filtrate A. To an appropriate quantity of the injection add sufficient
carefully to dryness. 0.1 M hydrochloric acid to produce a solution containing
On the residue, determine by infrared absorption 0.001 per cent w/v solution of Triflupromazine Hydrochloride.
spectrophotometry (2.4.6). Compare the spectrum with that When examined in the range 230 nm to 360 nm (2.4.7), the
obtained with triflupromazine hydrochloride RS or with the resulting solution shows an absorption maximum at about
reference spectrum of triflupromazine hydrochloride. 256 nm.
B. Complies with the test for identification of phenothiazines B. To a volume containing about 100 mg of Triflupromazine
(2.3.3), using as reference solution a 0.2 per cent w/v solution Hydrochloride add 5 ml of 8 M nitric acid and mix; a pink to
of triflupromazine hydrochloride RS in chloroform. amber colour develops which quickly turns dark brown and
C. When examined in the range 230 nm to 360 nm (2.4.7), a then changes to a clear solution having a yellow tint.
0.001 per cent w/v solution in 0.1 M hydrochloric acid shows
Tests
an absorption maximum at about 256 nm and a less well-defined
maximum at about 305 nm. pH (2.4.24). 3.5 to 5.2.

1204
IP 2007 TRIMETHOPRIM

Other tests. Complies with the tests stated under Parenteral Comply with the test stated under Tablets.
Preparations (Injections). Crush one tablet and transfer to a 100-ml volumetric flask with
Assay. To an accurately measured volume of the injection the aid of a mixture of 19 volumes of water and 1 volume of
containing about 20 mg of Triflupromazine Hydrochloride add hydrochloric acid. Shake well, dilute to volume with the same
sufficient of a mixture of 19 volumes of water and 1 volume of mixture and complete the procedure described under the Assay
hydrochloric acid to produce 100.0 ml and mix. Dilute 5.0 ml beginning at the words “mix and filter,...”.
of the solution to 100.0 ml with the same mixture and mix.
Calculate the content of C18H19F3N2S, HCl in the tablet.
Measure the absorbance of the resulting solution at the
maximum at about 256 nm (2.4.7). Other tests. Comply with the tests stated under Tablets.
Calculate the content of C18H19F3N2S, HCl taking 700 as the Assay. Weigh and powder 20 tablets. Weigh accurately a
specific absorbance at 256 nm. quantity of the powder containing about 50 mg of
Triflupromazine Hydrochloride and shake for 15 minutes with
Storage. Store protected from light. 200 ml of a mixture of 19 volumes of water and 1 volume of
hydrochloric acid. Dilute to 500.0 ml with the same solvent
mixture, mix and filter, rejecting the first few ml of the filtrate.
Triflupromazine Tablets Dilute 10.0 ml to 100.0 ml with the same solvent mixture.
Measure the absorbance of the filtrate at the maximum at about
Triflupromazine Hydrochloride Tablets; Flupromazine 256 nm (2.4.7).
Hydrochloride Tablets; Flupromazine Tablets Calculate the content of C18H19F3N2S, HCl taking 700 as the
Triflupromazine Tablets contain not less than 90.0 per cent specific absorbance at 256 nm.
and not more than 110.0 per cent of the stated amount of Storage. Store protected from light and moisture.
triflupromazine hydrochloride, C18H19F3N2S, HCl.
NOTE — In the following procedures, the test and standard
specimens and the solutions containing them should be
protected by carrying out the tests without delay and in Trimethoprim
subdued light.
NH2
Identification OCH3
N
A. Shake a quantity of the powdered tablets containing 20 mg
of Triflupromazine Hydrochloride with 30 ml of 1 M H2 N N OCH3
hydrochloric acid for 10 minutes, filter, make the filtrate alkaline OCH3
to litmus paper with 5 M sodium hydroxide and extract with
two quantities, each of 20 ml, of light petroleum (boiling C14H18N4O3 Mol. Wt. 290.3
range 60º to 80º). Combine the extracts, wash with 10 ml of Trimethoprim is 5-(3,4,5-trimethoxybenzyl)pyrimidine-2,4-
water, shake with 5 g of anhydrous sodium sulphate, filter and diamine.
evaporate the filtrate carefully to dryness.
Trimethoprim contains not less than 98.5 per cent and not
On the residue, determine by infrared absorption more than 101.0 per cent of C14H18N4O3, calculated on the
spectrophotometry (2.4.6). Compare the spectrum with that dried basis.
obtained with triflupromazine hydrochloride RS or with the
Description. A white or yellowish white powder; odourless or
reference spectrum of triflupromazine hydrochloride.
almost odourless.
B. Extract a quantity of the powdered tablets containing 5 mg
of Triflupromazine Hydrochloride with 5 ml of acetone, filter Identification
and evaporate the filtrate to dryness. Test A may be omitted if tests B and C are carried out. Tests B
When examined in the range 230 nm to 360 nm (2.4.7), a and C may be omitted if test A is carried out.
0.001 per cent w/v solution in 0.1 M hydrochloric acid shows A. Determine by infrared absorption spectrophotometry (2.4.6).
an absorption maximum at about 256 nm. Compare the spectrum with that obtained with trimethoprim
RS or with the reference spectrum of trimethoprim.
Tests
B. Dissolve 25 mg in 25 ml of ethanol (95 per cent) and dilute
Uniformity of content. For tablets containing 10 mg or less. 2.0 ml to 100 ml with 0.1 M sodium hydroxide.

1205
TRIMETHOPRIM AND SULPHAMETHOXAZOLE ORAL SUSPENSION IP 2007

When examined in the range 230 nm to 360 nm (2.4.7), the Assay. Weigh accurately about 0.25 g, dissolve in 50 ml of
resulting solution shows an absorption maximum at about anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
287 nm; absorbance at about 287 nm, about 0.49. acid, determining the end-point potentiometrically (2.4.25).
C. Dissolve about 25 mg in 5 ml of 0.005 M sulphuric acid, Carry out a blank titration.
with heating if necessary, add 2 ml of a 1.6 per cent w/v solution 1 ml of 0.1 M perchloric acid is equivalent to 0.02903 g of
of potassium permanganate in 0.1 M sodium hydroxide. Heat C14H18N4O3.
to boiling and to the hot solution add 0.4 ml of formaldehyde
Storage. Store protected from light.
solution. Mix, add 1 ml of 0.5 M sulphuric acid, mix and heat
to boiling. Cool and filter. Add 2 ml of chloroform to the filtrate
and shake vigorously. The chloroform layer exhibits a green
fluorescence when examined in ultraviolet light at 365 nm. Trimethoprim and Sulphamethoxazole
Tests Oral Suspension
Appearance of solution. A 5.0 per cent w/v solution in a mixture Sulphamethoxazole and Trimethoprim Oral Suspension;
of 10 volumes of chloroform, 9 volumes of methanol and Co-trimoxazole Oral Suspension; Co-trimoxazole Mixture
2 volumes of water is not more intensely coloured than
Trimethoprim and Sulphamethoxazole Oral Suspension is a
reference solution BYS7 (2.4.1).
suspension of Trimethoprim and Sulphamethoxazole in a
Related substances. Determine by thin-layer chromatography suitable flavoured vehicle. It contains 5 parts of
(2.4.17), coating the plate with silica gel GF254. Sulphamethoxazole for 1 part, by weight, of Trimethoprim.
Mobile phase. A mixture of 85 volumes of ethyl acetate, Trimethoprim and Sulphamethoxazole Oral Suspension
10 volumes of methanol, 5 volumes of water and 2 volumes of contains not less than 90.0 per cent and not more than
anhydrous formic acid. 110.0 per cent of the stated amounts of trimethoprim,
Test solution. Dissolve 0.4 g of the substance under C14H18N4O3, and sulphamethoxazole, C10H11N3O3S.
examination in 10 ml of a mixture of 10 volumes of chloroform,
9 volumes of methanol and 2 volumes of water. Identification
Reference solution. A 0.008 per cent w/v solution of the Determine by thin-layer chromatography (2.4.17), coating the
substance under examination in a mixture of 10 volumes of plate with silica gel G.
chloroform, 9 volumes of methanol and 2 volumes of water.
Mobile phase. A mixture of 20 volumes of chloroform,
Apply to the plate 5 µl of each solution. Allow the mobile 2 volumes of methanol and 1 volume of dimethylformamide.
phase to rise 17 cm. Dry the plate in a current of cold air for
Test solution. Add 20 ml of methanol (or a suitable volume of
5 minutes and examine in ultraviolet light at 254 nm. Place an
methanol to yield 0.4 per cent w/v solution of Trimethoprim)
evaporating dish containing a mixture of 2 volumes of a
to 5 ml of the suspension, mix, shake with 10 g of anhydrous
1.5 per cent w/v solution of potassium permanganate,
sodium sulphate, centrifuge and use the supernatant liquid.
1 volume of 7 M hydrochloric acid and 1 volume of water at
the bottom of a closed tank and allow to stand for 15 minutes. Reference solution (a). A 2.0 per cent w/v solution of
Place the dried plate in the closed tank and expose to the sulphamethoxazole RS in methanol.
chlorine vapour for 5 minutes. Remove the plate from the tank
Reference solution (b). A 0.4 per cent w/v solution of
and remove the chlorine in a current of cold air until an area
trimethoprim RS in methanol.
below the line of application does not give a blue colour on
the addition of 0.05 ml of potassium iodide and starch Apply to the plate 5 µl of each solution. After development,
solution. Spray the plate with potassium iodide and starch dry the plate in air, spray with dilute potassium
solution and examine in daylight. By both methods of iodobismuthate solution. One of the principal spots in the
visualisation any secondary spot in the chromatogram chromatogram obtained with the test solution corresponds to
obtained with the test solution is not more intense than the that in the chromatogram obtained with reference solution (a)
spot in the chromatogram obtained with the reference solution. and the other corresponds to that in the chromatogram
obtained with reference solution (b)
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm). Tests
Sulphated ash (2.3.18). Not more than 0.1 per cent.
pH (2.4.24). 5.0 to 6.5.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105º. Other tests. Complies with the tests stated under Oral liquids.

1206
IP 2007 TRIMETHOPRIM AND SULPHAMETHOXAZOLE TABLETS

Assay. For trimethoprim — Weigh accurately about 4 g, add Trimethoprim And Sulphamethoxazole
30 ml of 0.1 M sodium hydroxide, shake and extract with four
quantities, each of 50 ml, of chloroform, washing each extract Tablet
with the same two quantities, each of 10 ml, of 0.1 M sodium Sulphamethoxazole and Trimethoprim Tablets; Co-
hydroxide. Reserve the combined aqueous solution and trimoxazole Tablets
washings for the Assay for sulphamethoxazole. Extract the
combined chloroform extracts with four quantities, each of Trimethoprim and Sulphamethoxazole Tablets contain 5 parts
50 ml, of 1 M acetic acid. Wash the combined extracts with of Sulphamethoxazole for 1 part, by weight, of Trimethoprim.
5 ml of chloroform and dilute the aqueous extracts to 250.0 ml Trimethoprim and Sulphamethoxazole Tablets contain not less
with 1 M acetic acid. To 10.0 ml of this solution add 10 ml of than 92.5 per cent and not more than 107.5 per cent of the
1 M acetic acid and sufficient water to produce 100.0 ml, mix stated amounts of trimethoprim, C 14 H 18 N 4 O 3, and
and measure the absorbance of the resulting solution at the sulphamethoxazole, C10H11N3O3S.
maximum at about 271 nm (2.4.7).
Identification
Calculate the content of C14H18N4O3 taking 204 as the specific
absorbance at 271 nm. Determine the weight per ml of the A. To a quantity of the powdered tablets containing 50 mg of
suspension (2.4.29), and calculate the content of C14H18N4O3, Trimethoprim add 30 ml of 0.1 M sodium hydroxide and extract
weight in volume. with two quantities, each of 50 ml, of chloroform. Wash the
combined chloroform extracts with two quantities, each of
For sulphamethoxazole — Carry out the following procedure
10 ml, of 0.1 M sodium hydroxide and then with 10 ml of
protected from light.
water. Combine the aqueous extract and washings (solution
Dilute the combined aqueous solution reserved in the Assay A) and reserve for test B. Shake with 5 g of anhydrous sodium
for trimethoprim to 250.0 ml with water, filter and dilute 5.0 ml sulphate, filter and evaporate to dryness using a rotary
of the filtrate to 200.0 ml with water (solution A). To 2.0 ml of evaporator.
solution A add 0.5 ml of 4 M hydrochloric acid and 1 ml of a On the residue, determine by infrared absorption
0.1 per cent w/v solution of sodium nitrite and allow to stand spectrophotometry (2.4.6). Compare the spectrum with that
for 2 minutes. Add 1 ml of a 0.5 per cent w/v solution of obtained with trimethoprim RS or with the reference spectrum
ammonium sulphamate and allow to stand for 3 minutes. Add of trimethoprim.
1 ml of a 0.1 per cent w/v solution of N- (1-napthyl)
ethylenediamine dihydrochloride and allow to stand for B. Filter solution A, add, dropwise, sufficient 2 M hydrochloric
10 minutes. Dilute the solution to 25.0 ml with water and acid to the filtrate to make it just acidic and extract with 50 ml
measure the absorbance of the resulting solution at about of ether. Wash the ether layer with 10 ml of water, shake with
538 nm (2.4.7), using as the blank a solution prepared in the 5 g of anhydrous sodium sulphate, filter and evaporate the
same manner but using 2 ml of water in place of solution A. filtrate to dryness using a rotary evaporator. Dissolve the
Weigh accurately about 0.25 g of sulphamethoxazole RS, residue in the minimum volume of a 5 per cent w/v solution of
dissolve in 50 ml of 0.1 M sodium hydroxide and dilute to sodium carbonate, add 1 M hydrochloric acid dropwise until
250.0 ml with water. Dilute 5.0 ml of the resulting solution to precipitation is complete and filter. Wash the residue sparingly
200.0 ml with water (solution B). Repeat the procedure using with water and dry at 105º.
2.0 ml of solution B and beginning at the words “add 0.5 ml of On the residue, determine by infrared absorption
4 M hydrochloric acid......”. spectrophotometry (2.4.6). Compare the spectrum with that
obtained with sulphamethoxazole RS or with the reference
Calculate the content of C10H11N3O3S from the values of the
spectrum of sulphamethoxazole.
absorbances obtained. Calculate the content of C10H11N3O3S,
weight in volume from the weight per ml determined in the C. Determine by thin-layer chromatography (2.4.17), coating
Assay for trimethoprim. the plate with silica gel G.
Storage. Store protected from light and moisture. The Mobile phase. A mixture of 20 volumes of chloroform,
suspension should not be allowed to freeze. 2 volumes of methanol and 1 volume of dimethylformamide.

Labelling. The label states (1) the content of Trimethoprim Test solution. Shake a quantity of the powdered tablets
and of Sulphamethoxazole in each 5 ml of the suspension; (2) containing 0.4 g of Sulphamethoxazole with 20 ml of methanol
that the contents should be shaken before use; (3) that a and filter.
suspension containing 40 mg of Trimethoprim and 200 mg of Reference solution (a). A 2.0 per cent w/v solution of
Sulphamethoxazole in 5 ml is meant for paediatric use. sulphamethoxazole RS in methanol.

1207
TRIMETHOPRIM TABLETS IP 2007

Reference solution (b). A 0.4 per cent w/v solution of On the residue, determine by infrared absorption
trimethoprim RS in methanol. spectrophotometry (2.4.6). Compare the spectrum with that
Apply to the plate 5 µl of each solution. After development, obtained with trimethoprim RS or with the reference spectrum
dry the plate in air, spray with dilute potassium of trimethoprim.
iodobismuthate solution. One of the principal spots in the B. Shake a quantity of the powdered tablets containing 0.1 g
chromatogram obtained with the test solution corresponds to of Trimethoprim with 60 ml of 0.1 M hydrochloric acid for
that in the chromatogram obtained with reference solution (a) 20 minutes, add sufficient 0.1 M hydrochloric acid to produce
and the other corresponds to that in the chromatogram 100 ml, filter and dilute 5 ml of the filtrate to 250 ml with 0.1 M
obtained with reference solution (b) sodium hydroxide.

Tests When examined in the range 230 nm to 360 nm (2.4.7), the


resulting solution shows an absorption maximum at about
Other tests. Comply with the tests stated under Tablets. 287 nm.
Assay. For trimethoprim — Weigh accurately a quantity of Tests
the powdered tablets containing about 50 mg of Trimethoprim,
add 30 ml of 0.1 M sodium hydroxide and extract with four Related substances. Determine by liquid chromatography
quantities, each of 50 ml, of chloroform, washing each extract (2.4.14).
with the same two quantities, each of 10 ml, of 0.1 M sodium Test solution. Shake a quantity of the powdered tablets
hydroxide. Combine the chloroform extracts and extract with containing 0.2 g of Trimethoprim with 50 ml of the mobile
four quantities, each of 50 ml, of 1 M acetic acid. Wash the phase, filter and use the filtrate.
combined extracts with 5 ml of chloroform and dilute the
aqueous extracts to 250.0 ml with 1 M acetic acid. To 10.0 ml Reference solution. Dilute 1 volume of the test solution to
of the solution add 10 ml of 1 M acetic acid and sufficient 100 volumes with the mobile phase.
water to produce 100.0 ml, mix and measure the absorbance of Chromatographic system
the resulting solution at the maximum at about 271 nm (2.4.7). – a stainless steel column 20 cm x 5 mm, packed with
Calculate the content of C14H18N4O3 taking 204 as the specific octadecylsilane bonded to porous silica (5 µm),
absorbance at 271 nm. – mobile phase: 0.14 per cent w/v solution of sodium
perchlorate in methanol (60 per cent) adjusted to pH
For sulphamethoxazole — Weigh and powder 20 tablets. 3.1 with 0.1 M hydrochloric acid,
Weigh accurately a quantity of the powder containing about – flow rate. 1.3 ml per minute,
0.2 g of Sulphamethoxazole, dissolve as completely as possible – spectrophotometer set at 280 nm,
in 60 ml of water and 10 ml of hydrochloric acid. Add 3 g of – a 20 µl loop injector.
potassium bromide, cool in ice and carry out the nitrite titration
(2.3.31). The column efficiency, determined using the peak due to
trimethoprim in the chromatogram obtained with reference
1 ml of 0.1 M sodium nitrite is equivalent to 0.02533 g of solution, should be at least 8,000 theoretical plates per metre.
C10H11N3O3S.
In the chromatogram obtained with the test solution the sum
Storage. Store protected from light and moisture. of the areas of any secondary peaks is not greater than the
Labelling. The label states the quantities of Trimethoprim area of the principal peak in the chromatogram obtained with
and of Sulphamethoxazole in each tablet. reference solution.
Other tests. Comply with the tests stated under Tablets.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Trimethoprim Tablets quantity of the powder containing about 0.1 g of Trimethoprim,
add 100 ml of glacial acetic acid, shake for 20 minutes, dilute
Trimethoprim Tablets contain not less than 95.0 per cent and
to 200.0 ml with glacial acetic acid and filter. To 5.0 ml of the
not more than 105.0 per cent of the stated amount of
filtrate add 15 ml of glacial acetic acid and dilute to 100.0 ml
trimethoprim, C14H18N4O3.
with water. Measure the absorbance of the resulting solution
Identification at the maximum at about 271 nm (2.4.7).
Calculate the content of C14H18N4O3 taking 204 as the specific
A. Shake a quantity of the powdered tablets containing 0.1 g
absorbance at 271 nm.
of Trimethoprim with 10 ml of chloroform, filter and evaporate
the filtrate to dryness. Storage. Store protected from moisture.

1208
IP 2007 TRIPROLIDINE TABLETS

Triprolidine Hydrochloride the chromatogram obtained with the test solution any spot
corresponding to Z-triprolidine is not more intense than the
spot in the chromatogram obtained with reference solution
H (b) and any other secondary spot is not more intense than the
N
spot in the chromatogram obtained with reference solution (a).
N , HCl , H2O
Heavy metals (2.3.13). 1.0 g complies with the limit test for
CH3 heavy metals, Method B (20 ppm).
Sulphated ash (2.3.18). Not more than 0.1 per cent.
C19H22N2,HCl,H2O Mol. Wt. 332.9
Water (2.3.43). 4.0 to 6.0 per cent, determined on 0.4 g.
Triprolidine Hydrochloride is (E)-2-(3-pyrrolidin-1-yl-1-(4-
tolyl)prop-1-enyl)pyridine hydrochloride monohydrate. Assay. Weigh accurately about 0.25 g and dissolve in a mixture
of 50 ml of anhydrous glacial acetic acid and 0.5 ml of acetic
Triprolidine Hydrochloride contains not less than 98.0 per
anhydride. Titrate with 0.1 M perchloric acid, using crystal
cent and not more than 101.0 per cent of C19H22N2, HCl,
violet solution as indicator. Carry out a blank titration.
calculated on the anhydrous basis.
Description. A white, crystalline powder; almost odourless. 1 ml of 0.1 M perchloric acid is equivalent to 0.01574 g of
C19H22N2, HCl.
Identification Storage. Store protected from light and moisture.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with triprolidine
hydrochloride RS or with the reference spectrum of triprolidine Triprolidine Tablets
hydrochloride.
Triprolidine Hydrochloride Tablets
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.001 per cent w/v solution shows absorption maxima at about Triprolidine Tablets contain not less than 90.0 per cent and
230 nm and 276 nm; absorbance at about 230 nm, about not more than 110.0 per cent of the stated amount of triprolidine
0.50 and at about 276 nm, about 0.25. hydrochloride, C19H22N2,HCl,H2O.
C. When examined in the range 230 nm to 360 nm (2.4.7), a Identification
0.002 per cent w/v solution in 0.05 M sulphuric acid shows
an absorption maximum at about 290 nm; absorbance at about A. Extract a quantity of the powdered tablets containing
290 nm, about 0.6. 10 mg of Triprolidine Hydrochloride with ether, filter, discard
the ether extract and dry the residue. Extract the residue with
D. Dissolve 0.1 g in 2 ml of 2 M hydrochloric acid and add
chloroform, filter and evaporate the filtrate to dryness. Add
0.5 ml of potassium mercuri-iodide solution; a pale yellow
0.1 ml of ether, stir and allow to evaporate.
precipitate is produced.
E. A 5 per cent w/v solution gives the reactions of chlorides On the residue, determine by infrared absorption
(2.3.1). spectrophotometry (2.4.6). Compare the spectrum with that
obtained with triprolidine hydrochloride RS or with the
Tests reference spectrum of triprolidine hydrochloride.
Related substances. Determine by thin-layer chromatography B. In the test for Related substances, the principal spot in the
(2.4.17), coating the plate with silica gel HF254. chromatogram obtained with the test solution corresponds to
that in the chromatogram obtained with reference solution (b).
Mobile phase. A mixture of equal volumes of 2-butanone and
dimethylformamide. Tests
Test solution. Dissolve 0.1 g of the substance under
Related substances. Determine by thin-layer chromatography
examination in 10 ml of methanol.
(2.4.17), coating the plate with silica gel HF254.
Reference solution (a). Dissolve 10 mg of the substance under
Mobile phase. A mixture of equal volumes of 2-butanone and
examination in 100 ml of methanol.
dimethylformamide.
Reference solution (b). A 0.02 per cent w/v solution of
Test solution. Extract a quantity of powdered tablets containing
Z-triprolidine RS in methanol.
10 mg of Triprolidine Hydrochloride with methanol, filter,
Apply to the plate 5 µl of each solution. After development, evaporate to dryness and dissolve the residue in 1 ml of
dry the plate in air and examine in ultraviolet light at 254 nm. In methanol.

1209
TRISODIUM EDETATE CONCENTRATE FOR INJECTION IP 2007

Reference solution (a). A 0.02 per cent w/v solution of Disodium Edetate and Sodium Hydroxide. It should be diluted
Z-triprolidine RS in methanol. with a suitable diluent in accordance with the manufacturer’s
Reference solution (b). A 1 per cent w/v solution of instructions.
triprolidine hydrochloride RS in methanol. Trisodium Edetate Concentrate for Injection contains not less
Reference solution (c). A 0.01 per cent w/v solution of than 19.0 per cent w/v and not more than 21.0 per cent w/v
triprolidine hydrochloride RS in methanol. solution of trisodium edetate, C10H13N2Na3O8.
Apply to the plate 5 µl of each solution. After development, Description. A colourless solution.
dry the plate in air and examine in ultraviolet light at 254 nm. In
the chromatogram obtained with the test solution any spot Identification
corresponding to Z-triprolidine is not more intense than the
spot in the chromatogram obtained with reference solution (a) A. Dissolve 2 g in 25 ml of water, add 6 ml of lead nitrate
and any other secondary spot is not more intense than the solution, shake and add 3 ml of potassium iodide solution;
spot in the chromatogram obtained with reference solution no yellow precipitate is produced. Make alkaline to red litmus
(c). paper with 2 M ammonia and add 5 ml of ammonium oxalate
solution; no precipitate is produced.
Uniformity of content. For tablets containing 10 mg or less.
B. Dissolve 0.5 g in 10 ml of water, add 0.5 ml of a 10 per cent
Comply with the test stated under Tablets.
w/v solution of calcium chloride, make alkaline to red litmus
Powder one tablet, weigh accurately a quantity of the powder paper with 2 M ammonia and add 3 ml of ammonium oxalate
containing 7.5 mg of Triprolidine Hydrochloride and carry out solution; no precipitate is produced.
the Assay beginning at the words “add 15 ml of water....”.
C. Evaporate to dryness and ignite. The residue gives the
Calculate the content of C19H22N2,HCl,H2O in the tablet. reactions of sodium salts (2.3.1).
Other tests. Comply with the tests stated under Tablets.
Tests
Assay. Weigh and powder 20 tablets. Weigh accurately a
quantity of the powder containing about 7.5 mg of Triprolidine pH (2.4.24). 7.0 to 8.0.
Hydrochloride, add 15 ml of water and 1 g of sodium chloride,
shake for 2 to 3 minutes and add sufficient 5 M sodium Pyrogens (2.2.8). Complies with the test, using per kg of the
hydroxide to make it alkaline. Extract with four quantities, rabbit’s weight 5 ml of a solution prepared in the following
each of 20 ml, of ether and wash the combined extracts with manner. To 1 volume of the injection add 2.5 volumes of calcium
two quantities, each of 5 ml, of a mixture of equal volumes of a gluconate solution. Dilute the resulting solution with
saturated solution of sodium chloride and water. Extract the sufficient water for injections to give a final concentration of
ether solution with 20 ml of 0.1 M hydrochloric acid, wash 5.0 per cent w/v solution of trisodium edetate.
the ether with two quantities, each of 5 ml, of water and add
Other tests. Complies with the tests stated under Parenteral
the washings to the acid extract. Heat on a water-bath for
Preparations (Concentrated Solutions for Injection).
30 minutes, cool and add sufficient water to produce 50.0 ml.
Dilute 10.0 ml to 100.0 ml with 0.1 M hydrochloric acid. Assay. Dilute 10.0 ml to 100.0 ml with water and use this
Measure the absorbance of the resulting solution at the solution to titrate a mixture of 25.0 ml of 0.05 M magnesium
maximum at about 290 nm (2.4.7). sulphate and 10 ml of ammonia buffer pH 10.9 using mordant
Calculate the content of C19H22N2, HCl, H2O taking 290 as the black II mixed triturate as indicator.
specific absorbance at 290 nm. 1 ml of 0.05 M magnesium sulphate is equivalent to 0.01791 g
Storage. Store protected from light and moisture. of C10H13N2Na3O8.
Storage. Store in hermetically-sealed, lead-free glass
containers.
Labelling. The label states (1) the strength in terms of
Trisodium Edetate Concentrate for
anhydrous trisodium edetate contained in a suitable dose-
Injection volume; (2) ‘Trisodium Edetate Concentrate for Injection’; (3)
Trisodium Edetate Concentrate for Injection is a sterile that the solution must be diluted with either Sodium Chloride
solution in Water for Injections containing 20 per cent w/v Intravenous Infusion or Dextrose Intravenous Infusion before
solution of trisodium edetate prepared by the interaction of administration.

1210
IP 2007 TROPICAMIDE EYE DROPS

Tropicamide more than one such spot is more intense than the spot in the
chromatogram obtained with reference solution (b).
H3C OH Sulphated ash (2.3.18). Not more than 0.1 per cent.
N
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
N on 1.0 g by drying in an oven at 80º at a pressure not exceeding
0.7 kPa for 4 hours.
O
Assay. Weigh accurately about 0.5 g, dissolve in 80 ml of
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
C17H20N2O2 Mol. Wt. 284.4 acid, using 1-naphtholbenzein solution as indicator. Carry
Tropicamide is (RS)-N-ethyl-3-hydroxy-2-phenyl-N-(pyrid-4- out a blank titration.
ylmethyl)propionamide 1 ml of 0.1 M perchloric acid is equivalent to 0.02844 g of
Tropicamide contains not less than 99.0 per cent and not more C17H20N2O2.
than 101.0 per cent of C17H20N2O2, calculated on the dried Storage. Store protected from light and moisture.
basis.
Description. A white or almost white, crystalline powder;
odourless or almost odourless.
Tropicamide Eye Drops
Identification
Tropicamide Eye Drops are a sterile solution of Tropicamide
A. Determine by infrared absorption spectrophotometry (2.4.6). in Purified Water. They may contain stabilisers, suitable
Compare the spectrum with that obtained with tropicamide antimicrobial agents and suitable substances to increase the
RS or with the reference spectrum of tropicamide. viscosity of the solution.
B. When examined in the range 230 nm to 360 nm (2.4.7), a Tropicamide Eye Drops contain not less than 95.0 per cent
0.003 per cent w/v solution in 0.1 M hydrochloric acid shows and not more than 110.0 per cent of the stated amount of
an absorption maximum only at about 254 nm; absorbance at tropicamide, C17H20N2O2.
about 254 nm, about 0.54.
C. Dissolve 5 mg in 3 ml of a mixture of 9 ml of acetic anhydride, Identification
1 ml of 6 M acetic acid and 0.1 g of citric acid and heat on a
A. Shake a volume containing 20 mg of Tropicamide with
water-bath for 5 to 10 minutes; a reddish yellow colour is
10 ml of chloroform, dry the chloroform layer over anhydrous
produced.
sodium sulphate, filter and evaporate the filtrate to dryness.
Tests Dissolve the residue in minimum quantity of chloroform, add
dropwise to finely powdered potassium bromide IR, mix and
Related substances. Determine by thin-layer chromatography dry at 60º.
(2.4.17), coating the plate with silica gel GF254. On the residue determine by infrared absorption
Mobile phase. A mixture of 95 volumes of chloroform, spectrophotometry (2.4.6). Compare the spectrum with that
5 volumes of methanol and 0.5 volume of strong ammonia obtained with tropicamide RS or with the reference spectrum
solution. of tropicamide.
Test solution. Dissolve 0.1 g of the substance under B. When examined in the range 230 nm to 360 nm (2.4.7), the
examination in 10 ml of chloroform. final solution obtained in the Assay shows an absorption
maximum at about 254 nm.
Reference solution (a). A 0.005 per cent w/v solution of the
substance under examination in chloroform. Tests
Reference solution (b). A 0.002 per cent w/v solution of the
substance under examination in chloroform. pH (2.4.24). 4.0 to 5.8.
Related substances. Determine by thin-layer chromatography
Apply to the plate 20 µl of each solution. After development,
(2.4.17), coating the plate with silica gel GF254.
dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the Mobile phase. A mixture of 95 volumes of chloroform,
test solution is not more intense than the spot in the 5 volumes of methanol and 0.5 volume of strong ammonia
chromatogram obtained with reference solution (a) and not solution.

1211
TROXIDONE IP 2007

Test solution. A volume of the eye drops containing 0.2 mg of Identification


Tropicamide.
Test A may be omitted if tests B and C are carried out. Tests B
Reference solution (a). Dilute 1 volume of the eye drops to and C may be omitted if test A is carried out.
200 volumes with chloroform.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution (b). Dilute 1 volume of the eye drops to Compare the spectrum with that obtained with troxidone RS.
500 volumes with chloroform.
B. To 2 ml of a 5.0 per cent w/v solution in carbon dioxide-free
Apply to the plate 20 µl of each solution. After development,
water (solution A) add 1 ml of barium hydroxide solution; a
dry the plate in air and examine in ultraviolet light at 254 nm.
white precipitate is produced which dissolves on the addition
Any secondary spot in the chromatogram obtained with the
of 1 ml of 2 M hydrochloric acid.
test solution is not more intense than the spot in the
chromatogram obtained with reference solution (a) and not C. Dissolve 0.3 g in a mixture of 5 ml of ethanolic potassium
more than one such spot is more intense than the spot in the hydroxide solution and 5 ml of ethanol (95 per cent) and
chromatogram obtained with reference solution (b). allow to stand for 10 minutes. Add 0.05 ml of phenolphthalein
solution and carefully add hydrochloric acid until the solution
Other tests. Comply with the tests stated under Eye Drops.
is neutral. Evaporate to dryness on a water-bath, shake the
Assay. To a volume containing about 30 mg of Tropicamide residue with four quantities, each of 5 ml, of ether, filter the
add sufficient water to produce 100.0 ml. To 10.0 ml of the combined ether extracts and evaporate the filtrate to dryness.
resulting solution add 2 ml of a 10 per cent w/v solution of The residue, after recrystallisation from 5 ml of toluene and
anhydrous sodium carbonate and extract with four quantities, drying, melts at about 80º (2.4.21).
each of 20 ml, of chloroform. Wash the combined chloroform
extracts with 25 ml of phosphate buffer pH 6.5. Wash the Tests
aqueous layer with 10 ml of chloroform, combine the
chloroform extracts and shake with four quantities, each of Appearance of solution. Solution A is clear (2.4.1), and
20 ml, of 0.5 M sulphuric acid. Combine the acid extracts, colourless (2.4.1).
dilute to 100.0 ml with 0.5 M sulphuric acid and measure the Acidity or alkalinity. To 10 ml of solution A add 0.1 ml of
absorbance of the resulting solution at the maximum at about methyl red solution. Not more than 0.1 ml of 0.01 M
254 nm (2.4.7). hydrochloric acid or 0.01 M sodium hydroxide is required to
Calculate the content of C17H20N2O2 taking 172 as the specific change the colour of the solution.
absorbance at 254 nm. Heavy metals (2.3.13). 1.0 g complies with the limit test for
Storage. Store in a refrigerator (8º to 15º). It should not be heavy metals, Method B (20 ppm).
allowed to freeze. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying over anhydrous silica gel for 6 hours.

Troxidone Assay. Determine by gas chromatography (2.4.13).


Test solution. Weigh accurately 0.1 g of the substance under
Trimethadione
examination and dissolve in sufficient solutions prepared by
dissolving 0.125 g of 1-decanol (internal standard) in sufficient
H3 C O ethanol to produce 25.0 ml (solution B).
O
H3 C Reference solution. Dissolve 0.1 g of troxidone RS in sufficient
N
solution B to produce 10.0 ml.
O CH3
Chromatographic system
C6H9NO3 Mol. Wt. 143.1 – a stainless steel column 0.75 m x 3 mm, packed with
porous polymer beads (120 to 150 µm),
Troxidone is 3,5,5-trimethyloxazolidine-2,4-dione.
– temperature:
Troxidone contains not less than 98.0 per cent and not more column 210º,
than 102.0 per cent of C6H9NO3, calculated on the dried basis. inlet port at 240º and detector at 270º,
Description. Colourless or almost colourless crystals; odour, – flow rate. 30 ml per minute of the carrier gas.
slightly camphoraceous. Inject 1 µl of the test solution and the reference solution.

1212
IP 2007 TUBOCURARINE CHLORIDE

Calculate the content of C6H9NO3. Reference solution. Dissolve 0.1 g of troxidone RS in sufficient
Storage. Store protected from light and moisture. solution B to produce 10.0 ml.
Chromatographic system
– a stainless steel column 0.75 m x 3 mm, packed with
porous polymer beads (120 to 150 µm),
Troxidone Capsules – temperature:
column 210°,
Trimethadione Capsules inlet port at 240° and detector at 270°,
Troxidone Capsules contain not less than 95.0 per cent and – flow rate. 30 ml per minute of the carrier gas.
not more than 105.0 per cent of the stated amount of troxidone, Inject 1 µl of the test solution and the reference solution.
C6H9NO3.
Calculate the content of C6H9NO3 in the capsules.
Identification Storage. Store protected from moisture.
To a quantity of the contents of the capsules containing 1 g of
Troxidone add 25 ml of ether, set aside in a stoppered flask for
20 minutes, decant the ether through a filter, and if an insoluble
residue remains, digest it with another 10-ml portion of ether
Tubocurarine Chloride
as before, and filter into the first ether filtrate. Evaporate the
ether extracts to dryness with the aid of a current of air and H3 CO
dry the residue at a pressure of 2 kPa for 2 hours. The residue OH
complies with the following tests. N
O CH3
Test A may be omitted if tests B and C are carried out. Tests B
and C may be omitted if test A is carried out.
Cl, HCl, 5H2O
A. Determine by infrared absorption spectrophotometry (2.4.6). CH2 O
Compare the spectrum with that obtained with troxidone RS H3C
OH
or with the reference spectrum of troxidone. H3C N
B. To 2 ml of a 5.0 per cent w/v solution in carbon dioxide-free
OCH3
water (solution A) add 1 ml of barium hydroxide solution; a
white precipitate is produced which dissolves on the addition
of 1 ml of 2 M hydrochloric acid. C37H41ClN2O6,HCl,5H2O Mol. Wt. 771.7
C. Dissolve 0.3 g in a mixture of 5 ml of ethanolic potassium Tubocurarine Chloride is 7′,12′-dihydroxy-6,6′-dimethoxy-
hydroxide solution and 5 ml of ethanol (95 per cent) and 2,2′,2′-trimethyltubocuraranium chloride hydrochloride
allow to stand for 10 minutes. Add 0.05 ml of phenolphthalein pentahydrate.
solution and carefully add hydrochloric acid until the solution
Tubocurarine Chloride contains not less than 98.0 per cent
is neutral. Evaporate to dryness on a water-bath, shake the
and not more than 102.0 per cent of C37H41ClN2O6,HCl,
residue with four quantities, each of 5 ml, of ether, filter the
calculated on the anhydrous basis.
combined ether extracts and evaporate the filtrate to dryness.
The residue, after recrystallisation from 5 ml of toluene and Description. A white or yellowish white, crystalline powder.
drying, melts at about 80º (2.4.21).
Identification
Tests
Test A may be omitted if tests B, C, D and E are carried out.
Other tests. Comply with the tests stated under Capsules. Tests B, C and D may be omitted if tests A and E are carried
Assay. Determine by gas chromatography (2.4.13). out.

Test solution. To a quantity of the contents of the capsules A. Determine by infrared absorption spectrophotometry (2.4.6).
containing about 1.0 g of Troxidone add 25 ml of a 0.5 per cent Compare the spectrum with that obtained with tubocurarine
w/v solution of 1-decanol (internal standard) in ethanol, shake chloride RS or with the reference spectrum of tubocurarine
for 30 minutes, add sufficient of internal standard solution to chloride.
produce 100.0 ml, mix and centrifuge. Use the supernatant B. When examined in the range 230 nm to 360 nm (2.4.7), a
liquid. 0.005 per cent w/v solution shows an absorption maximum at

1213
TUBOCURARINE INJECTION IP 2007

about 280 nm and a minimum at about 255 nm, 0.56 to 0.62, water and 2 volumes of ferric chloride solution, prepared
calculated on the anhydrous basis. immediately before use. Any secondary spot in the
C. To 1 ml of a 2.5 per cent w/v solution add 0.2 ml of ferric chromatogram obtained with the test solution is not more
chloride solution and heat in a water-bath for 1 minute; a intense than the spot in the chromatogram obtained with
green colour is produced; 1 ml of water treated in the same reference solution (a) and not more than one such spot is
manner gives a brown colour. more intense than the spot in the chromatogram obtained
with reference solution (b).
D. To 20 ml of a 0.05 per cent w/v solution add 0.2 ml of
sulphuric acid and 2 ml of a 1 per cent w/v solution of Sulphated ash (2.3.18). Not more than 0.25 per cent, determined
potassium iodate, mix and warm on a water-bath for 30 minutes; on 0.2 g.
a yellow colour is produced. Water (2.3.43). 9.0 to 12.0 per cent, determined on 0.3 g.
E. Gives reaction A of chlorides and the reactions of alkaloids Assay. Weigh accurately about 25 mg, dissolve in sufficient
(2.3.1). water to produce 500.0 ml and measure the absorbance of the
resulting solution at the maximum at about 280 nm (2.4.7).
Tests
Calculate the content of C37H41ClN2O6,HCl from the absorbance
Appearance of solution. Dissolve 0.5 g in sufficient carbon obtained by repeating the operation using 25 mg, accurately
dioxide-free water to produce 50.0 ml (solution A). Solution A weighed, of tubocurarine chloride RS in place of the substance
is clear (2.4.1), and not more intensely coloured than reference under examination.
solution YS6 (2.4.1).
Storage. Store protected from moisture.
pH (2.4.24). 4.0 to 6.0, determined in Solution A.
Specific optical rotation (2.4.22). +210º to +222º, determined
in solution A 3 hours after preparation.
Chloroform-soluble substances. Not more than 2 per cent, Tubocurarine Injection
determined by the following method. Dissolve 0.25 g in 150 ml Tubocurarine Chloride Injection
of water contained in a separating funnel with a grease-free
stopcock. Add 5 ml of a saturated solution of sodium Tubocurarine Injection is a sterile solution of Tubocurarine
bicarbonate and extract with three quantities, each of 20 ml, Chloride in Water for Injections. It may contain suitable
of chloroform. Wash the combined chloroform extracts with buffering agents.
10 ml of water, filter the chloroform solution into a tared beaker Tubocurarine Injection contains not less than 95.0 per cent
and wash the filter with two successive quantities, each of and not more than 105.0 per cent of the stated amount of
5 ml, of chloroform. Add the washings to the filtrate. Remove tubocurarine chloride, C37H41ClN2O6,HCl,5H2O.
the chloroform on a water-bath, dry the residue at 105º for
Description. A colourless or faintly coloured solution.
1 hour, cool and weigh. The residue does not dissolve in 10 ml
of water but dissolves on the addition of 1 ml of 2 M Identification
hydrochloric acid.
Related substances. Determine by thin-layer chromatography A. Mix a volume containing 15 mg of Tubocurarine Chloride
(2.4.17), coating the plate with silica gel G. with 5 ml of acetone, evaporate the liquid at a pressure of
2 kPa and add successive quantities of 2 ml of acetone,
Mobile phase. The lower layer of a mixture of equal volumes evaporating each quantity at a pressure of 2 kPa until a dry
of chloroform, methanol and a 12.5 per cent w/v solution of residue is obtained.
trichloroacetic acid.
On the residue, determine by infrared absorption
Test solution. Dissolve 0.25 g of the substance under spectrophotometry (2.4.6). Compare the spectrum with that
examination in 10 ml of water. obtained with tubocurarine chloride RS or with the reference
Reference solution (a). A 0.0375 per cent w/v solution of the spectrum of tubocurarine chloride.
substance under examination in water. B. Dilute 1 ml to 30 ml with water. To 1 ml of the resulting
Reference solution (b). A 0.01875 per cent w/v solution of the solution add 0.5 ml of mercuric nitrate solution; a cherry-red
substance under examination in water. colour slowly develops.
Apply to the plate 5 µl of each solution. After development, Tests
dry the plate in a current of cold air and spray with a mixture of
1 volume of potassium ferricyanide solution, 1 volume of pH (2.4.24). 4.0 to 6.0.

1214
IP 2007 TUBOCURARINE INJECTION

Optical rotation (2.4.22).+0.172º to +0.206º for each mg of 1 volume of potassium ferricyanide solution, 1 volume of
tubocurarine chloride, C37H41ClN2O6,HCl,5H2O per ml stated water and 2 volumes of ferric chloride solution, prepared
on the label. immediately before use. Any secondary spot in the
Related substances. Determine by thin-layer chromatography chromatogram obtained with the test solution is not more
(2.4.17), coating the plate with silica gel G. intense than the spot in the chromatogram obtained with
reference solution (a) and not more than one such spot is
Mobile phase. The lower layer of a mixture of equal volumes more intense than the spot in the chromatogram obtained
of chloroform, methanol and a 12.5 per cent w/v solution of with reference solution (b).
trichloroacetic acid.
Other tests. Complies with the tests stated under Parenteral
Test solution. A volume of the injection containing 10 mg of Preparations (Injections).
Tubocurarine Chloride diluted to 1 ml.
Assay. Dilute a volume containing about 50 mg of Tubocurarine
Reference solution (a). Dilute 3 volumes of test solution to Chloride to 1000.0 ml with water and measure the absorbance
200 volumes with water. of the resulting solution at the maximum at about 280 nm (2.4.7).
Reference solution (b). Dilute 1 volume of reference solution Calculate the content of C37H41ClN2O6,HCl,5H2O taking 105 as
(a) to 2 volumes with water. the specific absorbance at 280 nm.
Apply to the plate 10 µl of each solution. After development, Storage. Store protected from moisture.
dry the plate in a current of cold air and spray with a mixture of

1215
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

U
Undecenoic Acid ....
Urea ....
Urea Cream ....
Urokinase ....

1217
IP 2007 UREA

Undecenoic Acid 1 ml of 0.5 M sodium hydroxide is equivalent to 0.09214 g of


C11H20O2.
Undecylenic Acid
Storage. Store protected from light and moisture.
H

H2C COOH
Urea
C11H20O2 Mol. Wt. 184.3
O
Undecenoic Acid is 10-undecenoic acid.
H2 N NH2
Undecenoic Acid contains not less than 97.0 per cent and not
more than 102.0 per cent of C11H20O2. CH4N2O Mol. Wt. 60.1
Description. A white or very pale yellow, crystalline mass or Urea is the diamide of carbonic acid.
colourless or pale yellow liquid; odour, characteristic.
Urea contains not less than 99.0 per cent and not more than
Identification 101.0 per cent of CH4N2O, calculated on the dried basis.
Description. A white, crystalline powder or transparent
A. Dissolve 0.1 g in a mixture of 2 ml of 1 M sulphuric acid and
crystals; odourless or almost odourless, but may gradually
5 ml of glacial acetic acid and add dropwise 0.25 ml of
develop a slight odour of ammonia upon long standing;
potassium permanganate solution; the colour of the
slightly hygroscopic.
permanganate solution is discharged.
B. Boil 2 g under a reflux condenser with 2 ml of freshly distilled Identification
aniline for 10 minutes, allow to cool, add 30 ml of ether and
Test A may be omitted if tests B and C are carried out. Tests B
extract with three quantities, each of 20 ml, of 2 M hydrochloric
and C may be omitted if test A is carried out.
acid and then with 20 ml of water. Evaporate the organic layer
to dryness on a water-bath. The residue, after recrystallising A. Determine by infrared absorption spectrophotometry (2.4.6).
twice from ethanol (70 per cent) and drying over phosphorus Compare the spectrum with that obtained with urea RS or
pentoxide at a pressure of 1.5 to 2.5 kPa for 3 hours melts at with the reference spectrum of urea.
66º to 68º. B. Heat 0.5 g in a test-tube; it liquefies, and ammonia is evolved
which is recognised by its characteristic odour. Heat further
Tests
until the liquid is turbid, cool and dissolve in 10 ml of water.
Congealing range (2.4.10). 21º to 24º. Add 1 ml of a 10 per cent w/v solution of sodium hydroxide
Refractive index (2.4.27). 1.447 to 1.450. and 0.05 ml of copper sulphate solution; a reddish violet colour
is produced.
Peroxide value (2.3.35). Not more than 10.
C. Dissolve 0.1 g in 1 ml of water and add 1 ml of nitric acid;
Iodine value (2.3.28). 131 to 140.
a white, crystalline precipitate is produced.
Fixed and mineral oils. Boil 1.0 g with 25 ml of water and 5 ml
of sodium carbonate solution for 3 minutes. The hot solution Tests
is not more opalescent than opalescence standard OS2 (2.4.1). Appearance of solution. Solution A is clear (2.4.1), and
Water-soluble acids. Shake 2.0 g with 20 ml of warm water, colourless (2.4.1).
allow to separate and filter the aqueous layer through a
Alkalinity. To 10 ml of a 5.0 per cent w/v solution (solution A)
moistened filter paper. To 5 ml of the filtrate add 0.01 ml of
add 0.1 ml of methyl red solution and 0.4 ml of 0.01 M
dilute phenolphthalein solution. Not more than 0.1 ml of
hydrochloric acid; the resulting solution is red to orange.
0.1 M sodium hydroxide is required to change the colour of
the solution. Biuret. Not more than 0.1 per cent, determined by the following
method. To 10 ml of a 20 per cent w/v solution add 5 ml of
Heavy metals (2.3.13). 2.0 g complies with the limit test for
water, 0.5 ml of a 0.5 per cent w/v solution of copper sulphate
heavy metals, Method B (10 ppm).
and 0.5 ml of 10 M sodium hydroxide and allow to stand for
Sulphated ash (2.3.18). Not more than 0.15 per cent. 5 minutes. Any reddish violet colour obtained is not more
Assay. Weigh accurately about 0.75 g, dissolve in 10 ml of intense than that in a standard prepared at the same time and
ethanol (95 per cent) and titrate with 0.5 M sodium hydroxide in the same manner using 10 ml of a 0.02 per cent w/v solution
using 0.1 ml of dilute phenolphthalein solution as indicator. of biuret in place of the substance under examination.

1219
UREA CREAM IP 2007

Ethanol-insoluble matter. Not more than 0.04 per cent, Apply to the plate 10 µl of each solution. After development,
determined by the following method. Dissolve 5.0 g in 50 ml of dry the plate in air, spray with a solution containing 0.5 per
warm ethanol (95 per cent), filter through a tared filter, wash cent w/v solution of 4-dimethylaminobenzaldehyde and
the filter with 20 ml of warm ethanol (95 per cent) and dry at 0.5 per cent v/v of sulphuric acid in ethanol. The principal
105º for 1 hour. spot in the chromatogram obtained with the test solution
Heavy metals (2.3.13). Dissolve 1.0 g in 20 ml of water and 5 ml corresponds to that in the chromatogram obtained with
of 0.1 M hydrochloric acid. The solution complies with the reference solution (a). The chromatogram obtained with
limit test for heavy metals, Method A (20 ppm). reference solution (b) shows a single, compact spot.

Sulphated ash (2.3.18). Not more than 0.1 per cent. B. To a quantity containing 0.1 g of Urea add 50 ml of water
and heat until dispersed, cool in ice and filter through glass
Loss on drying (2.4.19). Not more than 1.0 per cent, determined wool. Adjust the pH to 6.0 to 7.0 using 0.1 M hydrochloric
on 1.0 g by drying in an oven at 105º for 1 hour. acid or 0.1 M sodium hydroxide as necessary. To 5 ml add
Assay. Weigh accurately about 0.5 g, dissolve in sufficient of 5 ml of a 0.1 per cent w/v suspension of urease-active meal
a 10 per cent v/v solution of sulphuric acid to produce and allow to stand for 30 minutes in a stoppered flask at 37º.
100.0 ml and mix. Place 5.0 ml of the resulting solution in a When the resulting solution is heated in a water-bath a vapour
long-necked flask, add 10 ml of sulphuric acid and heat gently is produced that turns moist red litmus paper blue.
until evolution of gas ceases. Boil gently for 10 minutes, cool,
cautiously add 40 ml of water, cool again and place in a steam- Tests
distillation apparatus. Add 50 ml of 10 M sodium hydroxide Other tests. Comply with the tests stated under Creams.
and distil immediately by passing steam through the mixture.
Continue the distillation for 1 hour, collecting the distillate in Assay. Weigh accurately a quantity containing about 42 mg
40 ml of a 4 per cent w/v solution of boric acid. Titrate with of Urea, shake with 150 ml of hot water for 20 minutes, allow to
0.1 M hydrochloric acid, using 0.25 ml of methyl red- cool and dilute to 500.0 ml with water. Filter through a fine
methylene blue solution as indicator. Carry out a blank glass microfibre filter paper or by any other means, transfer
titration. 1.0 ml of the filtrate to a 100-ml volumetric flask, add 2 ml of a
0.1 per cent w/v suspension of urease-active meal, stopper
1 ml of 0.1 M hydrochloric acid is equivalent to 0.003003 g of the flask and allow to stand for 15 minutes at 37º. Immediately
CH4N2O. add 25 ml of a solution containing 12 g of sodium salicylate
Storage. Store protected from moisture. and 0.24 g of sodium nitroprusside in 200 ml and 25 ml of a
solution prepared by diluting a volume of sodium hypochlorite
solution containing 0.66 g of available chlorine with 0.2 M
Urea Cream sodium hydroxide to 1000 ml. Mix well, allow to stand at 37º
for 5 minutes and dilute to 100.0 ml with water. Measure the
Urea Cream contains Urea in a suitable basis. absorbance of the resulting solution at the maximum at about
Urea Cream contains not less than 90.0 per cent and not more 665 nm (2.4.17), using as the blank a solution prepared in the
than 110.0 per cent of the stated amount of urea, CH4N2O. same manner but using 1.0 ml of water in place of 1.0 ml of the
filtrate.
Identification
Calculate the content of CH4N2O from the absorbance obtained
A. Determine by thin-layer chromatography (2.4.17), coating by using 42 mg of urea RS in place of the substance under
the plate with silica gel G. examination.
Mobile phase. For the first development use 2,2,4- Storage. Store in accordance with the instructions of the
trimethylpentane. Dry the plate in air. For the second manufacturer.
development use a mixture of 99 volumes of ethanol and
1 volume of strong ammonia solution.
Test solution. Disperse with heating a quantity of the cream Urokinase
containing 50 mg of Urea in 1 ml of water, cool, add 4 ml of
acetone, mix and filter. Urokinase is an enzyme, obtained from human urine that
activates plasminogen. It consists of a mixture of low molecular
Reference solution (a). Dissolve 50 mg of urea RS in 1 ml of weight and high molecular weight forms, the high molecular
water and add 4 ml of acetone. weight form being predominant. The molecular weights of the
Reference solution (b). A mixture of equal volumes of the test low and high molecular weight forms are 33,000 and 54,000
solution and reference solution (a). respectively.

1220
IP 2007 UROKINASE

It is prepared in conditions designed to minimise microbial examination and multiplying the result by 6.25 to obtain the
and viral contamination. In particular, adequate measures, such content of protein.
as heating the substance in solution at 60º for 10 hours, are Hepatitis-B surface antigen. Examine by a suitably sensitive
taken to inactivate viruses. method such as radio-immunoassay; hepatitis-B surface
Urokinase contains not less than 70,000 Units of urokinase antigen is not detected.
activity per mg of protein. Abnormal toxicity (2.2.1). Complies with the test, using a
Description. A white or almost white, amorphous powder. solution containing 2,500 Units in 0.5 ml of saline solution.
Thromboplastic contaminants. Dissolve suitable quantities
Identification
of the substance under examination in barbitone buffer
A. Place 0.5 ml of citrated human plasma and 0.5 ml of citrated pH 7.4 to obtain solutions containing 5000, 2500, 1250, 625
bovine plasma in two separate haemolysis tubes maintained and 312 Units per ml. Into each of six haemolysis tubes, 1 cm
in a water-bath at 37º. To each tube add 0.1 ml of a solution of in internal diameter, place 0.1 ml of citrated rabbit plasma. Add
the substance under examination containing 1000 Units per 0.1 ml of one of each of the solutions of the substance under
ml in phosphate buffer pH 7.4 and 0.1 ml of a solution of examination to each of five of the tubes and 0.1 ml of barbitone
thrombin containing 20 Units per ml in phosphate buffer buffer pH 7.4 to the sixth (control). Incubate the six tubes at
pH 7.4 and shake immediately; in both tubes, a clot forms and 25º ± 0.5º for 5 minutes and then add 0.1 ml of a 0.3675 per cent
lyses within 30 minutes. w/v solution of calcium chloride. Using a stop-watch,
measure the clotting time for each solution and the control.
B. Carry out a suitable immunodiffusion test.
Plot the shortening of the recalcification time (control clotting
Tests time minus clotting time for each solution) against log
concentration. Extrapolate the best-fitting straight line through
Appearance of solution. A 0.1 per cent w/v solution in water is the five points until it reaches the log concentration axis. The
clear (2.4.1), and colourless (2.4.1). urokinase activity at the intersection point represents the limit
Molecular fractions. Determine by size-exclusion concentration for clotting activity (zero clotting activity). The
chromatography (2.4.16) zero clotting activity is not less than 150 Units per ml.
Test solution. Dissolve 0.1 g of the substance under Vasoactive substances. Anesthetise a rabbit by intraperitoneal
examination in 100 ml of 0.02 M phosphate buffer pH 8.0. injection of 0.15 g of phenobarbitone sodium per kg of body
weight. Dissolve in normal saline solution a sufficient
Chromatographic system quantity of the substance under examination to give a solution
– a column 90 cm x 16 mm, packed with a cross-linked containing 40,000 Units per ml. Administer by intravenous
dextran suitable for fractionation of proteins in the range infusion at a rate of 1 ml per minute a sufficient volume of the
of molecular weight from 4000 to 150,000 (such as solution of the substance under examination such that the
Sephadex G-100), at 5º, dose is 20,000 Units per kg of body weight. Measure the arterial
– mobile phase: 1.75 per cent w/v solution of sodium pressure and heart rate at intervals of 15 minutes for 5 hours
chloride in 0.02 M phosphate buffer pH 8.0, after the infusion. No significant and lasting alterations in
– flow rate. 6 ml per hour, arterial pressure or heart rate are produced, except those arising
– spectrophotometer set at 280 nm. from the effects of the anaesthetic.
Apply 1 ml of the test solution to the column, rinse twice with Assay. The potency of urokinase is determined by comparing
0.5-ml portions of the buffer and then carry out the elution. its ability to activate human plasminogen to form plasmin with
The eluate may be collected in 1-ml fractions. Plot the individual that of the Standard Preparation. The plasmin generated is
absorbances on a graph. Draw perpendicular lines towards determined by measurement of the time taken to lyse a fibrin
the axis of the abscissae from the minima before the high clot under the conditions of a suitable method of Assay.
molecular weight peak, between the high and the low molecular
weight peaks, and after the low molecular weight peak. Standard Preparation
Combine separately the high and low molecular weight
The Standard Preparation is the 1st International Reference
fractions and for each combined fraction determine the activity
Preparation for Urokinase, human, established in 1968,
by the method described under Assay. The ratio of the activity
consisting of partially purified freeze-dried urokinase from
in the combined high molecular weight fraction to that in the
human urine with lactose (supplied in ampoules containing
combined low molecular weight fraction is not less than 2.0.
4800 Units of urokinase activity) or another suitable preparation
Total protein. Determine by Method C for the determination the activity of which has been determined in relation to the
of nitrogen (2.3.30), using 10 mg of the substance under International Reference Preparation.

1221
UROKINASE IP 2007

Method into the remaining tubes 0.5 ml of a 1.0 per cent w/v solution of
bovine euglobulin fraction. Using a stop-watch, measure for
Unless otherwise prescribed, use phosphate buffer pH 7.4
each tube the time in seconds that elapses between the
containing 3 per cent w/v solution of bovine albumin for the
addition of the euglobulin and the lysis of the clot.
preparation of solutions and dilutions.
Using the logarithms of the lysis times, calculate the result of
Prepare a solution of the Standard Preparation containing the assay by standard statistical methods.
1000 Units of urokinase activity per ml and prepare a solution
of the preparation under examination expected to have the The estimated potency is not less than 90.0 per cent and not
same concentration; keep the solutions in ice and use within more than 111.0 per cent of the stated potency.
6 hours. Prepare three 1.5-fold serial dilutions of the solution The fiducial limits of error are not less than 80 per cent and not
of the Standard Preparation so that the longest clot-lysis time more than 125 per cent of the stated potency.
is less than 20 minutes and the shortest clot-lysis time is greater
than 3 minutes. Prepare three similar dilutions of the solution Urokinase intended for use in the manufacture of parenteral
of the preparation under examination. Keep the solutions in preparations or ophthalmic preparations complies with the
ice and use within 1 hour. Using 24 tubes 8 mm in diameter, following additional requirements.
label the tubes S1, S2, S3 for the dilutions of the Standard Bacterial endotoxins (2.2.3). Not more than 0.002 Endotoxin
Preparation and T1, T2, T3 for the dilutions of the preparation Unit per Unit of urokinase activity.
under examination, allocating four tubes to each dilution. Place
Sterility (2.2.11). Complies with the test for sterility.
the tubes in ice. Into each tube introduce 0.2 ml of the
appropriate dilution, 0.2 ml of phosphate buffer pH 7.4 Storage. Store protected from light in a refrigerator (2º to 8º).
containing 3 per cent w/v solution of bovine albumin and The containers should be sterile, tamper-evident and sealed
0.1 ml of a solution containing 20 Units of thrombin per ml. so as to exclude micro-organisms.
Place the tubes in a water-bath at 37º and allow to stand for Labelling. The label states (1) the number of Units of urokinase
2 minutes to attain temperature equilibrium. Using an automatic activity in the container; (2) the number of Units of urokinase
pipette, introduce into the bottom of the first tube 0.5 ml of a activity per mg of protein; (3) the storage conditions; (4)
1.0 per cent w/v solution of bovine euglobulin fraction whether or not it is intended for use in the manufacture of
ensuring mixing. At 5-second intervals introduce successively parenteral preparations or ophthalmic preparations.

1222
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

V
Vanillin ....
Vasopressin Injection ....
Verapamil Hydrochloride ....
Verapamil Injection ....
Verapamil Tablets ....
Vinblastine Sulphate ....
Vinblastine Injection ....
Vincristine Sulphate ....
Vincristine Injection ....
Vinorelbine Tartrate ....
Vinorelbine Tartrate Injection ....
Vitamin A Concentrate Oil ....
Vitamin A Concentrate Powder ....
Water-Miscible Vitamin A Concentrate ....
Vitamins A And D Capsules ....
Concentrated Vitamin D Solution ....
Concentrated Vitamins A And D Solution ....

1223
IP 2007 VASOPRESSIN INJECTION

Vanillin Use an unsaturated tank and allow the mobile phase to rise
10 cm. Apply to the plate 5 µl of each solution. After
development, dry the plate in cold air and examine in ultraviolet
CHO
light at 254 nm. Any secondary spot in the chromatogram
obtained with test solution (a) is not more intense than the
spot in the chromatogram obtained with reference solution
OCH3 (a). Spray with dinitrophenylhydrazine-aceto-hydrochloric
OH solution and examine in daylight. Any secondary spot in the
chromatogram obtained with test solution (a) is not more
C8H8O3 Mol. Wt. 152.2 intense than the spot in the chromatogram obtained with
reference solution (a).
Vanillin is 4-hydroxy-3-methoxybenzaldehyde.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Vanillin contains not less than 99.0 per cent and not more than
101.0 per cent of C8H8O3, calculated on the dried basis. Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying over phosphorus pentoxide for 4 hours.
Description. A white or slightly yellow powder or crystalline
needles; odour, characteristic of vanilla. Assay. Weigh accurately about 0.12 g, dissolve in 20 ml of
ethanol (95 per cent), add 60 ml of carbon dioxide-free water.
Identification Titrate with 0.1 M sodium hydroxide, determining the end-
point potentiometrically (2.4.25).
Test A may be omitted if tests B, C and D are carried out. Tests
B, C and D may be omitted if test A is carried out. 1 ml of 0.1 M sodium hydroxide is equivalent to 0.01521 g of
C8H8O3.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with vanillin RS. Storage. Store protected from light and moisture.
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution Vasopressin Injection
(b). Examine the chromatograms in daylight after spraying.
Vasopressin Injection is a sterile aqueous solution containing
C. To 5 ml of a saturated solution add 0.2 ml of ferric chloride the water-soluble pressor principle obtained from the posterior
solution; a blue colour is produced. Heat to 80o; the solution lobe of the pituitary of healthy oxen or other mammals or by
becomes brown and a white precipitate is produced on cooling. synthesis.
D. Melting range (2.4.21). 81º to 84º. Vasopressin Injection contains a pressor activity equivalent
to not less than 90.0 per cent and not more than 110.0 per cent
Tests
of the stated number of Units.
Appearance of solution. A 5.0 per cent w/v solution in ethanol Description. A clear, colourless or practically colourless liquid;
(95 per cent) is clear (2.4.1), and not more intensely coloured odour, faint and characteristic.
than reference solution BS6 (2.4.1).
Related substances. Determine by thin-layer chromatography Identification
(2.4.17), coating the plate with silica gel GF254.
A. Inject into the vein of a mammal, anaesthetised by a general
Mobile phase. A mixture of 98.5 volumes of dichloromethane, anaesthetic or by destruction of the brain; it causes a rise of
1 volume of methanol and 0.5 volume of anhydrous acetic blood pressure.
acid.
B. Inject under the skin of a mammal and at the same time
Test solution (a). Dissolve 0.2 g of the substance under administer a volume of water by mouth; it causes a delay in
examination in 10 ml of methanol. the excretion of the water.
Test solution (b). Dissolve 0.2 g of the substance under
examination in 100 ml of methanol. Tests
Reference solution (a). Dissolve 10 mg of the substance under pH (2.4.24). 2.5 to 4.5.
examination in 100 ml of methanol. Oxytocin activity. Not more than 1.2 Units per 20 Units of
Reference solution (b). A 0.2 per cent w/v solution of vanillin vasopressor activity, determined by the biological assay of
RS in methanol. oxytocin.

1225
VERAPAMIL HYDROCHLORIDE IP 2007

Vasopressor activity. Not more than 0.5 Unit per 20 Units of Dilute the extract of the Standard Preparation and the
oxytocic activity, when assayed by the following method. preparation under examination with saline solution so that
The vasopressor activity is estimated by comparing the activity the volume to be injected is 0.1 ml to 0.5 ml.
of the preparation under examination with that of the Standard Choose two doses of the Standard Preparation such that the
Preparation of arginine vasopressin under the conditions of a elevation of the blood pressure is about 4 kPa for the lower
suitable method of assay. dose and about 7 kPa but always submaximal for the higher
Standard Preparation dose, the ratio of low to high dose being determined by the
response and usually being 3 to 5. As an initial approximation
The Standard Preparation is the Ist International Standard for doses of 3 and 5 milliUnits may be tried. Choose two doses of
Arginine vasopressin, established in 1978, consisting of the preparation under examination with the same inter-dose
freeze-dried synthetic arginine vasopressin peptide acetate ratio, matching the effects of the dose of the Standard
with human albumin and citric acid (supplied in ampoules Preparation as closely as possible. Inject doses at intervals of
containing 8.20 Units), or another suitable preparation the 10 to 15 minutes.
potency of which has been determined in relation to that of
the International Standard. The two doses of the Standard Preparation and the two doses
of the preparation under examination should be given in a
Suggested Method randomised block or a Latin square design and four to five
Inject slowly into the tail vein of a male albino rat weighing responses to each should be recorded.
about 300 g a solution of a suitable a-adrenoceptor blocking Measure all the responses and calculate the result of the assay
agent, for example 10 ml per kg of body weight of a solution by standard statistical methods.
prepared by dissolving 5 mg of phenoxybenzamine
hydrochloride in 0.1 ml of ethanol (95 per cent), adding Storage. Store in a refrigerator (2º to 8º).
0.05 ml of 1 M hydrochloric acid and diluting to 5 ml with Labelling. The label states (1) the number of Units of the
saline solution. After 18 hours, anaesthetise the rat with an vasopressor activity per ml; (2) either the animal source of the
anaesthetic that will maintain a prolonged and uniform blood vasopressin or that it is synthetic.
pressure. After 45 to 60 minutes, tie the rat on its back to the
operating table by its hind legs. Cannulate the trachea with a
short polyethylene tube of external diameter about 2.5 mm
and dissect a carotid artery ready for cannulation. Then Verapamil Hydrochloride
cannulate the femoral vein close to the inguinal ligament.
Verapamil Chloride; Iproveratril Hydrochloride
Retract the abdominal muscles to expose the inguinal ligament.
Retract the superficial pudendal vein to one side and dissect
the femoral vein towards the inguinal ligament from the
corresponding artery. When dissecting, a deep branch
reaching the femoral vein must be found and tied off to prevent
bleeding during cannulation. Tie a short polyethylene cannula
of external diameter about 1 mm into the femoral vein by two
ligatures and join by a short piece of flexible tubing to a 1-ml C27H38N2O4, HCl, Mol. Wt. 491.1
burette with an attached thistle funnel containing saline
solution at about 37º. Firmly fix a wet absorbent cotton swab Verapamil Hydrochloride is (2RS)-2-(3,4-dimethoxyphenyl)-
to the thigh so as to cover the incision and cannula. At this 5-[[2-(3,4-dimethoxyphenyl)ethyl](methyl)amino]-2-(1-
stage inject through the venous cannula 200 Units of heparin, methylethyl)pentanenitrile hydrochloride.
dissolved in saline solution, per 100 g of body weight. Then Verapamil Hydrochloride contains not less than 99.0 per cent
tie in a carotid cannula of external diameter about 1 mm and and not more than 101.0 per cent of C27H38N2O4, HCl, calculated
connect by a column of saline solution containing heparin on the dried basis.
with a suitable pressure measuring device such as a mercury
Description. A white, crystalline powder.
manometer of internal diameter about 2 to 3 mm.
The central and peripheral nervous system including both Identification
vagus and associated sympathetic nerves is left intact. No
Test A may be omitted if tests B, C and D are carried out. Tests
artificial respiration is necessary. Taking care that no air is
B and C may be omitted if tests A and D are carried out.
injected, inject all solutions through the venous cannula by
means of a 1-ml syringe graduated in 0.01 ml and wash in with A. Determine by infrared absorption spectrophotometry (2.4.6).
0.2 ml of saline solution from the burette. Compare the spectrum with that obtained with verapamil

1226
IP 2007 VERAPAMIL INJECTION

hydrochloride RS or with the reference spectrum of verapamil with reference solution (c) is clearly visible. Ignore any spot
hydrochloride. remaining on the line of application.
B. When examined in the range 220 nm to 360 nm (2.4.7), a B. Carry out test A but using a mixture of 85 volumes of
0.002 per cent w/v solution in 0.01 M hydrochloric acid shows cyclohexane and 15 volumes of diethylamine as the mobile
absorption maxima at about 229 nm and 278 nm and there may phase and applying separately to the plate 10 µl of each of
be a shoulder at about 282 nm. The ratio of the absorbance at test solution (a), reference solutions (b) and (c) and heat at
the maximum at about 278 nm to that at the maximum at about 110º for 90 minutes after the second development.
229 nm is 0.35 to 0.39.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
C. In test A for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Loss on drying (2.4.19). Not more than 0.5 per cent, determined
that in the chromatogram obtained with reference solution (a). on 1.0 g by drying in an oven at 105º.

D. Gives reaction B of chlorides (2.3.1). Assay. Weigh accurately about 0.4 g, dissolve in 40 ml of
anhydrous glacial acetic acid, add 6 ml of mercuric acetate
Tests solution. Titrate with 0.1 M perchloric acid, determining the
end-point potentiometrically (2.4.25). Carry out a blank titration.
Appearance of solution. A 5.0 per cent w/v solution in carbon
dioxide-free water prepared with the aid of gentle heat is clear 1 ml of 0.1 M perchloric acid is equivalent to 0.04911 g of
(2.4.1), and colourless (2.4.1). C27H38N2O4, HCl.
pH (2.4.24). 4.5 to 6.0, determined in a 5.0 per cent w/v solution Storage. Store protected from light and moisture.
prepared with the aid of gentle heat.
Related substances. A. Determine by thin-layer
chromatography (2.4.17), coating the plate with silica gel G. Verapamil Injection
Mobile phase. A mixture of 70 volumes of toluene, 20 volumes Verapamil Hydrochloride Injection; Verapamil Chloride
of methanol, 5 volumes of glacial acetic acid and 5 volumes
Injection; Iproveratril Hydrochloride Injection
of acetone.
Test solution (a). Dissolve 0.5 g of the substance under Verapamil Injection is a sterile solution of Verapamil
examination in 10 ml of chloroform. Hydrochloride in Water for Injections.

Test solution (b). Dilute 1 ml of test solution (a) to 100 ml with Verapamil Injection contains not less than 90.0 per cent and
chloroform. not more than 110.0 per cent of the stated amount of verapamil
hydrochloride, C27H38N2O4, HCl.
Reference solution (a). A 0.05 per cent w/v solution of
verapamil hydrochloride RS in chloroform. Identification
Reference solution (b). Dilute 5 ml of reference solution (a) to
A. Dilute a volume containing 10 mg of Verapamil
100 ml with chloroform.
Hydrochloride to 5 ml with 0.1 M hydrochloric acid, extract
Reference solution (c). A 0.001 per cent w/v solution of with 5 ml of ether, discard the ether extract and make the
verapamil hydrochloride RS in chloroform. aqueous layer just alkaline with 2 M potassium carbonate.
Apply to the plate 10 µl of each solution. After development, Extract with 5 ml of ether, filter the ether layer through
dry the plate in air for 10 minutes and repeat the development. anhydrous sodium sulphate and evaporate to dryness. The
Dry the plate at 110º for 30 minutes and allow to stand until the residue complies with the following test.
odour of solvent is no longer detectable. Spray with a solution Determine by infrared absorption spectrophotometry (2.4.6).
prepared by dissolving 5 g of ferric chloride hexahydrate Compare the spectrum with that obtained with verapamil
and 2 g of iodine in sufficient of a mixture of equal volumes of hydrochloride RS treated in the same manner or with the
acetone and a 20 per cent w/v solution of (+)-tartaric acid to reference spectrum of verapamil.
produce 100 ml, applying a total of 15 to 20 ml of the reagent
for a plate (20 cm x 20 cm), and examine immediately. Any B. To a volume containing 5 mg of Verapamil Hydrochloride
secondary spot in the chromatogram obtained with test add 0.2 ml of a 5 per cent w/v solution of mercuric chloride; a
solution (a) is not more intense than the principal spot in the white precipitate is produced.
chromatogram obtained with reference solution (b) and not C. To a volume containing 5 mg of Verapamil Hydrochloride
more than three such spots are more intense than the spot in add 0.5 ml of 3 M sulphuric acid and 0.2 ml of dilute potassium
the chromatogram obtained with reference solution (c). The permanganate solution; a violet precipitate is produced which
test is not valid unless the spot in the chromatogram obtained quickly dissolves to produce a very pale yellow solution.

1227
VERAPAMIL TABLETS IP 2007

Tests Identification
pH (2.4.24). 4.5 to 6.0. A. Shake a quantity of the powdered tablets containing 0.1 g
Related substances. A. Determine by thin-layer of Verapamil Hydrochloride with 25 ml of 0.1 M hydrochloric
chromatography (2.4.17), coating the plate with silica gel G. acid, filter, extract the filtrate with 25 ml of ether, discard the
ether extract and make the aqueous layer just alkaline with
Mobile phase. A mixture of 70 volumes of toluene, 20 volumes 2 M potassium carbonate. Extract with 25 ml of ether, filter the
of methanol, 5 volumes of glacial acetic acid and 5 volumes ether layer through anhydrous sodium sulphate and
of acetone. evaporate to dryness. The residue complies with the following
Test solution. Evaporate a volume containing 5 mg of Verapamil tests.
Hydrochloride carefully to dryness on a water-bath in a current
Determine by infrared absorption spectrophotometry (2.4.6).
of nitrogen and dissolve the residue as completely as possible
Compare the spectrum with that obtained with verapamil
in 0.25 ml of chloroform.
hydrochloride RS treated in the same manner or with the
Reference solution. Dilute 1 volume of the test solution to reference spectrum of verapamil.
100 volumes with chloroform and dilute 1 volume of the
B. Shake a quantity of the powdered tablets containing 0.1 g
resulting solution to 10 volumes with chloroform.
of Verapamil Hydrochloride with 10 ml of dichloromethane,
Apply to the plate 30 µl of each solution. After development, filter, evaporate the filtrate to dryness and dissolve the residue
dry the plate in air for 10 minutes and repeat the development. in 10 ml of water (solution A). To 2 ml of the resulting solution
Dry the plate at 110º for 30 minutes and allow to stand until the add 0.2 ml of a 5 per cent w/v solution of mercuric chloride; a
odour of solvent is no longer detectable. Spray with a solution white precipitate is produced.
prepared by dissolving 5 g of ferric chloride hexahydrate
C. To 2 ml of solution A obtained in test B add 0.5 ml of 3 M
and 2 g of iodine in sufficient of a mixture of equal volumes of
sulphuric acid and 0.2 ml of dilute potassium permanganate
acetone and a 20 per cent w/v solution of (+)-tartaric acid to
solution; a violet precipitate is produced which quickly
produce 100 ml, applying a total of 15 to 20 ml of the reagent
dissolves to produce a very pale yellow solution.
for a plate (20 cm x 20 cm), and examine immediately. Any
secondary spot in the chromatogram obtained with the test Tests
solution is not more intense than the principal spot in the
chromatogram obtained with the reference solution. Ignore Related substances. A. Determine by thin-layer
any spot remaining on the line of application. chromatography (2.4.17), coating the plate with silica gel G.
B. Carry out test A but using a mixture of 85 volumes of Mobile phase. A mixture of 70 volumes of toluene, 20 volumes
cyclohexane and 15 volumes of diethylamine as the mobile of methanol, 5 volumes of glacial acetic acid and 5 volumes
phase and applying separately to the plate 30 µl of each of the of acetone.
test solution and the reference solution.
Test solution. Shake a quantity of the powdered tablets
Other tests. Comply with the tests stated under Parenteral containing 0.1 g of Verapamil Hydrochloride with 10 ml of
Preparations (Injections). dichloromethane, filter, wash the filter with a further 5 ml of
Assay. Dilute an accurately measured volume containing 5 mg dichloromethane, evaporate the combined filtrate and
of Verapamil Hydrochloride to 100.0 ml with 0.01 M washings to dryness and dissolve the residue in 2 ml of
hydrochloric acid and measure the absorbance of the resulting chloroform.
solution at the maximum at about 278 nm (2.4.7). Calculate the Reference solution. Dilute 1 volume of the test solution to
content of C 27H38N2O4, HCl taking 118 as the specific 100 volumes with chloroform and dilute 1 volume of the
absorbance at 278 nm. resulting solution to 10 volumes with chloroform.
Storage. Store protected from light. Apply to the plate 10 µl of each solution. After development,
dry the plate in air for 10 minutes and repeat the development.
Dry the plate at 110º for 30 minutes and allow to stand until the
Verapamil Tablets odour of solvent is no longer detectable. Spray with a solution
prepared by dissolving 5 g of ferric chloride hexahydrate
Verapamil Hydrochloride Tablets; Verapamil Chloride
and 2 g of iodine in sufficient of a mixture of equal volumes of
Tablets; Iproveratril Hydrochloride Tablets acetone and a 20 per cent w/v solution of (+)-tartaric acid to
Verapamil Tablets contain not less than 92.5 per cent and not produce 100 ml, applying a total of 15 to 20 ml of the reagent
more than 107.5 per cent of the stated amount of verapamil for a plate (20 cm x 20 cm), and examine immediately. Any
hydrochloride, C27H38N2O4, HCl. The tablets may be coated. secondary spot in the chromatogram obtained with the test

1228
IP 2007 VINBLASTINE SULPHATE

solution is not more intense than the spot in the chromatogram Identification
obtained with the reference solution. Ignore any spot remaining
on the line of application. A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with vinblastine
B. Carry out test A but using a mixture of 85 volumes of sulphate RS.
cyclohexane and 15 volumes of diethylamine as the mobile
B. In the Assay, the principal peak in the chromatogram
phase and applying separately to the plate 10 µl of each of the
obtained with the test solution corresponds to the peak due
test solution and the reference solution and heat at 110º for
to vinblastine sulphate in the chromatogram obtained with
90 minutes after the second development.
reference solution (b).
Other tests. Comply with the tests stated under Tablets.
C. A 10 per cent w/v solution gives the reaction of sulphates
Assay. Weigh and powder 20 tablets. Weigh accurately a (2.3.1).
quantity of the powder containing 0.1 g of Verapamil
Hydrochloride, shake with 150 ml of 0.1 M hydrochloric acid Tests
for 10 minutes, add sufficient 0.1 M hydrochloric acid to Appearance of solution. A 0.5 per cent w/v solution in carbon
produce 200.0 ml and filter. Dilute 10.0 ml of the filtrate to dioxide-free water is clear (2.4.1), and not more intensely
100.0 ml with water and measure the absorbance of the coloured than reference solution YS7 (2.4.1).
resulting solution at the maximum at about 278 nm (2.4.7).
Calculate the content of C27H38N2O4, HCl taking 118 as the pH (2.4.24). 3.5 to 5.0, determined in a 0.15 per cent w/v solution.
specific absorbance at 278 nm. Related substances. In the Assay, the area of any secondary
peak in the chromatogram obtained with the test solution is
not greater than that of the principal peak in the chromatogram
obtained with reference solution (c) and the sum of the areas
Vinblastine Sulphate of any such peaks is not greater than 2.5 times the area of the
principal peak in the chromatogram obtained with reference
solution (c). Ignore any peak with an area less than that of the
OH CH3 peak in the chromatogram obtained with reference solution (d).
N
Loss on drying (2.4.19). Not more than 15.0 per cent,
CH3 determined by Method B, on an appropriately calibrated
N H instrument using about 3.0 mg, accurately weighed. Heat the
H O
N COCH3 , H2SO4 substance under examination at the rate of 5º per minute
H H
H between ambient temperature and 200º in a current of nitrogen
H3COOC O for chromatography with a flow rate of 40 ml per minute. From
H3CO N HO the thermogram, determine the accumulated loss in weight
O
CH3 CH3 between ambient temperature and a point on the plateau before
decomposition is indicated (at about 160º).

C46H58N4O9,H2SO4 Mol. Wt. 909.1 Assay. Determine by liquid chromatography (2.4.14).


Test solution. Dissolve 0.1 g of the substance under
Vinblastine Sulphate is methyl (3aR,4R,5S,5aR,10bR,13aR)-
examination in 100 ml of water.
4-acetoxy-3a-ethyl-9-[(5S,7R,9S)-5-ethyl-5-hydroxy-9-
methoxycarbonyl-1,4,5,6,7,8,9,10-octahydro-2H-3,7- Reference solution (a). A solution containing 0.1 per cent
methanoazacycloundecino[(5,4-b)indol-9-yl]-6-formyl-5- w/v each of vinblastine sulphate RS and vincristine sulphate
hydroxy-8-methoxy-3a,4,5,5a,6,11,12,13a-octahydro-1H- RS in water.
indolizino[8,1-cd]carbazole-5-carboxylate sulphate. Reference solution (b). A 0.1 per cent w/v solution of
Vinblastine Sulphate contains not less than 95.0 per cent and vinblastine sulphate RS in water.
not more than 104.0 per cent of C46H58N4O9, H2SO4, calculated Reference solution (c). A 0.002 per cent w/v solution of
on the dried basis. vinblastine sulphate RS in water.
Description. A white or yellowish, amorphous or crystalline Reference solution (d). A 0.0001 per cent w/v solution of
powder; very hygroscopic. vinblastine sulphate RS in water.
CAUTION— Handle Vinblastine Sulphate with great care Chromatographic system
since it is a potent cytotoxic agent. Avoid contact with eyes; – a stainless steel column 25 cm x 4.6 mm, packed with
irritant to tissues. octylsilane bonded to porous silica (5 µm), (b) a guard

1229
VINBLASTINE INJECTION IP 2007

column packed with a suitable silica gel placed between The constituted solution complies with the requirements for
the pump and the injection device, Clarity of solution and Particulate matter stated under
– mobile phase: a mixture of 50 volumes of methanol, Parenteral Preparations (Injections).
38 volumes of a 1.5 per cent v/v solution of diethylamine Storage. The constituted solution should be used immediately
adjusted to pH 7.5 with phosphoric acid and 12 volumes after preparation but, in any case, within the period
of acetonitrile, recommended by the manufacturer.
– flow rate. 1 ml per minute,
– spectrophotometer set at 262 nm, Vinblastine Injection contains not less than 90.0 per cent and
– a 10 µl loop injector. not more than 110.0 per cent of the stated amount of
vinblastine sulphate, C46H58N4O9,H2SO4.
NOTE — Store all solutions in ice before use.
The contents of the sealed container comply with the
Inject each solution and record the chromatograms for 3 times
requirements stated under Parenteral Preparations
the retention time of the peak due to vinblastine.
(Powders for Injection) and with the following requirements.
The assay is not valid unless the resolution between the peaks
due to vincristine and vinblastine in the chromatogram Identification
obtained with reference solution (a) is at least 4 and the signal-
CAUTION — Handle Vinblastine Injection with great care
to-noise ratio of the peak in the chromatogram obtained with
since it is a potent cytotoxic agent. Avoid contact with eyes;
reference solution (d) is at least 5.
irritant to tissues.
Calculate the percentage content of C46H58N4O9, H2SO4.
A. When examined in the range 230 nm to 360 nm (2.4.7), the
Vinblastine Sulphate intended for use in the manufacture of solution obtained in the Assay shows an absorption maximum
parenteral preparations without a further appropriate at about 267 nm.
procedure for the removal of bacterial endotoxins complies
B. In the test for Related substances, the principal spot in the
with the following additional requirement.
chromatogram obtained with the test solution corresponds to
Bacterial endotoxins (2.2.3). Not more than 10.0 Endotoxin that in the chromatogram obtained with reference solution (a).
Units per mg.
C. To 1 ml add 0.2 ml of a freshly prepared 1 per cent w/v
Vinblastine Sulphate intended for use in the manufacture of solution of vanillin in hydrochloric acid; a pink colour is
parenteral preparations without a further appropriate produced in about 1 minute (distinction from Vincristine
sterilisation procedure complies with the following Sulphate).
additional requirement.
Tests
Sterility (2.2.11). Complies with the test for sterility.
Storage. Store protected from light and moisture in a deep pH (2.4.24). 3.5 to 5.0, determined in a 0.15 per cent w/v solution
freezer (Below -18º). If the material is intended for use in the of the dried contents.
manufacture of parenteral preparations, it should be stored in Related substances. Determine by thin-layer chromatography
sterile, tamper-evident glass containers and sealed so as to (2.4.17), coating the plate with silica gel GF254.
exclude micro-organisms.
Mobile phase. A mixture of 80 volumes of toluene, 40 volumes
Labelling. The label states whether or not the contents are of chloroform and 6 volumes of diethylamine.
suitable for use in the manufacture of parenteral preparations.
Test solution. Dissolve a quantity of the contents of a container
in sufficient methanol to produce a solution containing the
equivalent of 1 per cent w/v of dried vinblastine sulphate.

Vinblastine Injection Reference solution (a). A 1 per cent w/v solution of vinblastine
sulphate RS in methanol.
Vinblastine Sulphate Injection
Reference solution (b). A 0.02 per cent w/v solution of
Vinblastine Injection is a sterile material consisting of vincristine sulphate RS in methanol.
Vinblastine Sulphate with or without auxiliary substances. It
Apply to the plate 5 µl of each solution. After development,
is filled in a sealed container.
dry the plate in air and examine in ultraviolet light at 254 nm.
The injection is constituted by dissolving the contents of the Any secondary spot in the chromatogram obtained with the
sealed container in the requisite amount of sterile 0.9 per cent test solution is not more intense than the spot in the
w/v solution of Sodium Chloride, immediately before use. chromatogram obtained with reference solution (b).

1230
IP 2007 VINCRISTINE SULPHATE

Bacterial endotoxins (2.2.3). Not more than 10.0 Endotoxin B. In the Assay, the principal peak in the chromatogram
Units per mg of the dried contents. obtained with the test solution corresponds to the peak due
Assay. Weigh the contents of 20 sealed containers. Weigh to vincristine sulphate in the chromatogram obtained with
accurately a quantity of the mixed contents containing about reference solution (b).
20 mg of dried vinblastine sulphate and dissolve it in 100.0 ml C. A 10 per cent w/v solution gives the reaction for sulphates
of methanol. Dilute 10.0 ml to 100.0 ml with methanol and (2.3.1).
measure the absorbance of the resulting solution at the
maximum at about 267 nm (2.4.7). Calculate the content of Tests
C46H58N4O9,H2SO4 taking 185 as the specific absorbance at
267 nm. Appearance of solution. A 0.5 per cent w/v in carbon dioxide-
free water is clear (2.4.1), and not more intensely coloured
Storage. Store in sealed containers in a deep freezer (Below - than reference solution YS7 (2.4.1).
18º).
pH (2.4.24). 3.5 to 4.5. Determined in a 0.1 per cent w/v solution.
Labelling. The label states (1) the strength in terms of the
weight of dried vinblastine sulphate contained in it; (2) the Related substances. In the Assay, the area of any secondary
names of auxiliary substances, if any; (3) that the contents are peak in the chromatogram obtained with the test solution is
to be used by intravenous injection only; (4) the storage not greater than that of the principal peak in the chromatogram
conditions. obtained with reference solution (c) and the sum of the areas
of any such peaks is not greater than 2.5 times the area of the
principal peak in the chromatogram obtained with reference
Vincristine Sulphate solution (c). Ignore any peak with an area less than that of the
peak in the chromatogram obtained with reference solution
OH CH3 (d).
N
Loss on drying (2.4.19). Not more than 12.0 per cent,
CH3 determined by Method B, on an appropriately calibrated
N H instrument using about 3.0 mg, accurately weighed. Heat the
H O
N COCH3 , H2SO4 substance under examination at the rate of 5º per minute
H H
H between ambient temperature and 200º in current of nitrogen
H3COOC O for chromatography with a flow rate of 40 ml per minute.
H3CO N HO
O Assay. Determine by liquid chromatography (2.4.14).
CHO CH3
Test solution. Dissolve 0.1 g of the substance under
C46H56N4O10,H2SO4 Mol. Wt. 923.1 examination in 100 ml of water.
Vinblastine Sulphate is methyl (3aR,4R,5S,5aR,10bR,13aR) 4- Reference solution (a). A solution containing 0.1 per cent
acetoxy-3a-ethyl-9-[(5S,7R,9S)-5-ethyl-5-hydroxy-9- w/v each of vinblastine sulphate RS and vincristine sulphate
methoxycarbonyl-1,4,5,6,7,8,9,10-octahydro-2H-3,7- RS in water.
methanoazacycloundecino[(5,4-b)indol-9-yl]-5-hydroxy- 8-
Reference solution (b). A 0.1 per cent w/v solution of
methoxy-6-methyl-3a,4,5,5a,6,11,12,13a-octahydro-1H-
vincristine sulphate RS in water.
indolizino[8,1-cd]carbazole-5-carboxylate sulphate.
Vincristine Sulphate contains not less than 95.0 per cent and Reference solution (c). A 0.002 per cent w/v solution of
not more than 104.0 per cent of C46H56N4O10,H2SO4, calculated vincristine sulphate RS in water.
on the dried basis. Reference solution (d). A 0.0001 per cent w/v solution of
Description. A white to slightly yellowish, amorphous or vincristine sulphate RS in water.
crystalline powder; very hygroscopic. Chromatographic system
CAUTION— Handle Vincristine Sulphate with great care – a stainless steel column 25 cm x 4.6 mm, packed with
since it is a potent cytotoxic agent. Avoid contact with eyes; octylsilane bonded to porous silica (5 µm), (b) a guard
irritant to tissues. column packed with a suitable silica gel placed between
the pump and the injection device,
Identification – mobile phase: a mixture of 70 volumes of methanol and
A. Determine by infrared absorption spectrophotometry (2.4.6). 30 volumes of a 1.5 per cent v/v solution of diethylamine
Compare the spectrum with that obtained with vincristine adjusted to pH 7.5 with phosphoric acid,
sulphate RS. – flow rate. 1 ml per minute,

1231
VINCRISTINE INJECTION IP 2007

– spectrophotometer set at 297 nm, Vincristine Injection contains not less than 90.0 per cent and
– a 10 µl loop injector. not more than 110.0 per cent of the stated amount of vincristine
NOTE — Store all solutions in ice before use. sulphate, C46H56N4O10,H2SO4.
The contents of the sealed container comply with the
Inject each solution and record the chromatograms for 3 times
requirements stated under Parenteral Preparations
the retention time of the peak due to vincristine.
(Powders for Injection) and with the following requirements.
The assay is not valid unless the resolution between the peaks
due to vincristine and vinblastine in the chromatogram Identification
obtained with reference solution (a) is at least 4 and the signal-
to-noise ratio in the peak in the chromatogram obtained with CAUTION — Handle Vincristine Injection with great care
reference solution (d) is at least 5. since it is a potent cytotoxic agent. Avoid contact with eyes;
irritant to tissues.
Calculate the content of C46H56N4O10,H2SO4.
A. When examined in the range 230 nm to 360 nm (2.4.7), the
Vincristine Sulphate intended for use in the manufacture of solution obtained in the Assay shows an absorption maximum
parenteral preparations without a further appropriate at about 297 nm.
procedure for the removal of bacterial endotoxins complies
with the following additional requirement. B. In the test for Related substances, the principal spot in the
chromatogram obtained with the test solution corresponds to
Bacterial endotoxins (2.2.3). Not more than 62.5 Endotoxin
that in the chromatogram obtained with reference solution (b).
Units per mg.
C. Shake a quantity containing 1 mg of dried vincristine
Vincristine Sulphate intended for use in the manufacture of
sulphate with 3 ml of chloroform, filter and wash the filter with
parenteral preparations without a further appropriate
2 ml of chloroform. Reserve the residue for test D. Evaporate
sterilisation procedure complies with the following
the combined chloroform solutions to dryness at 40º. Add 0.2
additional requirement.
ml of a freshly prepared 1 per cent w/v solution of vanillin in
Sterility (2.2.11). Complies with the test for sterility. hydrochloric acid to the residue; an orange colour is produced
Storage. Store protected from light in a deep freezer (Below - in about 1 minute (distinction from vinblastine sulphate).
18º). If the material is intended for use in the manufacture of D. Dissolve the residue reserved in test C in 1 ml of water. The
parenteral preparations, it should be stored in sterile, tamper- solution complies with the following tests.
evident glass containers and sealed so as to exclude micro-
Heat 0.5 ml with 1 ml of potassium cupri-tartrate solution; a
organisms.
copius precipitate of copper oxide is produced.
Labelling. The label states whether or not the contents are
Heat 0.5 ml with 0.3 ml of a 6.5 per cent w/v solution of cupric
suitable for use in the manufacture of parenteral preparations.
acetate in a 1 per cent v/v solution of glacial acetic acid; no
precipitate is produced (distinction from fructose, glucose
and galactose).

Vincristine Injection Tests


Vincristine Sulphate Injection Appearance of solution. A solution prepared by dissolving
the contents of a sealed container in 10 ml of carbon dioxide-
Vincristine Injection is a sterile material consisting of Vincristine
free water is clear (2.4.1).
Sulphate with or without auxiliary substances. It is filled in a
sealed container. pH (2.4.24). 3.5 to 5.0, determined in a solution containing
0.15 per cent w/v solution of the dried contents.
The injection is constituted by dissolving the contents of the
sealed container in the requisite amount of sterile 0.9 per cent Related substances. Determine by thin-layer chromatography
w/v solution of Sodium Chloride, immediately before use. (2.4.17), coating the plate with silica gel GF254.
The constituted solution complies with the requirements for Mobile phase. A mixture of 80 volumes of toluene, 40 volumes
Clarity of solution and Particulate matter stated under of chloroform and 6 volumes of diethylamine.
Parenteral Preparations (Injections). Test solution. Dissolve a quantity of the contents of a container
Storage. The constituted solution should be used immediately in sufficient methanol (75 per cent) to produce a solution
after preparation but, in any case, within the period containing the equivalent of 1 per cent w/v of dried vincristine
recommended by the manufacturer. sulphate.

1232
IP 2007 VINORELBINE TARTRATE

Reference solution (a). A 0.02 per cent w/v solution of CAUTION – Vinorelbine Tartrate is cytotoxic; extra care
vinblastine sulphate RS in methanol. required to prevent inhaling particles and exposing the skin
Reference solution (b). A 1 per cent w/v solution of vincristine to it.
sulphate RS in methanol. Identification
Apply to the plate 5 µl of each solution. After development,
A. Dissolve 10 mg in 5 ml of water, add 0.5 ml of 5 M sodium
dry the plate in air and examine in ultraviolet light at 254 nm.
hydroxide, and extract with 5 ml of methylene chloride. Filter
Any secondary spot in the chromatogram obtained with the
the organic layer through anhydrous sodium sulphate, and
test solution is not more intense than the spot in the
evaporate to dryness.
chromatogram obtained with reference solution (a).
Determine by Infrared absorption spectrophotometry (2.4.6).
Bacterial endotoxins (2.2.3). Not more than 62.5 Endotoxin Compare the spectrum with that obtained with vinorelbine
Units per mg of dried vincristine sulphate. tartrate RS treated in the same manner.
Assay. Weigh the contents of 20 sealed containers. Weigh B. In the test for Related substances, the principal peak in the
accurately a quantity of the mixed contents of the containers chromatogram of the test solution corresponds to that due to
containing about 20 mg of dried vincristine sulphate and vinorelbine tartarate in the chromatogram obtained with the
dissolve it in 100.0 ml of methanol. Dilute 10.0 ml to 100.0 ml refrence solution.
with methanol and measure the absorbance of the resulting
solution at the maximum at about 297 nm (2.4.7). Calculate the C. It gives the reactions for tartrate (2.3.1).
content of C46H56N4O10,H2SO4 taking 177 as the specific Tests
absorbance at 297 nm.
Appearance of solution. A 1.0 per cent w/v solution is clear
Storage. Store in sealed containers in a deep freezer (Below -
(2.4.1).
18º).
Light absorption (2.4.7). The absorbance of 1.0 per cent w/v
Labelling. The label states (1) the strength in terms of the solution, at about 420 nm is not more than 0.03.
weight of dried vincristine sulphate contained in it; (2) the
names of auxilary substances, if any; (3) that the contents are pH (2.4.24). 3.3 to 3.8, determined on a 1.0 per cent w/v solution.
to be used by intravenous injection only; (4) the storage Related substances. Determine by liquid chromatography
conditions. (2.4.14).
Test solution. Dissolve 35 mg of the substance under
examination in 25 ml of mobile phase.
Vinorelbine Tartrate Reference solution (a). A 0.14 per cent w/v solution of
vinorelbine tartrate RS in mobile phase.
N CH3 Reference solution (b). Dilute 1 ml of the reference solution
(a) to 100 ml with mobile phase.
H
N Chromatographic system as described under Assay.
H
O N Inject reference solution (a). The test is not valid unless the
OCH3 H OH
COOH
H
CH3 , 2 HOOC column efficiency is not less than 2000 theoretical plates and
O OH
H3CO NH O
H the tailing factor is not more than 2.0.
H3C OH CH3
O OCH3 Inject the test solution and reference solution (b). In the
chromatogram obtained with the test solution, the area of any
C45H54N4O8, 2C4H6O6 Mol. Wt. 1079.11 secondary peak is not more than 0.3 times the area of the peak
in the chromatogram obtained with the reference solution (b)
Vinorelbine Tartrate is the ditartrate salt of vinorelbine, a
(0.3 per cent) and the sum of areas of all the secondary peaks
semisynthetic Vinca alkaloid; structurally relate to
is not more than the area of the peak in the chromatogram
vinblastine.
obtained with the reference solution (b)
Vinorelbine Tartrate contains not less than 98.0 per cent and (1.0 per cent). Ignore any secondary peak having area less
not more than 102.0 per cent of C45H54N4O8.2C4H6O6, calculated than 0.1 per cent.
on the anhydrous basis. Test solution. Weigh accurately about 0.2 g of the substance
Description. A white to yellow or light brown amorphous under examination, add 0.5 ml of water and 0.5 ml of N,N’-
powder. dimethyl formamide in to a 10 ml head space vial and seal it.

1233
VINORELBINE INJECTION IP 2007

Reference solution (a). To 50 ml of N,N’-dimethyl formamide, Chromatographic system


add 600 mg of methanol and 1000 mg of acetone, diluted to – a stainless steel column 15 cm x 3.9 mm, packed with
100 ml with N,N’-dimethyl formamide. octadecylsilane bonded to porous silica (5 µm),
Reference solution (b). To 50 ml of N,N’-dimethyl formamide, – column temperature 40°,
add 2400 mg of dichloromethane, 2880 mg of tetrahydrofuran – mobile phase: 62 volumes of methanol containing 1.22
and 240 mg of chloroform, diluted to 100 ml with N,N’-dimethyl g of sodium 1-decane sulphonate and 38 volumes of
formamide. Dilute 5 ml of the solution to 100 ml with N,N’- phosphate buffer solution, prepared by dissolving 6.9
dimethyl formamide. g of monobasic sodium phosphate in 900 ml of water,
adjust the pH to 4.2 with orthophosphoric acid and
Reference solution (c). Dilute 10 ml each of reference solutions dilute to 1000 ml with water,
(a) and (b) to 50 ml with N,N’-dimethyl formamide. Mix 0.5 ml – flow rate. 1 ml per minute,
of this solution with 0.5 ml of water in to a 10 ml head space – spectrophotometer set at 267 nm,
vial and seal it. – a 20 µl loop injector.
Chromatographic system Inject the reference solution. The test is not valid unless the
– a capillary column 30 m x 0.32 mm, coated with 1 per cent relative standard deviation for replicate injections is not more
vinyl and 5 per cent phenylmethylpolysiloxane (0.5 µm), than 2.0 per cent.
– temperature: Inject the test solution and reference solution.
column. 40º for 12 minutes increase @ 30º per minute to
220º hold for 5 minutes, Calculate the content of C45H54N4O8.2C4H6O6.
inlet port 120ºand detector. 250º, Storage. Store protected from light, at a temperature not
– nitrogen as carrier gas with a flow rate 0.5 ml per min. exceeding 25º.
Headspace conditions
Sample oven temperature 75º, Sample valve temperature 95º ,
Transfer line 100º, vial equilibrium 30 minutes, vial Vinorelbine Injection
Pressurisation 0.2 minute, sample loop fill 0.2 minute, loop
equilibrium 0.05 minute sample injection 1 minute. Vinorelbine Tartrate Injection
– a flame ionisation detector, Vinorelbine Injection is a sterile solution of vinorelbine tartrate
Inject 1 ml of the reference solution (c). The test is not valid in Water for Injection.
unless the resolution between two adjacent peaks is not less
than 1.5. Vinorelbine Injection contains not less than 90.0 per cent and
not more than 110.0 per cent of the stated amount of
Inject 1 ml of the test solution and reference solution (c). In
vinorelbine, C45H54N4O8.
the chromatogram obtained with test solution, the area of
peaks due to methanol, acetone, dichloromethane, chloroform Description. A clear, colourless to pale yellow solution.
and tetrahydrofuran is not more than the area of peaks
Identification
obtained in the chromatogram due to reference solution (c).
Sulphated ash (2.3.18). Not more than 0.1 per cent. A. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to the peak in the
Water (2.3.43). Not more than 4.0 per cent, determined on 0.1 g. chromatogram obtained with the reference solution.
Bacterial endotoxins (2.2.3). Not more than 1.5 Endotoxin Unit
B. When examined in the range 220 nm to 380 nm (2.4.7), a
per mg of vinorelbine base.
solution containing 0.01 per cent w/v of Vinorelbine Tartrate,
Microbial contamination (2.2.9). Total viable aerobic count, exhibits the maxima at about 267 nm.
not more than 100 cfu per g, total combined molds and yeast
does not exceed 50 cfu per g. It also meets the requirement of Tests
the tests for the absence of Staphylococcus aureus, pH (2.4.24). 3.0 to 3.8.
Pseudomonas aeruginosa, Salmonella species, Escherichia
coli, Enterobacteria and Closteridia. Light absorption. The absorbance of the injection at about
420 nm (2.4.7), is not more than 0.06.
Assay. Determine by liquid chromatography (2.4.14).
Related substances. Determine by liquid chromatography
Test solution. Dissolve 35 mg of the substance under (2.4.14).
examination in 25.0 ml of mobile phase.
Test solution. Accurately measured volume of injection
Reference solution. A 0.14 per cent w/v solution of vinorelbine containing 10 mg of Vinorelbine Tartrate, dilute to 10 ml with
tartrate RS in mobile phase. mobile phase.

1234
IP 2007 VITAMIN A CONCENTRATE OIL

Reference solution (a). A 0.1 per cent w/v solution of Vitamin A Concentrate Oil
vinorelbine tartrate RS in mobile phase.
Synthetic Vitamin A Concentrate (Oily Form); Synthetic
Reference solution (b). Dilute 1 ml of the reference solution
Retinol Concentrate (Oily Form)
(a) to 100 ml with mobile phase.
Vitamin A Concentrate Oil consists of an ester or a mixture of
Chromatographic system as described under Assay.
esters of retinol (as acetate, propionate or palmitate) prepared
Inject reference solution (a). The test is not valid unless the by synthesis. It may be diluted with a suitable vegetable oil. It
column efficiency is not less than 2000 theoretical plates and may contain suitable stabilising agents such as antioxidants.
the tailing factor is not more than 2.0.
Vitamin A Concentrate Oil contains not less than 500,000 Units
Inject the test solution and reference solution (b). Run the of Vitamin A per g, and not less than 95.0 per cent and not
chromatograms three times the retention time of the peak due more than 110.0 per cent of the stated number of Units of
to vinorelbine. In the chromatogram obtained with the test Vitamin A per g.
solution, the area of any secondary peak is not more than 0.2
Description. A yellow to brownish yellow, oily liquid; odour,
times the area of the peak in the chromatogram obtained with
faint and characteristic.
the reference solution (b) (0.2 per cent) and the sum of areas
of all the secondary peaks is not more than twice the area of Identification
the peak in the chromatogram obtained with the reference
solution (b) (2.0 per cent). A. When examined in the range 230 nm to 360 nm (2.4.7), a
solution in 2-propanol containing 10 to 15 Units per ml shows
Other tests. Complies with the tests stated under Parenteral an absorption maximum at about 325 nm to 327 nm.
Preparation (Injections).
B. Determine by thin-layer chromatography (2.4.17), coating
Bacterial endotoxins (2.2.3). Not more than 3.0 Endotoxin Unit the plate with silica gel G.
per mg of vinorelbine tartrate.
Mobile phase. A mixture of 80 volumes of cyclohexane and
Sterility (2.2.11). Complies with the test for sterility. 20 volumes of ether.
Assay. Determine by liquid chromatography (2.4.14). Test solution. Prepare a solution containing 5 Units per µl of
Test solution. Accurately measured volume of injection the substance under examination in cyclohexane.
containing 10 mg of Vinorelbine Tartrate, diluted to 100.0 ml Reference solution (a). Prepare a solution containing 5 Units
with water. per µl of retinyl acetate RS in cyclohexane.
Reference solution. A 0.1 per cent w/v solution of vinorelbine Reference solution (b). Prepare a solution containing 5 Units
tartrate RS in water. per µl of retinyl propionate RS in cyclohexane.
Chromatographic system Reference solution (c). Prepare a solution containing 5 Units
– a stainless steel column 15 cm x 3.9 mm, packed with per µl of retinyl palmitate RS in cyclohexane.
octadecylsilane bonded to porous silica (5 µm),
Apply to the plate 2 µl of each solution. After development,
– column temperature 40°,
dry the plate in air, spray with antimony trichloride solution.
– mobile phase: a mixture of 62 volumes of methanol
The principal spot or spots in the chromatogram obtained
containing 1.22 g of sodium1-decane sulphonate and
with the test solution correspond to one or more of the spots
38 volumes of phosphate buffer solution prepared by
in the chromatograms obtained with reference solutions (a),
dissolving 6.9 g of monobasic sodium phosphate in
(b) and (c).
900 ml of water adjusted the pH to 4.2 with
orthophosphoric acid and dilute to 1000 ml with water, C. Dissolve a quantity containing 10 to 15 Units in 1 ml of
– flow rate. 1 ml per minute, chloroform and add 5 ml of antimony trichloride solution; a
– spectrophotometer set at 267 nm, transient bright blue colour is produced immediately.
– a 20 µl loop injector.
Tests
Inject the reference solution. The test is not valid unless the
relative standard deviation is not more than 2.0 per cent. Acid value (2.3.23). Not more than 2.0, determined on 2.0 g.
Inject the test solution and the reference solution. Peroxides. Add 0.3 g to 25.0 ml of a mixture of 6 volumes of
Calculate the content of C45H54N4O8. toluene and 4 volumes of methanol (solution A). Add in a
test-tube, in the following order, mixing after each addition,
Storage. Store protected from light, in single-dose container. 0.3 ml of a 1.8 per cent w/v solution of ammonium thiocyanate,

1235
VITAMIN A CONCENTRATE POWDER IP 2007

10.0 ml of methanol, 0.3 ml of ferrous sulphate solution and A. When examined in the range 230 nm to 360 nm (2.4.7), a
15.0 ml of toluene and add 1.0 ml of solution A. The colour solution in 2-propanol containing 10 to 15 Units per ml shows
produced after 5 minutes is not more intense than that obtained an absorption maximum at about 325 nm to 327 nm.
in a solution prepared at the same time and in the same manner
B. Determine by thin-layer chromatography (2.4.17), coating
but using a solution prepared in the following manner in place
the plate with silica gel G.
of solution A. Add 1.0 ml of a 27.0 per cent w/v solution of
ferric chloride hexahydrate to 99 ml of a mixture of 6 volumes Mobile phase. A mixture of 80 volumes of cyclohexane and
of toluene and 4 volumes of methanol and dilute 2.0 ml to 20 volumes of ether.
100.0 ml with the same solvent mixture.
Test solution. Evaporate 10 ml of solution A to dryness in a
Assay. Carry out the assay of vitamin A, Method A (2.3.41). current of nitrogen and dissolve the residue in 0.5 ml of
cyclohexane.
Storage. Store protected from light, in well-filled containers.
Once the container has been opened, its contents should be Reference solution (a). Prepare a solution containing 5 Units
used as soon as possible; any part of the contents not used at per µl of retinyl acetate RS in cyclohexane.
once should be protected by an atmosphere of an inert gas.
Reference solution (b). Prepare 5 Units per µl of retinyl
Labelling. The label states (1) the number of Units of Vitamin propionate RS in cyclohexane.
A per g; (2) the name of the ester or esters; (3) the name(s) of
Reference solution (c). Prepare a solution containing 5 Units
any added stabilising agent(s); (4) the method of restoring
per µl of retinyl palmitate RS in cyclohexane.
the solution if partial crystallisation has occurred.
Apply to the plate 2 µl of each solution. After development,
dry the plate in air, spray with antimony trichloride solution.
The principal spot or spots in the chromatogram obtained
with the test solution correspond to one or more of the spots
Vitamin A Concentrate Powder in the chromatograms obtained with reference solutions (a),
(b) and (c).
Synthetic Vitamin A Concentrate (Powder Form);
Synthetic Retinol Concentrate (Powder Form) C. Dilute 2 ml of solution A to 50 ml with n-pentane and
evaporate 1 ml of the solution to dryness in a current of
Vitamin A Concentrate Powder consists of an ester or a mixture nitrogen. Dissolve the residue in 1 ml of chloroform and add
of esters of retinol (as acetate, propionate or palmitate) prepared 5 ml of antimony trichloride solution; a transient bright blue
by synthesis and dispersed in a matrix of Gelatin, Acacia or colour is produced immediately.
any other suitable material. It may contain suitable stabilising
agents such as antioxidants. Tests
Vitamin A Concentrate Powder contains not less than Related substances and degradation products. Using the
250,000 Units of Vitamin A per g, and not less than 95.0 per relative absorbances obtained in the Assay, the ratio A300/
cent and not more than 115.0 per cent of the stated number of A325 is not more than 0.612 and the sum of the ratios A300/A325
Units of Vitamin A per g. and A350/A325 is not more than 1.054, where A300, A325 and A350
Description. A yellowish powder usually in the form of pellets are the absorbances measured at about 300 nm, 325 nm and
or particles of almost uniform size. 350 nm respectively.
Assay. Carry out the assay of vitamin A, Method B (2.3.41),
Identification adding 5 ml of water to the mixture for saponification, using
2-propanol as the blank and taking 0.612 as the maximum
To a quantity containing 50,000 Units of Vitamin A add 1.5 ml
value of the ratio A300/A325.
of 2 M ammonia previously heated to 60º and heat in a water-
bath at 60º, shaking occasionally. After 10 minutes add 40 ml Storage. Store protected from light. Once the container has
of ethanol, dilute to 200 ml with ether and shake. Allow to been opened, its contents should be used as soon as possible;
stand for a few minutes and use the supernatant liquid any part of the contents not used at once should be protected
(solution A) for the following tests. by an atmosphere of an inert gas.
Certain samples may not react sufficiently during the course Labelling. The label states (1) the number of Units of Vitamin
of the above treatment. In such cases the volume of solution A per g; (2) the name of the ester or esters; (3) the name of the
A used in the following tests should be increased which may principal excipient or excipients used; (4) the name of any
be as much as 10-fold. added stabilising agents.

1236
IP 2007 VITAMIN A AND D CAPSULES

Water-Miscible Vitamin A Concentrate in the chromatograms obtained with reference solutions (a),
(b) and (c).
Synthetic Vitamin A Concentrate (Water-dispersible
C. Evaporate 0.1 ml of solution A to dryness in a current of
Form); Synthetic Retinol Concentrate (Water-dispersible nitrogen, dissolve the residue in 1 ml of chloroform and add
Form) 5 ml of antimony trichloride solution; a transient bright blue
Water-miscible Vitamin A Concentrate consists of an ester or colour is produced immediately.
a mixture of esters of retinol (as acetate, propionate or palmitate)
prepared by synthesis to which suitable solubilisers have Tests
been added. It may contain suitable stabilising agents such Water miscibility. Mix about 1 g with 10 ml of water previously
as antimicrobial preservatives and antioxidants. heated to 50º and cool to 20º. Immediately after cooling, a
Water-miscible Vitamin A Concentrate contains not less than uniform, slightly opalescent and slightly yellow dispersion is
100,000 Units of Vitamin A per g, and not less than 95.0 per obtained.
cent and not more than 115.0 per cent of the stated number of Related substances and degradation products. Using the
Units of Vitamin A per g. relative absorbances obtained in the Assay, the ratio A300/
Description. A yellow or yellowish liquid of variable A325 is not more than 0.618 and the sum of the ratios A300/A325
opalescence and viscosity; odour, characteristic. Highly and A350/A325 is not more than 1.060, where A300, A325 and A350
concentrated solutions may become cloudy at low are the absorbances measured at about 300 nm, 325 nm and
temperatures or gel at room temperature. 350 nm respectively.
Assay. Carry out the assay of vitamin A, Method B (2.3.41),
Identification using 2-propanol as the blank and taking 0.618 as the maximum
To a quantity containing about 10,000 Units of Vitamin A add value of the ratio A300/A325.
5 ml of water and homogenise. Add 5 ml of ethanol (95 per Storage. Store protected from light at the temperature stated
cent) and 20 ml of n-pentane and shake vigorously for on the label. Once the container has been opened, its contents
30 seconds. Allow to stand for a few minutes and use the should be used as soon as possible; any part of the contents
supernatant liquid (solution A) for the following tests. not used at once should be protected by an atmosphere of an
A. Dilute solution A with sufficient 2-propanol so that the inert gas.
absorbance at the wavelength of maximum absorption is Labelling. The label states (1) the number of Units of Vitamin
0.3 to 0.7 (2.4.7). A per g; (2) the name of the ester or esters; (3) the name of the
When examined in the range 230 nm to 360 nm (2.4.7), the principal excipient or excipients used; (4) the temperature at
solution shows an absorption maximum at 325 to 327 nm. which it should be stored.

B. Determine by thin-layer chromatography (2.4.17), coating


the plate with silica gel G.
Vitamins A And D Capsules
Mobile phase. A mixture of 80 volumes of cyclohexane and
20 volumes of ether. Vitamins A and D Capsules contain Vitamin A Oil and a source
of vitamin D such as Cholecalciferol or Ergocalciferol in an
Test solution. Evaporate 10 ml of solution A to dryness in a edible vegetable oil.
current of nitrogen and dissolve the residue in 0.5 ml of
cyclohexane. Vitamins A and D Capsules contain not less than 90.0 per cent
of the stated number of Units of vitamin A and vitamin D.
Reference solution (a). Prepare a solution containing 5 Units
per µl of retinyl acetate RS in cyclohexane. Tests
Reference solution (b). Prepare a solution containing 5 Units Other tests. Comply with the tests stated under Capsules.
per µl of retinyl propionate RS in cyclohexane.
Assay. For vitamin A — Weigh accurately a portion of the
Reference solution (c). Prepare a solution containing 5 Units mixed contents of 20 capsules containing about 500 Units of
per µl of retinyl palmitate RS in cyclohexane. Vitamin A and carry out the assay of vitamin A, Method A
Apply to the plate 2 µl of each solution. After development, (2.3.41).
dry the plate in air, spray with antimony trichloride solution. For vitamin D — Weigh accurately a portion of the mixed
The principal spot or spots in the chromatogram obtained contents of 20 capsules containing about 5000 Units of vitamin
with the test solution correspond to one or more of the spots D and carry out the assay of vitamin D (2.3.42).

1237
CONCENTRATED VITAMIN D SOLUTION IP 2007

Storage. Store protected from light and moisture. Labelling. The label states (1) the number of Units of vitamin
Labelling. The label states the number of Units of vitamin A D per g; (2) the storage conditions; (3) the nature and
and vitamin D per capsule. concentration of any stabilising agent added.

Concentrated Vitamin D Solution Concentrated Vitamins A And D


Concentrated Vitamin D Solution is a solution of
Solution
Cholecalciferol or Ergocalciferol in an edible vegetable oil. It Concentrated Vitamins A and D Solution is a solution of
may contain suitable stabilising agents such as antioxidants. Vitamin A Oil and a source of vitamin D such as Cholecalciferol
or Ergocalciferol in an edible vegetable oil. It may contain
Concentrated Vitamin D Solution contains not less than 10,000
suitable stabilising agents such as antioxidants.
Units of vitamin D and not less than 90.0 per cent of the stated
number of Units of vitamin D. Concentrated Vitamins A and D Solution contains not less
than 50,000 Units of vitamin A and 5000 Units of vitamin D per
Description. A pale yellow to yellow, oily liquid; odour, faint
g, and not less than 90.0 per cent and not more than 110.0 per
but not rancid.
cent of the stated number of Units of Vitamin A and vitamin D
Identification per g.

Dissolve a quantity containing about 1000 Units of vitamin D Tests


in 1 ml of chloroform and add 10 ml of antimony trichloride
Acid value (2.3.23). Not more than 2.5, determined on 2.0 g.
solution; a pinkish red colour appears at once.
Assay. For vitamin A — Carry out the assay of vitamin A,
Tests Method A (2.3.41).
Acid value (2.3.23). Not more than 2.5, determined on 2.0 g. For vitamin D — Carry out the assay of vitamin D (2.3.42).
Assay. Carry out the assay of vitamin D (2.3.42). Storage. Store protected from light and moisture.
Storage. Store protected from light, in well-filled containers at Labelling. The label states (1) the number of Units of vitamin
a temperature of 6º to 15º. The contents of an opened container A and vitamin D per gram; (2) the storage conditions.
should be used as soon as possible.

1238
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

W
Warfarin Sodium ....
Warfarin Sodium Clathrate ....
Warfarin Tablets ....
Purified Water ....
Water For Injections ....
Water For Injections in Bulk ....
Sterile Water For Injections ....
Wool Fat ....
Hydrous Wool Fat ....

1239
IP 2007 WARFARIN SODIUM CLATHRATE

Warfarin Sodium maximum at about 385 nm, measured within 15 minutes of


preparation, is not more than 0.20 (2.4.7).
O O Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Mobile phase. A mixture of 50 volumes of chloroform,
NaO CH3 50 volumes of cyclohexane and 20 volumes of glacial acetic
acid.
O
Test solution (a). Dissolve 0.2 g of the substance under
C19H15NaO4 Mol. Wt. 330.3 examination in 10 ml of acetone.
Warfarin Sodium is sodium 2-oxo-3-[(1RS)-3-oxo-1- Test solution (b). Dissolve 0.4 g of the substance under
phenylbutyl]-2H-1-benzopyran-4-olate. examination in 100 ml of acetone.
Warfarin Sodium contains not less than 98.0 per cent and not Reference solution (a). A 0.002 per cent w/v solution of the
more than 102.0 per cent of C19H15NaO4, calculated on the substance under examination in acetone.
anhydrous basis. Reference solution (b). A 0.4 per cent w/v solution of warfarin
Description. A white powder; hygroscopic. sodium RS in acetone.
Reference solution (c). A solution containing 0.1 per cent
Identification w/v of acenocoumarol RS and 0.2 per cent w/v of the
Test A may be omitted if tests B, C, D and E are carried out. substance under examination in acetone.
Tests B and D may be omitted if tests A, C and E are carried Apply to the plate 20 µl of each solution. After development,
out. dry the plate in air and examine in ultraviolet light at 254 nm.
A. Dissolve 1 g in 25 ml of water, add 2 ml of 2 M hydrochloric Any secondary spot in the chromatogram obtained with test
acid and filter. Wash the residue with water and dry over solution (a) is not more intense than the spot in the
phosphorus pentoxide. chromatogram obtained with reference solution (a). The test
is not valid unless the chromatogram obtained with reference
On the residue, determine by infrared absorption solution (c) shows two clearly separated spots and the
spectrophotometry (2.4.6). Compare the spectrum with that chromatogram obtained with reference solution (a) shows a
obtained with warfarin sodium RS or with the reference clearly visible spot.
spectrum of warfarin sodium.
Water (2.3.43). Not more than 4.0 per cent, determined on
B. In the test for Related substances, the principal spot in the 0.75 g.
chromatogram obtained with test solution (b) corresponds to
that in the chromatogram obtained with reference solution Assay. Weigh accurately about 0.1 g and dissolve in sufficient
(b). 0.01 M sodium hydroxide to produce 100.0 ml. Dilute 10.0 ml
to 100.0 ml with 0.01 M sodium hydroxide and dilute 5.0 ml to
C. Dissolve 1 g in 10 ml of water, add 5 ml of nitric acid and 50.0 ml with 0.01 M sodium hydroxide. Measure the
filter. To the filtrate add 2 ml of potassium dichromate solution, absorbance of the resulting solution at the maximum at about
shake for 5 minutes and allow to stand for 20 minutes; the 308 nm (2.4.7). Calculate the content of C19H15NaO4 taking 431
solution is not greenish blue when compared with a blank. as the specific absorbance at 308 nm.
D. The residue obtained in test A, after washing with water Storage. Store protected from light and moisture.
and drying at 105º, melts at 159° to 163° (2.4.21).
E. The filtrate obtained in test A gives the reactions of sodium
salts (2.3.1). Warfarin Sodium Clathrate
Tests Warfarin Sodium Clathrate is a clathrate form of Warfarin
Appearance of solution. A 5.0 per cent w/v solution is clear Sodium consisting principally of Warfarin Sodium and
(2.4.1), and colourless (2.4.1). Isopropyl Alcohol in a 2.1 molecular ratio.

pH (2.4.24). 7.6 to 8.6, determined in a 1.0 per cent w/v solution. Warfarin Sodium Clathrate contains not less than 97.0 per
cent and more than 102.0 per cent of C19H15NaO4 calculated on
Phenolic ketones. Absorbance of a 12.5 per cent w/v solution the anhydrous, isopropyl alcohol-free basis and not less than
in a 5.0 per cent w/v solution of sodium hydroxide at the 8.0 per cent and not more than 8.5 per cent of isopropy alcohol.

1241
WARFARIN SODIUM CLATHRATE IP 2007

Description. A white crystalline powder; hygroscopic. Reference solution (c). A solution containing 0.1 per cent
w/v of acenocomarol RS and 0.2 per cent w/v of the substance
Identification under examination in acetone.
Test A may be omitted if test B, C, D and E are carried out. Apply to the plate 20 µl of each solution. After development,
Tests B and D may be omitted if tests A, C and E are carried dry the plate in air and examine in ultraviolet light at 254 nm.
out. Any secondary spot in the chromatogram obtained with test
A. Dissolve about 1 g in 25 ml of water, add 2 ml of 2 M solution (a) is not more intense than the spot in the
hydrochloric acid and filter. Wash the precipitate 5 to 6 times chromatogram with reference solution (a). The test in not valid
with water. Dry the residue over phosphorus pentoxide. unless the chromatogram obtained with reference solution (c)
show two clearly separated spots and the chromatogram
On the residue, determine by infrared absorption obtained with reference solution (a) shows a clearly visible
spectrophotometry (2.4.6). Compare the spectrum with that spot.
obtained with warfarin sodium RS or with the reference
spectrum of warfarin sodium. Water (2.3.43). Not more than 4.0 per cent, determined on
0.75 g.
B. In the test for Related substances, the principal spot in the
chromatogram obtained with test solution (b) corresponds to Isopropyl alcohol. Determine by gas chromatography (2.4.13).
that in the chromatogram obtained with reference solution (b).
Test solution. Dissolve 0.5 g of the substance under
C. Dissolve 1 g in 10 ml of water, add 5 ml of nitric acid and examination in sufficient water to produce 10 ml.
filter. To the filtrate add 2 ml of potassium dichromate solution,
shake for 5 minutes and allow to stand for 20 minutes; the Reference solution (a). A solution containing 5.0 per cent
solution is not greenish-blue when compared with a blank. w/v of the substance under examination and 0.5 per cent v/v
of propan-I-ol (internal standard).
D. The residue obtained in test A, after washing with water
and drying at 105° melts at 159° to 163°. Reference solution (b). A solution containing 0.5 per cent v/v
each of propan-2-ol and the internal standard.
E. The filtrate obtained in test A gives the reactions of sodium
salts (2.3.1). Chromatographic system
– a glass column 1.5 m x 4 mm, packed with porous polymer
Tests beads (125 to 150 mm) (such as Porapak Q),
– temperature:
Appearance of solution. A 5.0 per cent w/v solution is clear
column 150°,
(2.4.1), and colourless (2.4.1).
inlet port at 180° and detector at 200°,
pH (2.4.24). 7.2 to 8.3, determined in a 1.0 per cent w/v solution. – flow rate. 40 ml per minute of the carrier gas.
Phenolic ketones. Absorbance of a 12.5 per cent w/v solution The column temperature may be varied so that the resolution,
in a 5.0 per cent w/v solution of sodium hydroxide at the R, between propan-I-ol and propanol-2 ol is not less than 2.0,
maximum at about 385 nm, measured within 15 minutes of the tailing factor. T, for the propan-2-ol is not less than 2.0 the
preparation, is not more than 0.20 (2.4.7). tailing factor, T, for the propan-2-ol peak is not more than 1.5
Related substances. Determine by thin-layer chromatography and the relative standard deviation of the ratio of the area due
(2.4.17), coating the plate with silica gel GF254. to the peak of propanol-2-ol to that due to propan-1-ol for five
Mobile phase. A mixture of 50 volumes of chloroform, replicate injections of reference solution (b) is not more than
50 volumes of cyclohexane and 20 volumes of glacial acetic 2.0 per cent.
acid. Calculate the content of isopropyl alcohol.
Test solution (a). Dissolve 0.2 g of the substance under Assay. Weigh accurately about 0.1 g and dissolve in sufficient
examination in 10 ml of acetone. 0.01 M sodium hydroxide to produce 100.0 ml. Dilute 10.0 ml
Test solution (b). Dissolve 0.4 g of the substance under to 100.0 ml with 0.01 M sodium hydroxide and dilute 5.0 ml to
examination in 100 ml of acetone. 50.0 ml with 0.01 M sodium hydroxide. Measure the
absorbance of the resulting solution at the maximum at about
Reference solution (a). A 0.002 per cent w/v solution of the
308 nm (2.4.7). Calculate the content of C19H15NaO4 taking 431
substance under examination in acetone.
as the specific absorbance at 308 nm.
Reference solution (b). A 0.4 per cent w/v solution of warfarin
sodium RS in acetone. Storage. Store protected from light and moisture.

1242
IP 2007 WARFARIN TABLETS

Warfarin Tablets the chromatograms obtained with reference solutions (b) and
(c) respectively and any other secondary spot is not more
Warfarin Sodium Tablets intense than the spot in the chromatogram obtained with
Warfarin Tablets contain not less than 92.5 per cent and not reference solution (a).
more than 107.5 per cent of the stated amount of warfarin Uniformity of content. Comply with the test stated under
sodium, C19H15NaO4. Tablets.
Determine by liquid chromatography (2.4.14).
Identification
Test solution. Shake one tablet with 10 ml of 0.01 M sodium
A. Extract a quantity of the powdered tablets containing 0.1 g hydroxide for 15 minutes, add 10 ml of a 2 per cent v/v solution
of Warfarin Sodium with 30 ml of water, add 0.1 ml of 2 M of glacial acetic acid in acetonitrile, centrifuge for 10 minutes
hydrochloric acid, filter, wash the precipitate with water and and use the clear supernatant liquid.
dry. Warm the residue gently with 3 ml of ethanol (95 per
cent), filter and add the filtrate to 25 ml of water containing Chromatographic system
0.1 ml of 2 M hydrochloric acid. Filter, wash the precipitate – a stainless steel column 10 cm x 4.6 mm, packed with
with water and dry it at 105°. octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 55 volumes of acetonitrile,
On the residue, determine by infrared absorption 45 volumes of water and 1 volume of glacial acetic
spectrophotometry (2.4.6). Compare the spectrum with that acid,
obtained with warfarin sodium RS or with the reference – flow rate. 2 ml per minute,
spectrum of warfarin sodium. – spectrophotometer set at 283 nm,
B. The final residue obtained in test A melts at about 159° – a 20 µl loop injector.
(2.4.21). Determine the content of C19H15NaO4 in the tablet.

Tests Dissolution (2.5.2).


Apparatus. No 1
Related substances. Determine by thin-layer chromatography
Medium. 900 ml of a 0.68 per cent w/v solution of potassium
(2.4.17), coating the plate with silica gel GF254.
dihydrogen phosphate with the pH adjusted to 6.8 by the
Mobile phase. A mixture of 50 volumes of chloroform, addition of 1 M sodium hydroxide
50 volumes of cyclohexane and 20 volumes of glacial acetic Speed and time. 100 rpm and 45 minutes.
acid.
For tablets containing 2 mg or less of warfarin sodium, use
Test solution. Shake a quantity of the powdered tablets three tablets for each test; for tablets containing more than 2
containing 40 mg of Warfarin Sodium with 30 ml of water for mg of warfarin sodium, use a single tablet for each test.
15 minutes, add 0.1 ml of hydrochloric acid and extract with
Withdraw a suitable volume of the medium and filter. Measure
three quantities, each of 10 ml, of chloroform, drying each
the absorbance of a layer of suitable thickness of the filtrate,
extract with anhydrous sodium sulphate. Evaporate the
suitably diluted if necessary, at the maxima at about 307 nm
combined extracts at a temperature not exceeding 40° and
and 360 nm (2.4.7), and calculate the difference between the
dissolve the residue in 2 ml of acetone.
two absorbances (DA). Calculate the total content of warfarin
Reference solution (a). Dilute 1 volume of the test solution to sodium, C19H15NaO4, in the medium taking 428 as the specific
200 volumes with acetone. absorbance value of DA.
Reference solution (b). A 0.002 per cent w/v solution of (E)-4- D. Not less than 70 per cent of the stated amount of
phenylbut-3-en-2-one in acetone. C19H15NaO4.
Reference solution (c). A 0.02 per cent w/v solution of Other tests. Comply with the tests stated under Tablets.
4-hydroxycoumarin in acetone.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Apply to the plate 20 µl of each solution. After development, quantity of the powder containing about 20 mg of Warfarin
dry the plate in air and examine immediately in visible light Sodium and shake with 250.0 ml of 0.01 M sodium hydroxide
noting the position of any coloured spots and then examine in for 15 minutes and filter. To 20.0 ml of the filtrate add 0.15 ml of
ultraviolet light at 254 nm, ignoring any spot that was noted in hydrochloric acid and extract with three quantities, each of
visible light. Any spots corresponding to (E)-4-phenylbut-3- 15 ml, of chloroform. Extract the combined chloroform layers
en-2-one and 4-hydroxycoumarin in the chromatogram obtained with three quantities, each of 20 ml, of 0.01 M sodium hydroxide.
with the test solution are not more intense than the spots in Dilute the combined aqueous layers to 100.0 ml with 0.01 M

1243
PURIFIED WATER IP 2007

sodium hydroxide, filter and measure the absorbance of the Nitrates. To 5 ml in a test-tube immersed in ice add 0.4 ml of a
resulting solution at the maximum at about 307 nm (2.4.7). 10 per cent w/v solution of potassium chloride, 0.1 ml of
Calculate the content of C19H15NaO4 taking 431 as the specific diphenylamine solution and, dropwise with shaking, 5 ml of
absorbance at 307 nm. sulphuric acid. Transfer the tube to a water-bath at 50° and
Storage. Store protected from light. allow to stand for 15 minutes. Any blue colour in the solution
is not more intense than that in a solution prepared at the
same time and in the same manner using a mixture of 4.5 ml of
nitrate-free water and 0.5 ml of nitrate standard solution
(2 ppm NO3) (0.2 ppm).
Purified Water
Sulphates (2.3.17). To 10 ml add 0.1 ml of 2 M hydrochloric
H2O Mol. Wt. 18.0 acid and 0.1 ml of barium chloride solution. The appearance
Purified Water is prepared by distillation, by means of ion of the solution does not change for at least 1 hour.
exchange or by any other appropriate means from suitable Oxidisable substances. To 100 ml add 10 ml of 1 M sulphuric
potable water that complies with all relevant statutory acid and 0.1 ml of 0.02 M potassium permanganate and boil
regulations. for 5 minutes; the solution remains faintly pink.
During production and subsequent storage, it is recommended Residue on evaporation. Evaporate 100 ml to dryness on a
that adequate measures are taken to ensure that the microbial water-bath and dry to constant weight at 105°. The residue
quality is controlled and monitored. Appropriate alert and weighs not more than 1 mg (0.001 per cent).
action limits are set so as to detect adverse trends. Under
controlled conditions, an appropriate action limit is a total Purified Water intended for use in the manufacture of dialysis
viable count (2.2.9) of 100 micro-organisms per ml, determined solutions and also without a further procedure for the
by membrane filtration. In addition, the test for oxidisable removal of bacterial endotoxins complies with the following
substances (given below) is carried out. The adequacy of additional requirements.
these measures may be determined by carrying out the test Aluminium (2.3.8). Not more than 10 ppb, determined using
for conductivity (2.4.9) off-line or in-line. the following solutions.
Description. A clear, colourless liquid; odourless and tasteless. Test solution. To 400 ml of the water under examination add
10 ml of acetate buffer solution pH 6.0 and 100 ml of distilled
Tests water.
Acidity or alkalinity. To 10 ml, freshly boiled and cooled in a Reference solution. Mix 2 ml of aluminium standard solution
borosilicate glass flask, add 0.05 ml of methyl red solution; (2 ppm Al), 10 ml of acetate buffer solution pH 6.0 and 98 ml
the resulting solution is not red. To 10 ml add 0.1 ml of of distilled water.
bromothymol blue solution; the resulting solution is not blue.
Blank solution. Mix 10 ml of acetate buffer solution pH 6.0
Ammonium. To 20 ml add 1 ml of alkaline potassium mercuri- and 100 ml of distilled water.
iodide solution, and allow to stand for 5 minutes. When
viewed vertically the solution is not more intensely coloured Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
than a solution prepared at the same time by adding 1 ml of Unit per ml.
alkaline potassium mercuri-iodide solution to a mixture of Storage. Store protected from light.
4.0 ml of ammonium standard solution (1 ppm NH4) and
16.0 ml of ammonia-free water (0.2 ppm).
Calcium and magnesium. To 100 ml add 2 ml of ammonia
buffer pH 10.0, 50 mg of mordant black II mixture and 0.5 ml
of 0.01 M disodium edetate; a pure blue colour is produced.
Water For Injections
Heavy metals (2.3.13). Evaporate 150 ml to 15 ml on a water-
bath; 12 ml of the solution complies with the limit test for H2O Mol. Wt. 18.0
heavy metals, Method D (0.1 ppm). Use lead standard solution
Water for Injections is water intended for use in the
(1 ppm Pb) to prepare the standard.
preparations of medicines for parenteral administration when
Chlorides (2.3.12). To 10 ml add 1 ml of 2 M nitric acid and water is used as a vehicle (Water for Injections in bulk) and for
0.2 ml of 0.1 M silver nitrate; the appearance of the solution dissolving or diluting substances or preparations for injectable
does not change for at least 15 minutes. preparations (Sterile Water for Injections).

1244
IP 2007 STERILE WATER FOR INJECTIONS

Water For Injections in Bulk allow to stand for 15 minutes. Any blue colour in the solution
is not more intense than that in a solution prepared at the
Production same time and in the same manner using a mixture of 4.5 ml of
nitrate-free water and 0.5 ml of nitrate standard solution
Water for Injections in bulk is obtained by distilling potable (2 ppm NO3) (0.2 ppm).
water or Purified Water from a neutral glass, quartz or suitable
metal still fitted with an effective device for preventing the Sulphates (2.3.17). To 10 ml add 0.1 ml of 2 M hydrochloric
entrainment of droplets; the still must be suitably maintained acid and 0.1 ml of barium chloride solution. The appearance
to ensure the production of a pyrogenic water. The first portion of the solution does not change for at least 1 hour.
of the distillate is discarded and the remainder is collected and Aluminium (2.3.8) For water for injections intended for use
stored in conditions designed to prevent the growth of micro- in the manufacture of dialysis solutions.
organisms and to avoid any other contamination.
Not more than 10 ppb, determined using the following
During production and subsequent storage, it is recommended solutions.
that adequate measures are taken to ensure that the microbial
Test solution. To 400 ml of the water under examination add
quality is controlled and monitored. Appropriate alert and
10 ml of acetate buffer solution pH 6.0 and 100 ml of distilled
action limits are set so as to detect adverse trends. The
water.
adequacy of these measures is determined by the following
tests that may be done off-line or in-line. Reference solution. Mix 2 ml of aluminium standard solution
(2 ppm Al), 10 ml of acetate buffer solution pH 6.0 and 98 ml
Total organic carbon (2.4.30). Not more than 0.5 mg per litre.
of distilled water.
Conductivity (2.4.9). Meets the requirements of the test.
Blank solution. Mix 10 ml of acetate buffer solution pH 6.0
Description. A clear and colourless liquid; odourless and and 100 ml of distilled water.
tasteless.
Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
Tests Unit per ml.
Storage. Store protected from moisture in containers designed
Acidity or alkalinity. To 10 ml, freshly boiled and cooled in a
to prevent the growth of micro-organisms.
borosilicate glass flask, add 0.05 ml of methyl red solution;
the resulting solution is not red. To 10 ml add 0.1 ml of Labelling. The label on the container in which the bulk has
bromothymol blue solution; the resulting solution is not blue. been distributed states that the contents have not been
sterilised.
Ammonium. To 20 ml add 1 ml of alkaline potassium mercuri-
iodide solution and allow to stand for 5 minutes. When viewed
vertically the solution is not more intensely coloured than a
solution prepared at the same time by adding 1 ml of alkaline Sterile Water For Injections
potassium mercuri-iodide solution to a solution containing
2.5 ml of dilute ammonium chloride solution and 7.5 ml of the Sterile Water for Injections is Water for Injections in bulk that
liquid under examination. has been distributed in suitable containers of glass or any
other material, sealed and sterilised by heat under conditions
Calcium and magnesium. To 100 ml add 2 ml of ammonia that ensure that the water complies with the test for bacterial
buffer pH 10.0, 50 mg of mordant black II mixture and 0.5 ml of endotoxins. It is free from any added substances. Each
0.01 M disodium edetate; a pure blue colour is produced. container contains a sufficient quantity of Water for Injections
Heavy metals (2.3.13). Evaporate 150 ml to 15 ml on a water- to permit the withdrawal of the nominal volume.
bath. 12 ml of the solution complies with the limit test for Description. A clear, colourless liquid; odourless.
heavy metals, Method D (0.1 ppm). Use lead standard solution
(1 ppm Pb) to prepare the standard. Tests
Chlorides (2.3.12). To 10 ml add 1 ml of 2 M nitric acid and Appearance of solution. When examined in suitable conditions
0.2 ml of 0.1 M silver nitrate; the appearance of the solution of visibility, it is clear (2.4.1) colourless (2.4.1) and practically
does not change for at least 15 minutes. free from suspended particles.
Nitrates. To 5 ml in a test-tube immersed in ice add 0.4 ml of a Acidity or alkalinity. To 20 ml add 0.05 ml of phenol red
10 per cent w/v solution of potassium chloride, 0.1 ml of solution. If the solution is yellow, it becomes red on the addition
diphenylamine solution and, dropwise with shaking, 5 ml of of 0.1 ml of 0.01 M sodium hydroxide; if red, it becomes yellow
sulphuric acid. Transfer the tube to a water-bath at 50° and on the addition of 0.15 ml of 0.01 M hydrochloric acid.

1245
WOOL FAT IP 2007

Ammonium. To 20 ml add 1 ml of alkaline potassium mercuri- Bacterial endotoxins (2.2.3). Not more than 0.25 Endotoxin
iodide solution and allow to stand for 5 minutes. When viewed Unit per ml.
vertically the solution is not more intensely coloured than a Sterility (2.2.11). Complies with the test for sterility.
solution prepared at the same time by adding 1 ml of alkaline
potassium mercuri-iodide solution to a solution containing Storage. Store in single dose containers of not larger than one
2.5 ml of dilute ammonium chloride solution and 7.5 ml of the litre size.
liquid under examination.
Calcium and magnesium. To 100 ml add 2 ml of ammonia
buffer pH 10.0, 50 mg of mordant black II mixture and 0.5 ml
of 0.01 M disodium edetate; a pure blue colour is produced. Wool Fat
Heavy metals (2.3.13). Evaporate 150 ml to 15 ml on a water- Anhydrous Lanolin
bath. 12 ml of the solution complies with the limit test for
Wool Fat is purified, anhydrous, waxy material obtained from
heavy metals, Method D (0.1 ppm). Use lead standard solution
the wool of sheep. It may contain Butylated Hydroxytoluene
(1 ppm Pb) to prepare the standard.
as an antioxidant.
Chlorides.To 10 ml add 1 ml of 2 M nitric acid and 0.2 ml of
Description. A pale yellow, unctuous substance; odour,
0.1 M silver nitrate; the appearance of the solution does not
characteristic.
change for at least 15 minutes.
For containers with a nominal volume of 100 ml or less, 15 ml Identification
complies with the limit test for chlorides (2.3.12) (0.5 ppm),
using a standard solution prepared by mixing 1.5 ml of chloride A. To a solution of 0.5 g in 5 ml of chloroform add 1 ml of
standard solution (5 ppm Cl) and 13.5 ml of water. acetic anhydride and 0.1 ml of sulphuric acid; a green colour
develops.
Nitrates. To 5 ml in a test-tube immersed in ice add 0.4 ml of a
10 per cent w/v solution of potassium chloride, 0.1 ml of B. To a solution of 50 mg in 5 ml of chloroform add 5 ml of
diphenylamine solution and, dropwise with shaking, 5 ml of sulphuric acid and shake; a red colour is produced and a
sulphuric acid. Transfer the tube to a water-bath at 50° and strong fluorescence appears in the lower layer.
allow to stand for 15 minutes. Any blue colour in the solution
Tests
is not more intense than that in a solution prepared at the
same time and in the same manner using a mixture of 4.5 ml of Melting range (2.4.21). 34° to 44°, determined by Method IV.
nitrate-free water and 0.5 ml of nitrate standard solution To fill the metal cup, melt the substance under examination on
(2 ppm NO3) (0.2 ppm). a water-bath, cool to about 50°, pour into the cup and allow to
Sulphates. To 10 ml add 0.1 ml of 2 M hydrochloric acid and stand at 15° to 20° for 24 hours.
0.1 ml of barium chloride solution. The appearance of the Acid value (2.3.23). Not more than 1.0, determined on 5.0 g
solution does not change for at least 1 hour. dissolved in 25 ml of the prescribed mixture of solvents.
Oxidisable substances. Boil 100 ml with 10 ml of 1 M sulphuric Peroxide value (2.3.35). Not more than 20.
acid, add 0.4 ml of 0.02 M potassium permanganate (for Sterile
Water for Injection in containers with fill volume of less than Saponification value. (2.3.37). 90 to 105. Heat for 4 hours.
50 ml) or 0.2 ml of 0.02 M potassium permanganate (for Sterile Water-absorption capacity. Weigh 10.0 g into a mortar. Add
Water for Injection in containers with fill volume of 50 ml or water in quantities of 0.2 to 0.5 ml from a burette and stir
more) and boil for 5 minutes. If a precipitate forms, cool in an vigorously, incorporating all the water before proceeding to
ice-bath to room temperature and filter through a sintered the next addition. The end-point is reached when visible
glass filter (porosity No.3). The pink colour of the solution droplets remain that cannot be incorporated; not less than
does not disappear completely. 20 ml of water is absorbed.
Residue on evaporation. Evaporate 100 ml to dryness on a Water-soluble acidic or alkaline substances. Shake
water-bath and dry the residue to constant weight at 105°. For vigorously 5.0 g, previously melted on a water-bath, for
containers with a nominal volume of 10 ml or less, the residue 2 minutes with 75 ml of water previously heated to 90° to 95°.
weighs not more than 4 mg (0.004 per cent) and for containers Allow to cool and filter through filter paper previously washed
with a nominal volume greater than 10 ml, the residue weighs with water. To 60 ml of the filtrate, which may not be clear, add
not more than 3 mg (0.003 per cent). 0.25 ml of bromothymol blue solution. Not more than 0.2 ml of
Particulate contamination (2.5.9). Complies with the 0.02 M hydrochloric acid or 0.15 ml of 0.02 M sodium
requirements of Method 1 or Method 2. hydroxide is required to change the colour of the solution.

1246
IP 2007 HYDROUS WOOL FAT

Water-soluble oxidisable substances. To 10 ml of the filtrate – flow rate. 40 ml per minute of the carrier gas.
obtained in the test for Water-soluble acidic or alkaline Calculate the content of butylated hydroxytoluene in the
substances add 1 ml of 1 M sulphuric acid and 0.1 ml of substance under examination from the heights or areas of the
0.02 M potassium permanganate; the solution is not peaks due to butylated hydroxytoluene and the internal
completely decolorised within 10 minutes. standard in the chromatograms obtained with test solution
Ammonia. To 10 ml of the filtrate obtained in the test for Water- (b) and the reference solution.
soluble acidic or alkaline substances add 1 ml of 1 M sodium
Sulphated ash (2.3.18). Not more than 0.15 per cent.
hydroxide and boil; the vapours do not turn red litmus paper
blue. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105° for 1 hour.
Chlorides. Boil 1.0 g with 20 ml of ethanol (90 per cent) under
a reflux condenser for 5 minutes, cool, add 40 ml of water and Storage. Store protected from moisture.
0.5 ml of nitric acid and filter. To the filtrate add 0.15 ml of a Labelling. The label states the proportion of any butylated
1 per cent w/v solution of silver nitrate in ethanol (90 per hydroxytoluene present.
cent). After 5 minutes, protected from light; any opalescence
produced is not more intense than that obtained by adding
0.15 ml of a 1 per cent w/v solution of silver nitrate in ethanol
(90 per cent) to a mixture of 0.2 ml of 0.02 M hydrochloric Hydrous Wool Fat
acid, 20 ml of ethanol (90 per cent), 40 ml of water and 0.5 ml
Hydrous Wool Fat is a mixture of 75 per cent w/w of Wool Fat
of nitric acid (150 ppm).
and 25 per cent w/w of Purified Water.
Paraffins. Prepare an alumina column 23 cm x 2 cm by adding
a slurry of anhydrous aluminium oxide and light petroleum Description. A pale yellow, unctuous substance; odour, faint
(40° to 60°) to a glass tube fitted with a tap and containing and characteristic. On heating, it separates at first into two
the light petroleum; the tap and absorbent cotton plugs should layers; with continued heating with stirring, water is driven
be free from grease. Allow to settle and reduce the depth of off and the residue which is transparent while warm, cools to
the solvent above the column to about 4 cm. Dissolve 3.0 g of form a yellowish, tenacious, soft mass.
the substance under examination in 50 ml of warm light
Identification
petroleum (40° to 60°), cool, pass the solution through the
column at a rate of 3 ml per minute and wash with 250 ml of the A. To a solution of 0.5 g in 5 ml of chloroform add 1 ml of
light petroleum. Distil the combined eluate and washings to acetic anhydride and 0.1 ml of sulphuric acid; a green colour
low bulk, evaporate to dryness on a water-bath and heat the develops.
residue at 105° for periods of 10 minutes until the difference
B. To a solution of 50 mg in 5 ml of chloroform add 5 ml of
between two successive weighings is not greater than 1 mg;
sulphuric acid and shake; a red colour is produced and a
the residue weighs not more than 30 mg.
strong fluorescence appears in the lower layer.
Butylated hydroxytoluene (if present). Not more than 200 ppm,
determined by gas chromatography (2.4.13). Tests
Test solution (a). A 10 per cent w/v solution of the substance Melting range (2.4.21). 34° to 44°, determined by Method IV.
under examination in carbon disulphide. To fill the metal cup, melt the substance under examination on
Test solution (b). A solution containing 10 per cent w/v of the a water-bath, cool to about 50°, pour into the cup and allow to
substance under examination and 0.002 per cent w/v of methyl stand at 15° to 20° for 24 hours.
n-decanoate (internal standard) in carbon disulphide. Acid value (2.3.23). Not more than 1.0, determined on 5.0 g
Reference solution. A solution containing 0.002 per cent w/v dissolved in 25 ml of the prescribed mixture of solvents.
each of butylated hydroxytoluene and the internal standard. Paraffins. Prepare an alumina column 23 cm x 2 cm by adding
Chromatographic system a slurry of anhydrous aluminium oxide and light petroleum
– a glass column 1.5 m x 4 mm, packed with silanised (40° to 60°) to a glass tube fitted with a tap and containing
diatomaceous support (80 to 100 mesh) (such as the light petroleum; the tap and absorbent cotton plugs should
Diatomite) impregnated with 10 per cent w/w of silicone be free from grease. Allow to settle and reduce the depth of
gum rubber (methyl) (such as SE-30), the solvent above the column to about 4 cm. Dissolve 3 g of
– temperature: the substance under examination in 50 ml of warm light
column 150°, petroleum (40° to 60°), cool, pass the solution through the
inlet port at 180° and detector at 300°, column at a rate of 3 ml per minute and wash with 250 ml of the

1247
HYDROUS WOOL FAT IP 2007

light petroleum. Distil the combined eluate and washings to Ammonia. To 10 ml of the filtrate obtained in the test for Water-
low bulk, evaporate to dryness on a water-bath and heat the soluble acidic or alkaline substances add 1 ml of 1 M sodium
residue at 105° for periods of 10 minutes until the difference hydroxide and boil; the vapours do not turn red litmus paper
between two successive weighings is not greater than 1 mg; blue.
the residue weighs not more than 30 mg. Chlorides. Boil 1.0 g with 20 ml of ethanol (90 per cent) under
Peroxide value (2.3.35). Not more than 15. a reflux condenser for 5 minutes, cool, add 40 ml of water and
0.5 ml of nitric acid and filter. To the filtrate add 0.15 ml of a1
Saponification value (2.3.37). 67 to 79. Heat for 4 hours.
per cent w/v solution of silver nitrate in ethanol (90 per
Water-absorption capacity. Weigh 10.0 g of the residue cent). After 5 minutes, protected from light, any opalescence
obtained in the test for Wool fat content into a mortar. Add produced is not more intense than that obtained by adding
water in quantities of 0.2 to 0.5 ml from a burette and stir 0.15 ml of a 1 per cent w/v solution of silver nitrate in ethanol
vigorously, incorporating all the water before proceeding to (90 per cent) to a mixture of 0.2 ml of 0.02 M hydrochloric
the next addition. The end-point is reached when visible acid, 20 ml of ethanol (90 per cent), 40 ml of water and 0.5 ml
droplets remain that cannot be incorporated; not less than of nitric acid (150 ppm).
20 ml of water is absorbed.
Wool fat content. 72.5 to 77.5 per cent, determined by the
Water-soluble acidic or alkaline substances. Shake following method. Weigh accurately about 30 g in a tared
vigorously 6.7 g, previously melted on a water-bath, for porcelain dish containing a glass rod, heat on a water-bath
2 minutes with 75 ml of water previously heated to 90° to 95°, with continuous stirring to constant weight and weigh the
Allow to cool and filter through filter paper previously washed residue.
with water. To 60 ml of the filtrate, which may not be clear, add solution. Titrate with 0.1 M perchloric acid, using
0.25 ml of bromothymol blue solution. Not more than 0.2 ml of 1-naphtholbenzein solution as indicator. Carry out a blank
0.02 M hydrochloric acid or 0.15 ml of 0.02 M sodium titration.
hydroxide is required to change the colour of the solution.
1 ml of 0.1 M perchloric acid is equivalent to 0.02808 g of
Water-soluble oxidisable substances. 10 ml of the filtrate C16H24N2, HCl.
obtained in the test for Water-soluble acidic or alkaline
substances add 1 ml of 1 M sulphuric acid and 0.1 ml of Storage. Store protected from light and moisture.
0.02 M potassium permanganate; the solution is not
completely decolorised within 10 minutes.

1248
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

X
Xylometazoline Hydrochloride ....
Xylometazoline Nasal Drops ....
Xylose ....

1249
IP 2007 XYLOMETAZOLINE NASAL DROPS

Xylometazoline Hydrochloride ninhydrin in a mixture of 100 ml of 1-butanol and 3 ml of


glacial acetic acid. Heat at 100º for 10 minutes, allow to cool,
CH3 and spray with dilute potassium iodobismuthate solution.
H3C Any spot corresponding to N-(2-aminoethyl)-4-tert-butyl-
CH3
H3C N 2,6-xylyl- aceta0-mide in the chromatogram obtained with
, HCl the test solution is not more intense than the spot in the
HN chromatogram obtained with the reference solution.
CH3
Iron (2.4.14). Moisten the residue obtained in the test for
C16H24N2,HCl Mol. Wt. 280.8 Sulphated ash with 5 ml of hydrochloric acid, evaporate to
dryness and dissolve in sufficient water to produce 50 ml.
Xylometazoline Hydrochloride is 2-(4-tert-butyl-2,6- 10 ml of the resulting solution complies with the limit test for
dimethylbenzyl)-2-imidazoline hydrochloride. iron (50 ppm).
Xylometazoline Hydrochloride contains not less than 99.0 per Sulphates (2.3.17). 0. 75 g complies with the limit test for
cent and not more than 101.0 per cent of C16H24N2, HCl, sulphates (200 ppm).
calculated on the dried basis.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Description. A white or almost white, crystalline powder;
odourless or almost odourless. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
on 1.0 g by drying in an oven at 105º.
Identification Assay. Weigh accurately about 0.5 g, dissolve in 50 ml of
A. Determine by infrared absorption spectrophotometry (2.4.6). anhydrous glacial acetic acid, add 10 ml of mercuric acetate
Compare the spectrum with that obtained with xylometazoline solution. Titrate with 0.1 M perchloric acid, using
hydrochloride RS or with the reference spectrum of 1-naphtholbenzein solution as indicator. Carry out a blank
xylometazoline hydrochloride. titration.
B. When examined in the range 230 nm to 360 nm (2.4.7), a 1 ml of 0.1 M perchloric acid is equivalent to 0.02808 g of
0.05 per cent w/v solution in 0.1 M hydrochloric acid shows C16H24N2, HCl.
an absorption maximum at about 265 nm and a minimum at Storage. Store protected from light and moisture.
about 257 nm with two inflections at about 270 nm and
275 nm; absorbance at about 265 nm, about 0.5.
C. To 1 ml of a 0.05 per cent w/v solution, add 0.2 ml of a 5 per
cent w/v solution of sodium nitroprusside and 0.1 ml of 5 M Xylometazoline Nasal Drops
sodium hydroxide, allow to stand for 10 minutes and add 2 ml Xylometazoline Hydrochloride Nasal Drops
of sodium bicarbonate solution; a violet colour is produced.
Xylometazoline Nasal Drops are a solution of Xylometazoline
D. Gives the reactions of chlorides (2.3.1).
Hydrochloride in Purified Water.
Tests Xylometazoline Nasal Drops contain not less than 90.0 per
cent and not more than 110.0 per cent of the stated amount of
pH (2.4.24). 5.0 to 6.6, determined in a 5.0 per cent w/v solution.
xylometazoline hydrochloride, C16H24N2, HCl.
N-(2-Aminoethyl)-4-tert-butyl-2, 6-xylylacetamide. Determine
by thin-layer chromatography (2.4.17), coating the plate with Identification
silica gel HF254.
A. To a volume containing 50 mg of Xylometazoline
Mobile phase. A mixture of 200 volumes of methanol and Hydrochloride add 5 ml of 1 M sodium hydroxide, extract with
3 volumes of strong ammonia solution. 10 ml of dichloromethane, evaporate to dryness and dissolve
Test solution. Dissolve 0.2 g of the substance under the residue in 0.5 ml of dichloromethane.
examination in 10 ml of methanol. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution. A 0.01 per cent w/v solution of N-(2- Compare the spectrum with that obtained with xylometazoline
aminoethyl)-4-tert-butyl-2,6-xylyl- acetamide RS in hydrochloride RS treated in the same manner or with the
methanol. reference spectrum of xylometazoline.
Apply to the plate 5 µl of each solution. After development, B. To a volume containing 0.5 mg of Xylometazoline
dry the plate in air, spray with a solution containing 0.3 g of Hydrochloride add 0.2 ml of a 5 per cent w/v solution of sodium

1251
XYLOSE IP 2007

nitroprusside and 0.1 ml of 5 M sodium hydroxide, allow to the absorbance of the resulting solution at the maximum at
stand for 10 minutes and add 1 ml of sodium bicarbonate about 560 nm (2.4.7), using as blank a solution prepared by
solution; a violet colour is produced. treating 5 ml of water and 2.5 ml of 1 M sodium hydroxide in
the same manner beginning at the words “and 2.5 ml of a 5 per
Tests cent w/v solution of sodium nitroprusside,.....”.
pH (2.4.24). 5.6 to 6.6. Calculate the content of C16H24N2, HCl from the absorbance
N-(2-Aminoethyl)-4-tert-butyl-2,6-xylylacetamide. Determine obtained by repeating the operation using a 0.1 per cent w/v
by thin-layer chromatography (2.4.17), coating the plate with solution of xylometazoline hydrochloride RS in place of the
silica gel HF254. nasal drops.

Mobile phase. A mixture of 200 volumes of methanol and Storage. Store protected from light and moisture.
3 volumes of strong ammonia solution.
Test solution. Add a volume containing 10 mg of
Xylometazoline Hydrochloride to 30 ml of water, add 5 ml of Xylose
5 M sodium hydroxide, mix, extract with three quantities, each D-Xylose; D-Xylopyranose
of 20 ml, of dichloromethane, evaporate the combined extracts
to dryness and dissolve the residue in 1 ml of O
dichloromethane.
OH OH
Reference solution. A 0.03 per cent w/v solution of N-(2- HO
aminoethyl)-4-tert-butyl-2,6-xylylacetamide RS in
OH
dichloromethane.
Apply to the plate 5 µl of each solution. After development, C5H10O5 Mol. Wt. 150.1
dry the plate in air, spray with a solution containing 0.3 g of Xylose contains not less than 98.0 per cent and not more
ninhydrin in a mixture of 100 ml of 1-butanol and 3 ml of than 102.0 per cent of C5H10O5, calculated on the dried basis.
glacial acetic acid. Heat at 100º for 10 minutes, allow to cool,
Description. Colourless needles or a white, crystalline powder.
and spray with dilute potassium iodobismuthate solution.
Any spot corresponding to N-(2-aminoethyl)-4-tert-butyl- Identification
2,6-xylyl- acetamide in the chromatogram obtained with test
solution is not more intense than the spot in the chromatogram A. Determine by thin-layer chromatography (2.4.17), coating
obtained with reference solution. the plate with a suspension of silica gel G in a 0.3 per cent
w/v solution of sodium acetate to form a uniform layer 0.5 mm
Other tests. Comply with the tests stated under Nasal
thick.
Preparations.
Mobile phase. A mixture of 70 volumes of glacial acetic acid,
Assay. To a volume containing 10 mg of Xylometazoline
60 volumes of chloroform and 10 volumes of water.
Hydrochloride add 5 ml of water, 10 ml of 2 M hydrochloric
acid and 10 ml of dichloromethane and shake for 1 minute. Test solution. Dissolve 0.5 g of the substance under
Discard the dichloromethane layer and repeat the extraction examination in 10 ml of water.
with two further quantities, each of 10 ml, of dichloromethane. Reference solution (a). A 5.0 per cent w/v solution of xylose
Add to the aqueous extract 10 ml of 5 M sodium hydroxide RS in water.
and 10 ml of dichloromethane, shake for 1 minute and allow to
separate. Filter the dichloromethane extract through glass wool Reference solution (b). A mixture of equal volumes of the test
and repeat the extraction with four further quantities, each of solution and reference solution (a).
10 ml, of dichloromethane. Evaporate the combined Apply to the plate 2 µl of each solution. Develop the plate in
dichloromethane extracts almost to dryness on a water-bath, a continuous elution tank for about 4 hours. Dry the plate in
remove the final traces of solvent in a current of air and dissolve warm air, spray with a solution in acetone containing 1 per
the residue in 10.0 ml of 0.01 M hydrochloric acid. To 2.0 ml cent w/v solution of diphenylamine, 1 per cent v/v of aniline
of this solution add 3 ml of water, 2.5 ml of 1 M sodium and 1 per cent v/v of phosphoric acid and heat for 10 minutes
hydroxide and 2.5 ml of a 5 per cent w/v solution of sodium at 130º. The principal spot in the chromatogram obtained with
nitroprusside, mix and allow to stand protected from light for the test solution corresponds to that in the chromatogram
10 minutes. Add 10 ml of a freshly prepared 8.3 per cent w/v obtained with reference solution (a). The principal spot in the
solution of sodium bicarbonate, dilute to 100.0 ml with water, chromatogram obtained with reference solution (b) appears
allow to stand protected from light for 10 minutes and measure as a single, compact spot.

1252
IP 2007 XYLOSE

B. When heated with potassium cupri-tartrate solution it Sulphated ash (2.3.18). Not more than 0.1 per cent.
produces a copious precipitate of cuprous oxide. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Tests on 1.0 g by drying in an oven at 100º at a pressure not exceeding
0.7 kPa.
Appearance of solution. A 10.0 per cent w/v solution in carbon Assay. Carry out the following procedure keeping strict
dioxide-free water is clear (2.4.1), and colourless (2.4.1). control of time between steps.
Acidity. Dissolve 5.0 g in 50 ml of carbon dioxide-free water. Weigh accurately about 1.0 g of the substance under
Not more than 0.2 ml of 0.1 M sodium hydroxide is required to examination, dissolve in a saturated solution of benzoic acid
neutralise the solution using dilute phenolphthalein solution in a 100-ml volumetric flask and dilute to volume with the same
as indicator. solvent. Dilute 1.0 ml of this solution to 100.0 ml with the same
Specific optical rotation (2.4.22). +18.5º to +19.5º, determined solvent. To 1.0 ml of the resulting solution, in two different
at 20º in a 10.0 per cent w/v solution containing 0.4 per cent test-tubes, add 5 ml of 4-bromoaniline solution into each
v/v of 5 M ammonia. tube and mix. Loosely stopper one tube, place in a water-bath
maintained at 70º for 10 minutes, remove, cool rapidly to room
Arsenic (2.3.10). Dissolve 10.0 g in 50 ml of water and add
temperature and mix. Keep the tube in the dark for 70 minutes
10 ml of stannated hydrochloric acid AsT. The resulting
and measure the absorbance at the maximum at about 520 nm
solution complies with the limit test for arsenic (1 ppm).
(2.4.7), using the untreated solution in the second test tube as
Heavy metals (2.3.13). 1.0 g complies with the limit test for the blank. Simultaneously, carry out the operation using
heavy metals, Method A (20 ppm). 1.0 ml of a 0.01 per cent w/v solution of xylose RS in a saturated
Iron (2.3.14). A solution of 4.0 g in 25 ml of water complies solution of benzoic acid beginning at the words “add 5 ml of
with the limit test for iron (10 ppm). 4-bromoaniline solution.....”.

Chlorides (2.3.12). Dissolve 3.0 g in 20 ml of water. 5 ml of the Calculate the content of C5H10O5.
resulting solution complies with the limit test for chlorides Storage. Store protected from moisture.
(330 ppm).

1253
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

Z
Zidovudine ....
Zidovudine Capsules ....
Zidovudine Injection ....
Zidovudine Oral Solution ....
Zidovudine Tablets ....
Zidovudine, Lamivudine And Nevirapine Tablets ....
Zinc Chloride ....
Zinc Oxide ....
Zinc Oxide Cream ....
Zinc Stearate ....
Zinc Sulphate ....
Zinc Sulphate Eye Drops ....
Zinc Undecenoate ....
Zinc Undecenoate Ointment ....

1255
IP 2007 ZIDOVUDINE

Zidovudine spots in the chromatogram obtained with the reference


solution. No secondary spot in the chromatogram obtained
with the test solution is more intense than the principal spot
O in the chromatogram obtained with the reference solution
CH3 (0.5 per cent).
HN
Spray the plate with a mixture of 0.5 g of carbazole in 95 ml of
O N ethanol (95 per cent) and 5 ml of sulphuric acid, heat for
HO
O 10 minutes at 120º and compare the intensities of any secondary
spots observed in the chromatogram obtained with the test
solution with those of the principal spots in the chromatogram
N3 obtained with the reference solution. No spot corresponding
to triphenylmethanol (Rf value about 2.3 relative to the Rf of
C10H13N5O4 Mol. Wt. 267.2 zidovudine) is more intense than the corresponding spot in
the chromatogram obtained with the reference solution
Zidovudine is 1-(3-azido-2,3-dideoxy-β-D-ribofuranosyl)-5- (0.5 per cent). No secondary spot in the chromatogram
methylpyrimidine-2,4(1H,3H)-dione. obtained with the test solution is more intense than the
Zidovudine contains not less than 97.0 per cent and not more principal spot in the chromatogram obtained with the reference
than 102.0 per cent of C10H13N5O4, calculated on the anhydrous solution.
basis. B. Determine by liquid chromatography (2.4.14).
Description. A white or almost white powder.
Test solution. Dissolve 0.1 g of the substance under
examination in 100 ml of methanol.
Identification
Reference solution (a). A 0.001 per cent w/v solution of
A. Determine by infrared absorption spectrophotometry (2.4.6).
zidovudine RS in methanol.
Compare the spectrum with that obtained with zidovudine RS
or with the reference spectrum of zidovudine. Reference solution (b). A solution containing 0.1 per cent
w/v of zidovudine RS and 0.001 per cent w/v each of
B. In the Assay, the principal peak in the chromatogram
zidovudine-related compound B RS and zidovudine- related
obtained with the test solution corresponds to the peak due
compound C RS in methanol.
to zidovudine in the chromatogram obtained with reference
solution (a). Chromatographic system
C. Melting range (2.4.21). 122º to 125º. – a stainless steel column 15 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm),
Tests – mobile phase: A. water
B. methanol,
Specific optical rotation (2.4.22). +60.5° to +63.0°, determined
– flow rate. 1 ml per minute,
in a 1.0 per cent w/v solution in ethanol (95 per cent).
– a linear gradient programme using the conditions given
Related substances. A. Determine by thin-layer below,
chromatography (2.4.17), coating the plate with silica gel – spectrophotometer set at 265 nm,
GF254. – a 10 µl loop injector.
Mobile phase. A mixture of 90 volumes of dichloromethane Time Water Methanol
and 10 volumes of methanol. (in min.) (per cent v/v) (per cent v/v)
Test solution. Dissolve 0.2 g of the substance under 0 90 10
examination in 10 ml of methanol. 15 35 65
Reference solution. A solution containing 0.01 per cent w/v 30 35 65
each of zidovudine RS and triphenylmethanol in methanol. 35 5 95
Apply to the plate 10 µl of each solution. Allow the mobile 40 5 95
phase to rise to about three-fourths of the height of the plate.
45 90 10
Dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spots observed in the chromatogram obtained Inject reference solution (b). The test is not valid unless the
with the test solution correspond to those of the principal resolution between the peaks due to zidovudine and

1257
ZIDOVUDINE CAPSULES IP 2007

zidovudine-related compound B is not less than 1.5 and the Zidovudine Capsules
tailing factor for zidovudine is not more than 1.5.
Zidovudine Capsules contain not less than 90.0 per cent and
Separately inject the test solution and reference solution (a). not more than 110.0 per cent of the stated amount of
The area of the peak corresponding to zidovudine- related zidovudine, C10H13N5O4.
compound B is not greater than 1.0 per cent and of that to
zidovudine-related compound C is not greater than 2.0 per Identification
cent.
A.Transfer a quantity of the mixed contents of the capsules
The sum of the percentages of related substances by tests A
containing about 15 mg of Zidovudine to a 100-ml volumetric
and B is not greater than 3.0 per cent.
flask. Add about 80 ml of a mixture of 75 volumes of methanol
Heavy metals (2.3.13). 1.0 g complies with limit test for heavy and 25 volumes of water, shake for 10 minutes and dilute to
metals, Method B ( 20 ppm). volume with the same solvent mixture, mix and filter. Dilute
10 ml of the filtrate to 100 ml with the same solvent mixture.
Sulphated ash (2.3.18). Not more than 0.25 per cent.
When examined in the range 200 nm to 300 nm (2.4.7), the
Water (2.3.43). Not more than 1.0 per cent, determined on
resulting solution shows an absorption maximum at about
0.5 g.
265 nm.
Assay. Determine by liquid chromatography (2,4,14).
B. In the Assay, the principal peak in the chromatogram
Test solution. Weigh accurately about 100 mg of the substance obtained with the test solution corresponds to the peak due
under examination, dissolve in a suitable quantity of methanol to zidovudine in the chromatogram obtained with reference
in a 50-ml volumetric flask and make up to volume with the solution (c).
mobile phase. Dilute 5.0 ml of this solution to 50.0 ml with the
mobile phase. Tests
Reference solution (a). Weigh accurately about 100 mg of Related substances. Determine by liquid chromatography
zidovudine RS, dissolve in a suitable quantity of methanol in (2.4.14).
a 50-ml volumetric flask and make up to volume with the mobile
Test solution. Weigh accurately a quantity of the mixed
phase (solution A). Dilute 5.0 ml of solution A to 50.0 ml with
contents of 20 capsules containing about 100 mg of Zidovudine
the mobile phase.
and transfer to a 100-ml volumetric flask. Add about 60 ml of
Reference solution (b). Transfer 2.0 ml of a 0.005 per cent w/v the mobile phase, mix with the aid of ultrasound for 10 minutes,
solution of zidovudine-related compound B RS in the mobile dilute to volume with the mobile phase, mix and filter. Dilute
phase to a 50-ml volumetric flask, add 5.0 ml of solution A and 5.0 ml of the filtrate to 50.0 ml with the mobile phase.
make up to volume with the mobile phase.
Reference solution (a). Weigh accurately about 100 mg of
Chromatographic system zidovudine RS and transfer to a 100-ml volumetric flask.
– a stainless steel column 15 cm x 4.6 mm, packed with Dissolve in about 60 ml of the mobile phase and dilute to
octadecylsilane bonded to porous silica (5 µm), volume with the mobile phase.
– mobile phase: a mixture of 70 volumes of water and
Reference solution (b). Weigh accurately about 20 mg of
30 volumes of methanol,
thymine and transfer to a 200-ml volumetric flask, dissolve
– flow rate. 1 ml per minute,
and make up to volume with methanol.
– spectrophotometer set at 265 nm,
– a 20 µl loop injector. Reference solution (c). Transfer 10.0 ml of the reference
solution (a) and 2.0 ml of reference solution (b) to a 100-ml
Inject reference solution (b). The test is not valid unless the
volumetric flask and make up to the volume with the mobile
resolution between the peaks due to zidovudine and
phase.
zidovudine-related compound B is not less than 2.0.
Chromatographic system
Inject reference solution (a). The relative standard deviation – a stainless steel column 25 cm x 4.6 mm, packed with
for replicate injections is not more than 2.0 per cent. octadecylsilane bonded to porous silica (5 µm),
Separately inject the test solution and reference solution (a) – mobile phase: a mixture of 20 volumes of methanol and
and measure the responses for the principal peak. 80 volumes of water,
– flow rate. 1 ml per minute,
Calculate the content of C10H13N5O4.
– spectrophotometer set at 265 nm,
Storage. Store protected from light. – a 10 µl loop injector.

1258
IP 2007 ZIDOVUDINE INJECTION

Inject reference solution (c). The test is not valid unless the Storage. Store protected from moisture.
relative retention times are about 0.2 for thymine and 1.0 for
zidovudine, the resolution between zidovudine and thymine
is not less than 5.0, the tailing factor is not more than 2.0 and Zidovudine Injection
the relative standard deviation for replicate injections is not
more than 2.0 per cent. Zidovudine Injection is a sterile solution of Zidovudine in
Water for Injections.
Inject separately the test solution and reference solution (c)
and measure the responses for the thymine peak. The content Zidovudine Injection contains not less than 90.0 per cent and
of thymine in the capsules should not be more than 3.0 per not more than 110.0 per cent of the stated amount of
cent. C10H13N5O4.
Dissolution (2.5.2). Description. A clear, colourless solution.
Apparatus. No 1 Identification
Medium. 900 ml of water
Speed and time. 50 rpm and 45 minutes. A. When examined in the range 220 nm to 360 nm (2.4.7), a
0.0015 per cent w/v solution in a mixture of 75 volumes of
Withdraw a suitable volume of the medium and filter promptly methanol and 25 volumes of water shows absorption maxima
through a membrane filter disc having an average pore diameter similar to those obtained with a solution of zidovudine RS of
not greater than 1.0 µm, rejecting the first few ml of the filtrate the same concentration.
and dilute a suitable volume of the filtrate, if necessary with
water. B. In the Assay, the principal peak in the chromatogram
obtained with the test solution corresponds to that in the
Determine by liquid chromatography (2.4.14). chromatogram obtained with the reference solution.
Test solution. The filtrate obtained as given above.
Tests
Reference solution. A known quantity of zidovudine RS is
dissolved in 1 ml of methanol and suitably diluted with water pH (2.4.24). 3.5 to 7.0, in a mixture containing a volume of
to obtain a solution having a similar concentration as that of injection containing 150 mg of zidovudine and 5 ml of 0.12 M
the test solution. potassium chloride.
Chromatographic system Related substances. Determine by liquid chromatography
– a stainless steel column 25 cm x 4.6 mm, packed with (2.4.14).
octadecylsilane bonded to porous silica (5 µm), Test solution. Dilute an accurately measured volume of the
– mobile phase: a mixture of 20 volumes of methanol and injection containing 25 mg of Zidovudine, to 25 ml with
80 volumes of water, methanol.
– flow rate. 1 ml per minute,
– spectrophotometer set at 265 nm, Reference solution (a). A 0.1 per cent w/v solution of
– a 10 µl loop injector. zidovudine RS in methanol.
Reference solution (b). A solution containing 0.1 per cent w/
Inject the reference solution. The tailing factor is not more
v of zidovudine RS and 0.001 per cent w/v of zidovudine
than 2.0 for zidovudine peak and the relative standard deviation
related compound C (thymine).
for replicate injections is not more than 2.0 per cent.
Chromatographic system
Inject the test solution and the reference solution and measure
– a stainless steel column 25 cm × 4.0 mm, packed with
the peak responses of the major peak.
octadecylsilane bonded to porous silica ( 5 µm),
Calculate the content of C10H13N5O4 in the medium. – mobile phase: a mixture of 80 volumes of water and 20
D. Not less than 75 per cent of the stated amount of volumes of methanol,
C10H13N5O4. – flow rate. 1 ml per minute,
– spectrophotometer set at 265 nm,
Other tests. Comply with the tests stated under Capsules. – a 10 µl loop injector.
Assay. Determine by liquid chromatography (2.4.14), as Inject reference solution (b). The test is not valid unless the
described under Related substances. Inject separately the test resolution between zidovudine and thymine is not less than
solution and reference solution (c) and measure the responses 4.0, the tailing factor is not more than 2.0 and the relative
for the principal peak. standard deviation for replicate injections is not more than 2.0
Calculate the content of C10H13N5O4 in the capsules. per cent.

1259
ZIDOVUDINE ORAL SOLUTION IP 2007

Inject the test solution and reference solution (b). In the Tests
chromatogram obtained with the test solution the area of any
peak corresponding to thymine is not greater than twice the pH (2.4.24). 3.0 to 4.0, determined in a mixture containing a
area of the corresponding peak in the chromatogram obtained volume of the preparation under examination containing
with reference solution (b) (2.0 per cent). 150 mg of zidovudine and 5 ml of 0.12 M potassium chloride.

Other tests. Complies with the tests stated under Parenteral Related substances. Determine by liquid chromatography
preparations (Injections). (2.4.14).

Bacterial endotoxins (2.2.3). Not more than 1.0 Endotoxin Test solution. Transfer an accurately measured volume of the
Unit per mg of zidovudine. preparation under examination containing about 100 mg of
zidovudine to a 100-ml volumetric flask, dissolve and dilute to
Assay. Determine by liquid chromatography (2.4.14), as given volume with the mobile phase and mix. Dilute 5.0 ml of this
under the test for Related substances. solution to 50.0 ml with the mobile phase.
Inject alternately the test solution and reference solution (a). Reference solution (a). Weigh accurately about 100 mg of
Calculate the content of C10H13N5O4 in the injection. zidovudine RS and transfer to a 100-ml volumetric flask, add
about 50 ml of the mobile phase, mix with the aid of ultrasound
Storage. Store protected from light and moisture. to dissolve, dilute to volume with the mobile phase and mix.
Reference solution (b). Weigh accurately about 20 mg of
thymine RS and transfer to a 200-ml volumetric flask, add about
150 ml of the mobile phase, mix with the aid of ultrasound to
Zidovudine Oral Solution dissolve, dilute to volume with the mobile phase and mix.
Zidovudine Oral Solution is a solution of Zidovudine in a Reference solution (c). Transfer 10.0 ml of the reference
suitable flavoured vehicle. solution (a) and 2.0 ml of reference solution (b) to a 100-ml
volumetric flask and make up to the volume with the mobile
Zidovudine Oral Solution contains not less than 90.0 per cent
phase.
and not more than 110.0 per cent of the stated amount of
zidovudine, C10H13N5O4. Chromatographic system
– a stainless steel column 12.5 cm x 4.0 mm, packed with
Identification octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 90 volumes of 0.04 M sodium
A. Determine by thin-layer chromatography (2.4.17), coating
acetate, 9 volumes of methanol, 1 volume of acetonitrile
the plate with silica gel GF254.
and 0.2 volume of glacial acetic acid,
Mobile phase. A mixture of 40 volumes of 1-butanol, – flow rate. 1 ml per minute,
30 volumes of heptane, 30 volumes of acetone and 10 volumes – spectrophotometer set at 240 nm,
of strong ammonia solution. – a 10 µl loop injector.
Test solution. Dilute the preparation under examination with Inject reference solution (c). The test is not valid unless the
methanol to obtain a solution containing 5 mg of zidovudine relative retention times are about 0.12 for thymine and 1.0 for
per ml. zidovudine, the resolution between zidovudine and thymine
Reference solution. A 0.5 per cent w/v solution of zidovudine is not less than 4.0, the tailing factor is not more than 2.0 and
RS in a mixture of 75 volumes of methanol and 25 volumes of the relative standard deviation for replicate injections is not
water. more than 2.0 per cent.

Apply to the plate 5 µl of each solution. After development, Inject separately the test solution and reference solution (c)
dry the plate in air and examine in ultraviolet light at 254 nm. and measure the responses for the thymine peak. The content
The principal spot in the chromatogram obtained with the test of thymine in the capsules should not be more than 3.0 per
solution corresponds to that in the chromatogram obtained cent.
with the reference solution. Other tests. Comply with the tests stated under Oral Liquids.
B. In the Assay, the principal peak in the chromatogram Assay. Determine by liquid chromatography (2.4.14), as
obtained with the test solution corresponds to the peak due described under Related substances. Inject separately the test
to zidovudine in the chromatogram obtained with reference solution and reference solution (c) and measure the responses
solution (c). for the major peak.

1260
IP 2007 ZIDOVUDINE TABLETS

Calculate the content of C10H13N5O4 in the preparation under – mobile phase: a mixture of 90 ml of methanol and 40 ml
examination. of acetonitrile and a buffer solution prepared by
Storage. Store protected from light and moisture. dissolving 3.0 g of sodium acetate and 3.0 g of sodium
octanesulfonate in 900 ml of water and adjusting the
pH to 5.3 with glacial acetic acid,
– flow rate. 1.3 ml per minute,
Zidovudine Tablets – spectrophotometer set at 265 nm,
– a 20 µl loop injector.
Zidovudine Tablets contain not less than 90.0 per cent and Inject reference solution (c). The test is not valid unless the
not more than 110.0 per cent of the stated amount of resolution between zidovudine and the peak having relative
zidovudine, C10H13N5O4. The tablets may be coated. retention time of 1.2 is not less than 2.5, the tailing factor for
the zidovudine peak is not more than 2.0 and relative standard
Identification deviation for replicate injections is not more than 2.0 per cent.
A. In the Assay, the principal peak in the chromatogram Separately inject the test solution and record the
obtained with the test solution corresponds to the peak due chromatograms for 3 times the retention time of zidovudine.
to Zidovudine in the chromatogram obtained with reference The content of the impurity at a relative retention time of 0.17
solution (a). compared to that of the zidovudine peak is not greater than
1.5 per cent and that of any other impurity is not greater than
B. Remove the coating from a few tablets and crush them in a
0.2 per cent. The sum of all the impurities is not greater than
mortar so that no large pieces remain.
2.0 per cent.
On the powder determine by infrared absorption
Dissolution (2.5.2).
spectrophotometry (2.4.6). Compare the spectrum with that
obtained with zidovudine RS or with the reference spectrum Apparatus. No 1
of zidovudine. Medium. 900 ml of water
Speed and time. 50 rpm and 30 minutes.
Tests
Withdraw a suitable volume of the medium and filter promptly
Related substances. Determine by liquid chromatography through a membrane filter disc having an average pore diameter
(2.4.14) not greater than 1.0 µm, rejecting the first few ml of the filtrate
Test solution. Weigh and powder 20 tablets. Weigh accurately and dilute a suitable volume of the filtrate, if necessary with
a quantity of the powder containing about 100 mg of water.
Zidovudine and transfer to a 100-ml volumetric flask. Add Determine by liquid chromatography (2.4.14).
about 60 ml of the mobile phase and mix with the aid of
Test solution. The filtrate obtained as given above.
ultrasound for 10 minutes. Make up to volume with the mobile
phase, mix and filter through a membrane filter disc with an Reference solution. A known quantity of zidovudine RS is
average pore diameter not greater than 1.0 µm, rejecting the dissolved in 1 ml of methanol and suitably diluted with water
first few ml of the filtrate. Further dilute 5.0 ml of the filtrate to to obtain a solution having a similar concentration as that of
50.0 ml with the mobile phase. the test solution.
Reference solution (a). Weigh accurately about 100 mg of the Chromatographic system
zidovudine RS and transfer to a 100-ml volumetric flask. – a stainless steel column 15 cm x 4.6 mm, packed with
Dissolve in about 50 ml of the mobile phase and make up to octadecylsilane bonded to porous silica (5 µm),
volume with the mobile phase. – mobile phase: a mixture of 90 ml of methanol and 40 ml
of acetonitrile and a buffer prepared by dissolving
Reference solution (b). Weigh accurately about 20 mg of
3.0 g of sodium acetate and 3.0 g of sodium
thymine and transfer to a 200-ml volumetric flask, dissolve
1-octanesulphonate in 900 ml of water and adjusting
and make up to volume with methanol.
the pH to 5.3 with glacial acetic acid,
Reference solution (c). Transfer 10.0 ml of reference solution – flow rate. 1.3 ml per minute,
(a) and 2.0 ml of reference solution (b) to a 100-ml volumetric – spectrophotometer set at 265 nm,
flask and make up to volume with the mobile phase. – a 10 µl loop injector.
Chromatographic system Inject the reference solution. The tailing factor is not more
– a stainless steel column 25 cm x 4.6 mm, packed with than 2.0 for zidovudine peak and the relative standard deviation
octadecylsilane bonded to porous silica (5 µm), for replicate injections is not more than 2.0 per cent.

1261
ZIDOVUDINE, LAMIVUDINE AND NEVIRAPINE TABLETS IP 2007

Inject separately the test solution and the reference solution – flow rate. 1 ml per minute,
and measure the peak responses of the major peak. – a linear gradient programme using the conditions given
Calculate the content of C10H13N5O4 in the medium. below,
– spectrophotometer set at 270 nm,
D. Not less than 80 per cent of the stated amount of – a 20 µl loop injector.
C10H13N5O4.
Time 0.1 M ammonium acetate acetonitrile
Other tests. Comply with the tests stated under Tablets. (in min.) (per cent v/v) (per cent v/v)
Assay. Determine by liquid chromatography (2.4.14), as 0 95 5
described under Related substances. Separately inject the test 5 95 5
solution and reference solution (c) and measure the peak 25 20 80
responses for the major peak.
30 20 80
Calculate the content of C10H13N5O4 in the tablets.
31 95 5
Storage. Store protected from light and moisture. 35 95 5
Inject the reference solution. The test is not valid unless the
column efficiency determined from the zidovudine, lamivudine
Zidovudine, Lamivudine and and nevirapine peaks is not less than 3000 theoretical plates
and the tailing factor for the same peaks is not more than 2.0.
Nevirapine Tablets
Inject separately water and the test solution. Examine the
Zidovudine, Lamivudine and Nevirapine Tablets contain not
chromatogram obtained with water for any extraneous peaks
less than 90.0 per cent and not more than 110.0 per cent of the
and ignore the corresponding peaks observed in the
stated amounts of zidovudine, C10H 13N5O4, lamivudine,
chromatogram obtained with the test solution.
C8H11N3O3S and nevirapine, C15H14N4O. The tablets may be
coated. Any secondary peak observed in the chromatogram obtained
with the test solution corresponding to a relative retention
Identification time of 0.35 should not be more than 1.0 per cent. Any other
secondary peak observed in the chromatogram obtained with
A. In the Assay, the three principal peaks in the chromatogram
the test solution should not be more than 0.5 per cent and the
obtained with the test solution correspond to the peaks due
sum of the areas of all the secondary peaks should not be
to zidovudine, lamivudine and nevirapine in the chromatogram
more than 2.5 per cent when calculated by percentage area
obtained with the reference solution.
normalisation.
Tests Dissolution (2.5.2).
Related substances. Determine by liquid chromatography Apparatus. No 1
(2.4.14). Medium. 900 ml of 0.1 M hydrochloric acid
Test solution. Weigh accurately a quantity of the powdered Speed and time. 50 rpm and 30 minutes.
tablets containing about 100 mg of Zidovudine and transfer Withdraw a suitable volume of the medium and filter promptly
to a 200-ml volumetric flask. Add about 150 ml of water, mix through a membrane filter disc having an average pore diameter
with the aid of ultrasound for 10 minutes, dilute to volume not greater than 1.0 µm, rejecting the first few ml of the filtrate
with water, mix and filter. and dilute a suitable volume of the filtrate, if necessary with
Reference solution. Weigh accurately about 100 mg of water.
zidovudine RS, 50 mg of lamivudine RS and 65 mg of nevirapine Determine by liquid chromatography (2.4.14).
RS and transfer to a 200-ml volumetric flask. Add about 20 ml
of methanol, mix with the aid of ultrasound to dissolve, dilute Test solution. The filtrate obtained as given above.
to volume with water and mix. Reference solution. Weigh accurately about 300 mg of
Chromatographic system zidovudine RS, 150 mg of lamivudine RS and 200 mg of
– a stainless steel column 25 cm x 4.6 mm, packed with nevirapine RS and transfer to a 100-ml volumetric flask. Add
octylsilane bonded to porous silica (5 µm) (such as about 20 ml of methanol, mix with the aid of ultrasound to
Hypersil C8), dissolve and dilute to volume with a solvent mixture of equal
– mobile phase: A. 0.1 M ammonium acetate, volumes of methanol and water. Dilute 5.0 ml of this solution
B. acetonitrile, to 50.0 ml with 0.1 M hydrochloric acid.

1262
IP 2007 ZINC CHLORIDE

Chromatographic system Storage. Store protected from moisture.


– a stainless steel column 15 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 35 volumes of methanol and
65 volumes of a buffer solution prepared by dissolving Zinc Chloride
0.68 g of potassium dihydrogen phosphate and 1.0 g of ZnCl2 Mol. Wt. 136.3
sodium octanesulphonate in 1000.0 ml of water to which
1 ml of triethylamine is added and the pH of which is Zinc Chloride contains not less than 95.0 per cent and not
adjusted to 2.5 with phosphoric acid, more than 100.5 per cent of ZnCl2.
– flow rate. 1 ml per minute, Description. A white or practically white, crystalline powder;
– spectrophotometer set at 266 nm, odourless; very deliquescent.
– a 10 µl loop injector.
Inject the reference solution. The tailing factor for the Identification
individual zidovudine, lamivudine and nevirapine peaks is not A. To 2 g add 38 ml of carbon dioxide-free water prepared
more than 2.0 and the relative standard deviation for replicate from distilled water and add 2 M hydrochloric acid dropwise
injections of all the analyte peaks is not more than 2.0 per until solution is complete and dilute to 40 ml with carbon
cent. dioxide-free water prepared from distilled water (solution
Inject the test solution and the reference solution and measure A). Solution A gives the reaction of zinc salts (2.3.1).
the peak responses of the major peaks due to zidovudine,
B. A 5 per cent w/v solution in 2 M nitric acid gives the
lamivudine and nevirapine.
reactions of chlorides (2.3.1).
Calculate the contents of C10H 13N5O4, C 8H11N 3O3S and
C15H14N4O in the medium. Tests
D. Not less than 70 per cent of the stated amounts of pH (2.4.24). 4.6 to 6.0, determined in a solution prepared by
C10H13N5O4, C8H11N3O3S and C15H14N4O. dissolving 1.0 g in 9 ml of freshly boiled and cooled water,
Other tests. Comply with the tests stated under Tablets. ignoring any slight turbidity.
Assay. Determine by liquid chromatography (2.4.14). Aluminium, calcium, heavy metals, iron and magnesium. To
Test solution. Weigh and powder 20 tablets. Weigh accurately 8 ml of solution A add 2 ml of strong ammonia solution and
a quantity of the powder containing about 100 mg of shake; the solution is clear (2.4.1), and colourless (2.4.1). Add
Zidovudine and transfer to a 200-ml volumetric flask. Add 1 ml of a 9 per cent w/v solution of disodium hydrogen
about 20 ml of methanol, mix with the aid of ultrasound for phosphate; the resulting solution remains clear for at least
10 minutes, dilute to volume with water, mix and filter. Further 5 minutes. Add 0.2 ml of sodium sulphide solution; a white
dilute 10.0 ml of the filtrate to 25.0 ml with water. precipitate is produced and the supernatant liquid remains
colourless.
Reference solution. Weigh accurately about 100 mg of
zidovudine RS, 50 mg of lamivudine RS and 65 mg of nevirapine Ammonium salts. To 5 ml of a 10 per cent w/v solution add
RS and transfer to a 200-ml volumetric flask. Add about 20 ml 1 M sodium hydroxide until the precipitate first formed is
of methanol, mix with the aid of ultrasound to dissolve, dilute redissolved and then warm the solution; no odour of ammonia
to volume with water and mix. Further dilute 10.0 ml of this is perceptible.
solution to 25.0 ml with water. Oxychlorides. Dissolve 1.5 g in 1.5 ml of carbon dioxide-free
Follow the chromatographic procedure described under water; the solution is not more opalescent than opalescence
Dissolution. standard OS2 (2.4.1). Add 7.5 ml of ethanol (95 per cent); the
Inject the reference solution. The tailing factor for the solution may become cloudy within 10 minutes but becomes
individual peaks due to zidovudine, lamivudine and nevirapine clear on the addition of 0.2 ml of 2 M hydrochloric acid.
is not more than 2.0 and the relative standard deviation for Sulphates (2.3.17). 15 ml of solution A complies with the limit
replicate injections of all the analyte peaks is not more than test for sulphates (225 ppm).
2.0 per cent.
Assay. Weigh accurately about 3.0 g, dissolve in 125 ml of
Inject separately the test solution and the reference solution water, add 3 g of ammonium chloride and add sufficient water
and measure the responses for the major peaks. to produce 250.0 ml. To 25.0 ml of the resulting solution add
Calculate the contents of C10H 13N5O4, C 8H11N 3O3S and 100 ml of water and 10 ml of strong ammonia-ammonium
C15H14N4O in the tablets. chloride solution. Titrate with 0.1 M disodium edetate, using

1263
ZINC OXIDE IP 2007

eriochrome black T solution as indicator until a deep blue Assay. Dissolve 0.15 g in 10 ml of 2 M acetic acid and dilute to
colour is obtained. 50 ml with water. To the resulting solution add about 50 mg of
1 ml of 0.1 M disodium edetate is equivalent to 0.01363 g of xylenol orange triturate and sufficient hexamine to produce
ZnCl2. violet-pink colour. Add a further 2 g of hexamine and titrate
with 0.1 M disodium edetate until the solution becomes yellow.
Storage. Store protected from moisture, in non-metallic
1 ml of 0.1 M disodium edetate is equivalent to 0.008138 g of
containers.
ZnO.
Storage. Store protected from moisture.
Zinc Oxide
ZnO Mol. Wt. 81.4
Zinc Oxide contains not less than 99.0 per cent and not
Zinc Oxide Cream
more than 100.5 per cent of ZnO, calculated on the ignited Zinc Cream
basis.
Zinc Oxide Cream contains 32 per cent w/v of Zinc Oxide in a
Description. A soft, white or faintly yellowish white suitable water-in-oil emulsified base.
amorphous powder, free from grittiness. It gradually absorbs
carbon dioxide from air. Zinc Oxide Cream contains not less than 30.0 per cent and not
more than 34.0 per cent w/w of zinc oxide, ZnO.
Identification
Identification
A. It becomes yellow when strongly heated; the yellow colour
disappears on cooling. The residue obtained in the Assay is yellow when hot and
white when cool.
B. Dissolve 0.1 g in 1.5 ml of 2 M hydrochloric acid and dilute
to 5 ml with water. The solution gives the reaction of zinc salts Tests
(2.3.1).
Other tests. Complies with the tests stated under Creams.
Tests Assay. Weigh accurately about 0.5 g in a porcelain dish, heat
Alkalinity. Shake 1.0 g with 10 ml of boiling water, add 0.1 ml gently over a small flame until the base is completely volatilised
of phenolphthalein solution and filter. If the filtrate is red, not or charred. Increase the heat until all the carbon is removed.
more than 0.3 ml of 0.1 M hydrochloric acid is required to Dissolve the residue in 10 ml of 2 M acetic acid and add
discharge the colour. sufficient water to produce 50 ml. To the resulting solution
add about 50 mg of xylenol orange triturate and sufficient
Carbonate and substances insoluble in acids. Dissolve 1.0 g hexamine to produce violet-pink colour. Add a further 2 g of
in 15 ml of 2 M hydrochloric acid; no effervescence is hexamine and titrate with 0.1 M disodium edetate until the
produced and the solution is not more opalescent than solution becomes yellow.
opalescence standard OS2 (2.4.1), and colourless (2.4.1).
1 ml of 0.1 M disodium edetate is equivalent to 0.008138 g of
Arsenic (2.3.10). Dissolve 2.0 g in 15 ml of brominated ZnO.
hydrochloric acid AsT and 45 ml of water and remove the
excess of bromine with a few drops of stannous chloride
solution AsT. The resulting solution complies with the limit
test for arsenic (5 ppm).
Zinc Stearate
Iron (2.3.14). Dissolve 0.1 g in a mixture of 5 ml of water and
1 ml of hydrochloric acid and dilute to 40 ml with water. The (C17H35COO)2 Zn Mol. Wt. 632.3
resulting solution complies with the limit test for iron Zinc Stearate consists mainly of zinc stearate but many
(400 ppm). Use 0.5 ml of thioglycollic acid in the test. contain variable proportions of zinc palmitate (C15H31COO)2
Lead. Dissolve 2 g in a mixture of 20 ml water and 5 ml of Zn, and zinc oleate (C17H33COO)2Zn.
glacial acetic acid and add 0.25 ml of potassium chromate Zinc Stearate contains not less than 10.0 per cent and not
solution; the solution remains clear. more than 12.0 per cent of zinc, Zn.
Loss on ignition (2.4.20). Not more than 1.0 per cent, determined Description. A fine, white, bulky, amorphous powder, free from
on 2.0 g by igniting at 500º. grittiness; odour, faint and characteristic.

1264
IP 2007 ZINC SULPHATE

Identification Assay. Weigh accurately about 1.0 g and boil with 50 ml of 2 M


acetic acid until the fatty acid layer which separates is clear,
A. To 5.0 g add 50 ml of ether and 40 ml of a 7.5 per cent v/v adding more water if necessary to maintain the original volume.
solution of nitric acid in distilled water and heat under a Cool, filter and wash the filter and the flask thoroughly with
reflux condenser until dissolution is complete. Allow to cool, water until the last washing is not acidic to blue litmus paper.
separate the aqueous layer and shake the ether layer with two To the combined filtrate and washings add about 50 mg of
quantities, each of 4 ml, of distilled water. Combine the xylenol orange triturate and sufficient hexamine to produce
washings with the aqueous layer, wash with 15 ml of ether violet-pink colour. Add a further 2 g of hexamine and titrate
and heat on a water-bath until ether is completely eliminated. with 0.1 M disodium edetate until the colour changes to yellow.
Allow to cool and dilute to 50.0 ml with distilled water (solution
A). Evaporate the ether layer to dryness and dry the residue 1 ml of 0.1 M disodium edetate is equivalent to 0.00654 g of
at 105º. The freezing point of the residue is not lower than 53º Zn.
(2.4.11).
B. Neutralise 5 ml of solution A to red litmus paper with 10 M
sodium hydroxide. The solution gives the reactions of zinc
salts (2.3.1). Zinc Sulphate
ZnSO4, 7H2O Mol. Wt. 287.5
Tests
Zinc Sulphate contains not less than 99.0 per cent and not
Appearance of solution. Solution A is not more intensely more than 104.0 per cent of ZnSO4, 7H2O.
coloured than reference solution YS6 (2.4.1).
Description. Colourless, transparent crystals or a white,
Acidity or alkalinity. Shake 1.0 g with 5 ml of ethanol (95 per
crystalline powder; odourless; efflorescent.
cent) and add 20 ml of carbon dioxide-free water and 0.1 ml of
phenol red solution. Not more than 0.3 ml of 0.1 M
Identification
hydrochloric acid or 0.1 ml of 0.1 M sodium hydroxide is
required to change the colour of the solution. Dissolve 2.5 g in sufficient carbon dioxide-free water to
Alkalis and alkaline earths. Add 1.0 g to a mixture of 25 ml of produce 50 ml (solution A). Solution A gives the reactions of
water and 5 ml of hydrochloric acid, boil, filter immediately zinc salts and sulphates (2.3.1).
and wash with 25 ml of hot water. Add dilute ammonia
solution to make the filtrate just alkaline and then add Tests
ammonium sulphide solution in excess to precipitate the zinc Appearance of solution. Solution A is clear (2.4.1), and
as zinc sulphide completely. Filter, add 0.5 ml of sulphuric colourless (2.4.1).
acid to the filtrate, evaporate to dryness and ignite to constant
weight; the residue weighs not more than 20 mg. pH (2.4.24). 4.4 to 5.6, determined in solution A.
Arsenic (2.3.10). Mix 5.0 g with 10 ml of bromine solution and Arsenic (2.3.10). Dissolve 1.0 g in 50 ml of water and add 10 ml
evaporate to dryness on a water-bath. Ignite gently, dissolve of stannated hydrochloric acid AsT. The resulting solution
the cooled residue, ignoring any carbon, in 50 ml of water and complies with the limit test for arsenic (10 ppm).
14 ml of brominated hydrochloric acid AsT and remove the Iron (2.3.14). Dissolve 0.4 g in 20 ml of water. The resulting
excess of bromine with 2 ml of stannous chloride solution solution complies with the limit test for iron (100 ppm).
AsT. The resulting solution complies with the limit test for
arsenic (2 ppm). Chlorides (2.3.12). 20 ml of solution A complies with the limit
test for chlorides (250 ppm).
Heavy metals (2.3.13). Heat 5.0 g with 40 ml of 2 M acetic acid
and allow to cool. Filter, wash the residue with two successive Assay. Weigh accurately about 0.5 g and dissolve in 5 ml of
quantities, each of 5 ml, of warm water and dilute the combined 2 M acetic acid and dilute to 50 ml with water. To the resulting
filtrate and washings to 100.0 ml with water. 12 ml of the solution solution add about 50 mg of xylenol orange triturate and
complies with the limit test for heavy metals, Method D sufficient hexamine to produce violet-pink colour. Add a further
(20 ppm). Use 1.0 ml of lead standard solution (10 ppm Pb) to 2 g of hexamine and titrate with 0.1 M disodium edetate until
prepare the standard. the colour changes to yellow.
Chlorides (2.3.12). 10 ml of solution A complies with the limit 1 ml of 0.1 M disodium edetate is equivalent to 0.02875 g of
test for chlorides (250 ppm). ZnSO4, 7H2O.
Sulphates (2.3.17). 2.5 ml of solution A complies with the limit Storage. Store protected from moisture, in non-metallic
test for sulphates (0.6 per cent). containers.

1265
ZINC SULPHATE EYE DROPS IP 2007

Zinc Sulphate Eye Drops B. Dissolve 0.1 g in a mixture of 2 ml of 1 M sulphuric acid and
5 ml of glacial acetic acid and add, dropwise, 0.25 ml of
Zinc Sulphate Eye Drops are a sterile solution containing potassium permanganate solution; the pink colour of
0.25 per cent w/v of Zinc Sulphate in Purified Water. permanganate is discharged.
Zinc Sulphate Eye Drops contain not less than 0.22 per cent C. A mixture of 1 ml of the aqueous layer reserved in test A and
and not more than 0.28 per cent w/v of zinc sulphate, ZnSO4, 4 ml of water gives the reaction of zinc salts (2.3.1).
7H2O.
D. Melting range (2.4.21). 115º to 121º.
Description. A clear, colourless solution.
Tests
Identification
Alkalinity. Mix 1.0 g with 5 ml of ethanol (95 per cent) and
Give the reactions of zinc salts and of sulphates (2.3.1). 0.5 ml of phenol red solution, add 50 ml of carbon dioxide-
free water and examine immediately; no reddish colour is
Tests produced.
Other tests. Comply with the tests stated under Eye Drops. Alkalis and alkaline earths. Add 1.0 g to a mixture of 25 ml of
Assay. To 5.0 ml add 50 ml of water and 5 ml of ammonia buffer water and 5 ml of hydrochloric acid, boil, filter immediately
pH 10.9 and titrate with 0.01 M disodium edetate using and wash with 25 ml of hot water. Add dilute ammonia
mordant black II solution as indicator. solution to make the filtrate just alkaline and then add
ammonium sulphate solution in excess to precipitate the zinc
1 ml of 0.01 M disodium edetate is equivalent to 0.002875 g of as zinc sulphide completely. Filter, add 0.5 ml of sulphuric
ZnSO4, 7H2O. acid to the filtrate, evaporate to dryness and ignite to constant
Storage. Store in containers of glass or any other non-metallic weight; the residue weighs not more than 20 mg.
material and sealed so as to exclude micro-organisms. Sulphates (2.3.17). To 0.25 g add a mixture of 10 ml of distilled
water and 5 ml of 2 M hydrochloric acid. Cool, filter and
dilute to 20 ml with distilled water. The resulting solution
complies with the limit test for sulphates (600 ppm).
Zinc Undecenoate
Degree of unsaturation. Dissolve 0.1 g in a mixture of 5 ml of
CH2 H2C 2 M hydrochloric acid and 30 ml of glacial acetic acid and
O O titrate with 0.05 M bromine using 0.05 ml of ethoxychrysoidine
Zn hydrochloride solution, added towards the end of the titration,
O O as indicator. Not less than 9.1 ml and not more than 9.4 ml of
C22H38O4Zn Mol. Wt. 431.9 0.05 M bromine is required to discharge the red colour.
Zinc Undecenoate is zinc di(undec-10-enoate). Loss on drying (2.4.19). Not more than 1.5 per cent, determined
on 0.5 g by drying in an oven at 105º.
Zinc Undecenoate contains not less than 98.0 per cent and
not more than 102.0 per cent of C22H38O4Zn, calculated on the Assay. Weigh accurately about 0.35 g, add 25 ml of 2 M acetic
dried basis. acid, heat to boiling, cool and dilute to 50 ml with water. Add
about 50 mg of xylenol orange triturate and sufficient
Description. A white or almost white, fine powder. hexamine to produce a violet-pink colour. Add a further 2 g of
hexamine and titrate with 0.1 M disodium edetate until the
Identification colour changes to yellow.
A. To 2.5 g add 10 ml of water and 10 ml of 1 M sulphuric acid 1 ml of 0.1 M disodium edetate is equivalent to 0.04319 g of
and extract with two quantities, each of 10 ml, of ether. Reserve C22H38O4Zn.
the aqueous layer for test C. Wash the combined ether extracts
Storage. Store protected from light and moisture.
with water and evaporate to dryness. To the residue add 2 ml
of freshly distilled aniline and boil under a reflux condenser
for 10 minutes, cool and add 30 ml of ether. Extract with three
quantities, each of 20 ml, of 2 M hydrochloric acid and then Zinc Undecenoate Ointment
with 20 ml of water. Evaporate the ether extract to dryness on
Zinc Undecylenate Ointment
a water-bath. The residue, after recrystallising twice from
ethanol (70 per cent) and drying at a pressure not exceeding Zinc Undecenoate Ointment contains 20 per cent w/v of Zinc
2 kPa for 3 hours, melts at about 67º (2.4.21). Undecenoate in a suitable ointment basis.

1266
IP 2007 ZINC UNDECENOATE OINTMENT

Zinc Undecenoate Ointment contains not less than 18.0 per 300 ml, saturate it with sodium chloride and shake the mixture.
cent and not more than 22.0 per cent of zinc undecenoate, Transfer the aqueous layer to a second separator and extract
C22H38O4Zn, and not less than 4.5 per cent and not more than with another 100 ml of ether. Wash the combined ether extracts
5.5 per cent of free undecenoic acid, C11H20O2. with successive quantities, each of 10 ml, of water until the
washings are free from chloride. Transfer the ether solution to
Tests a beaker and evaporate on a water-bath to about 5 ml. Add
20 ml of carbon tetrachloride, mix, transfer the mixture to a
Other tests. Complies with the tests stated under Ointments.
small separator and drain the carbon tetrachloride layer into
Assay. For zinc undecenoate — Weigh accurately about a 100-ml volumetric flask. Rinse the beaker with three
2.0 g, add 20 ml of dilute hydrochloric acid and boil under a quantities, each of 5 ml, of carbon tetrachloride and transfer
reflux condenser for at least 20 minutes or until the fatty layer the rinsings to the volumetric flask, dilute to volume with
is clear. Filter while hot and wash the residue with hot water. carbon tetrachloride and mix. Evaporate 50.0 ml of the resulting
Cool the combined filtrate and washings, neutralise to litmus solution to about 5 ml, add 100 ml of ethanol (95 per cent),
paper with dilute ammonia solution, add 3 ml of dilute previously neutralised, and 0.15 ml of phenolphthalein
hydrochloric acid and 5 g of hexamine and titrate with solution and titrate the total undecenoic acid with 0.1 M
0.05 M disodium edetate using xylenol orange solution as sodium hydroxide.
indicator, until the colour changes to yellow.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01843 g of
1 ml of 0.05 M disodium edetate is equivalent to 0.02160 g of C11H20O2.
C22H38O4Zn.
Calculate the content of free undecenoic acid from the
For free undecenoic acid — Weigh accurately about 5.0 g, difference between the total undecenoic acid and the
add 100 ml of dilute hydrochloric acid and heat to 70º with undecenoic acid equivalent to the determined zinc
constant stirring. Cool and transfer to a separator with the aid undecenoate (the content of zinc undecenoate multiplied by
of four quantities, each of 25 ml, of ether and add the rinsings 0.8533 gives the equivalent of undecenoic acid).
to the mixture in the separator. Dilute the aqueous phase to
Storage. Store protected from light and moisture.

1267
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

VACCINES AND IMMUNOSERA FOR HUMAN USE

General Requirements .....................................................................................................

Monographs .....................................................................................................................
Adsorbed Diphtheria, Tetanus and Hepatitis B (rDNA) Vaccine ........................................
Adsorbed Diphtheria, Tetanus and Pertussis (Acellular Component) and
Haemophilus Type b Conjugate Vaccine ........................................................................
Adsorbed Diphtheria, Tetanus and Pertussis (Acellular Component) and
Hepatitis B (rDNA) Vaccine ...........................................................................................
Adsorbed Diphtheria, Tetanus and Pertussis (Acellular Component),
Inactivated Poliomyelitis Vaccine and Haemophilus Type b Conjugate Vaccine .................
Adsorbed Diphtheria, Tetanus, Pertussis (Acellular Component) and
Inactivated Poliomyelitis Vaccine ...................................................................................
Adsorbed Diphtheria, Tetanus, Pertussis and Poliomyelitis (Inactivated) Vaccine ....................
Adsorbed Diphtheria, Tetanus, Pertussis, Poliomyelitis (Inactivated) and
Haemophilus Type b Conjugate Vaccine ........................................................................
Adsorbed Pertussis Vaccine (Acellular Component) .........................................................
Adsorbed Pertussis Vaccine (Acellular, Co-purified) .........................................................
Bacillus Calmette-Guerin Vaccine (Freeze-Dried) ...............................................................
Diphtheria and Tetanus Vaccine (Adsorbed) ........................................................................
Diphtheria and Tetanus Vaccine (Adsorbed) for Adults and Adolescents ..............................
Diphtheria, Tetanus and Pertussis Vaccine (Adsorbed) .....................................................
Diphtheria, Tetanus, Pertussis (Whole Cell), Hepatitis B (rDNA) and
Haemophilus Type b Conjugate Vaccine (Adsorbed) ....................................................
Diphtheria, Tetanus, Pertussis (Whole Cell) Hepatitis B (rDNA)
Vaccine (Adsorbed) ...................................................................................................
Diphtheria, Tetanus, Pertussis (Whole Cell) and Haemophilus Type b
Conjugate Vaccine (Adsorbed) ......................................................................................
Diphtheria Vaccine (Adsorbed) ...........................................................................................
Haemophilus Type b Conjugate Vaccine .............................................................................
Hepatitis A (Inactivated) and Hepatitis B (rDNA) Vaccine (Adsorbed) .................................
Hepatitis B Vaccine (rDNA) .............................................................................................
Inactivated Hepatitis A Vaccine (Adsorbed) .......................................................................
Inactivated Hepatitis B Vaccine ...........................................................................................

1269
MONOGRAPHS INDIAN PHARMACOPOEIA 2007

Inactivated Influenza Vaccine (Split Virion) ...........................................................................


Inactivated Influenza Vaccine (Surface Antigen) ....................................................................
Inactivated Influenza Vaccine (Whole Virion) ....................................................................
Japanese Encephalitis Vaccine (Human) ..............................................................................
Measles and Rubella Vaccine (Live) .....................................................................................
Measles Vaccine (Live) ......................................................................................................
Measles, Mumps and Rubella Vaccine (Live) .......................................................................
Meningococcal Polysaccharide Vaccine ..............................................................................
Mumps Vaccine (Live) ......................................................................................................
Pertussis Vaccine ................................................................................................................
Pneumococcal Polysaccharide Vaccine ................................................................................
Poliomyelitis Vaccine (Inactivated) ......................................................................................
Poliomyelitis Vaccine, Live (Oral) .......................................................................................
Rabies Vaccine, Human ......................................................................................................
Rubella Vaccine (Live) .......................................................................................................
Tetanus Vaccine (Adsorbed) ...............................................................................................
Tick-borne Encephalitis Vaccine (Inactivated) .....................................................................
Tuberculin Purified Protein Derivative ...................................................................................
Typhoid (Strain Ty 21a) Vaccine, Live (Oral) ....................................................................
Typhoid Polysaccharide Vaccine .........................................................................................
Typhoid Vaccine ................................................................................................................
Typhoid Vaccine (Freeze Dried) .........................................................................................
Varicella Vaccine (Live) .......................................................................................................
Viper Venom ........................................................................................................................
Yellow Fever Vaccine (Live) ...............................................................................................

1270
IP 2007 VACCINES : GENERAL REQUIREMENTS

Vaccines : General Requirements be freeze-dried so that the water content is not more than 3.0
per cent w/w unless otherwise stated in the individual
Vaccines are preparations of antigenic substances that are monograph. They may be standardized in terms of interopacity
administered for the purpose of inducing in the recipient a units or, where appropriate, by numbers of living or killed
specific and active immunity against the infective agent or bacteria determined by direct cell count or by viable count.
toxin produced by it.
Bacterial toxoids. Bacterial toxoids are toxins or material
Vaccines may contain living micro-organisms suitably treated derived therefrom, the toxicity of which has been reduced to a
to attenuate their virulence but retain their antigenic potency very low level or completely eliminated by chemical or physical
or they may consist of pathogenic organisms which have means without destroying their immunizing potency. The toxins
been killed or inactivated. Some vaccines consist of antigenic are obtained from selected strains of specific micro-organisms,
fractions or substances produced by the same pathogenic grown in media free from ingredients known to cause toxic,
organisms but rendered harmless whilst retaining their allergic or other undesirable immunological reactions in
antigenic efficiency. Vaccines may be prepared from one humans. Toxoids produced by the action of formaldehyde are
species only or from a mixture of two or more species. known as formol toxoids.
Vaccines may be prepared by the method described in the Bacterial toxoids may be liquid or may be prepared by adsorbing
individual monographs or by the general methods given below on mineral carriers such as aluminium phosphate, aluminium
or in any other manner provided the identity of the antigens is hydroxide or any other suitable adsorbent; the adsorbed
maintained and the vaccines are free from microbial product may be separated, washed and suspended in a saline
contamination and extraneous agents. Suitable adjuvants may or other appropriate solution isotonic with blood.
be added during the preparation but streptomycin, penicillin
or other β-lactam antibiotics may not be added at any stage of Bacterial toxoids are clear or slightly opalescent liquids,
manufacture or in the final vaccine. A suitable bactericide may colourless or slightly yellow. Adsorbed toxoids may be white
be added to sterile and inactivated vaccines. The final products or greyish white suspensions or pale-yellow liquids with a
are distributed aseptically into sterile containers which are sediment at the bottom of the container. Freeze-dried
then sealed to exclude extraneous micro-organisms. Unless preparations are greyish white or yellowish white powders or
otherwise indicated in the monograph, the final vaccine may pellets.
be filled into single dose or multiple dose containers but Viral and rickettsial vaccines. Viral and rickettsial vaccines
vaccines in multiple dose containers must invariably contain are suspensions of viruses or rickettsiae and are prepared
a bactericide. from infected tissues or blood obtained from artificially infected
Bacterial Vaccines. Bacterial vaccines are either sterile animals, from cultures in fertile eggs, or from cell or tissue
suspensions of live or killed bacteria or sterile extracts of culture. Viral vaccines may be live or killed and they may be
derivatives of bacteria. They may be simple vaccines prepared freeze-dried. Live vaccines are usually prepared using
from one species or may be mixed vaccines prepared by attenuated strains of the specific organisms. Killed vaccines
blending two or more simple vaccines from different species may be inactivated by suitable chemical or physical means.
or strains. Mixed Vaccines. Mixed vaccines are mixtures of two or more
Bacterial vaccines may be prepared from cultures grown on vaccines. A suitable antibacterial substance may be added to
suitable solid or liquid media. The whole culture or parts of it inactivated or live viral and rickettsial vaccines provided that
may be used in preparing the vaccine. The identity, antigenic it has no action against the specific organisms.
potency and purity of each bacterial culture must be carefully
controlled. Production
Vaccines containing killed organisms may be prepared by General provisions. Requirements for production including
killing the organisms by chemical or physical means provided in-process testing are included in individual monographs.
the antigenic potency of the vaccine is preserved. Vaccines Where justified and authorized, certain tests may be omitted
containing living bacteria may be prepared from strains which where it can be demonstrated, for example by validation
are avirulent for humans but which stimulate the production studies, that the production process consistently ensures
of antibodies active against pathogenic strains of the same compliance with the test.
species. The final vaccines must be free from any substance Unless otherwise justified and authorized, vaccines are
known to cause toxic, allergic or other undesirable produced using a seed-lot system. The methods of preparation
immunological reactions in humans. are designed to maintain adequate immunogenic properties,
Bacterial vaccines are suspensions of varying degrees of to render the preparation harmless and to prevent
opacity in colourless or slightly coloured liquids or they may contamination with extraneous agents.

1
VACCINES : GENERAL REQUIREMENTS IP 2007

Unless otherwise justified and authorized, in the production Control eggs. For live vaccines produced in eggs, control
of a final lot of vaccine, the number of passages of a virus, or eggs are incubated and tested as prescribed in the monograph.
the number of subcultures of a bacterium, from the master Purification. Where applicable, validated purification
seed lot shall not exceed that used for production of the vaccine procedures may be applied.
shown in clinical studies to be satisfactory with respect to
safety and efficacy. Inactivation. Inactivated vaccines are produced using a
validated inactivation process whose effectiveness and
Vaccines are as far as possible free from ingredients known to
consistency have been demonstrated. Where there are
cause toxic, allergic or other undesirable reactions in man.
recognised potential contaminants of a harvest, for example
Suitable additives, including stabilizers and adjuvants may be
in vaccines produced in eggs from healthy, non-SPF flocks,
incorporated. Penicillin and streptomycin are neither used at
the inactivation process is also validated with respect to the
any stage of production nor added to the final product;
potential contaminants. A test for inactivation is carried out
however, master seed lots prepared with media containing
as soon as possible after the inactivation process, unless
penicillin or streptomycin may, where justified and authorized,
otherwise justified and authorised.
be used for production.
Intermediates. Where applicable, the stability of intermediates
Substrates for propagation. Substrates for propagation in given storage conditions shall be evaluated and a period of
comply with the relevant requirements of the Pharmacopoeia validity established.
or in the absence of such requirements with those of the
competent authority. Processing of cell banks and subsequent Final bulk. The final bulk is prepared by aseptically blending
cell cultures is done under aseptic conditions in an area where the ingredients of the vaccine.
no other cells are being handled. Serum and trypsin used in Adsorbents. Vaccines may be adsorbed on aluminium
the preparation of cell suspensions shall be shown to be free hydroxide, aluminium phosphate, calcium phosphate or other
from extraneous agents. suitable adsorbent; the adsorbents are prepared in special
Seed lot. The strain of bacterium or virus used in a master conditions which confer the appropriate physical form and
seed lot is identified by historical records that include adsorptive properties.
information on the origin of the strain and its subsequent Antimicrobial preservative. A suitable antimicrobial
manipulation. No micro-organism other than the seed strain preservative may be included in sterile and inactivated
shall be present in a seed lot. vaccines and is invariably added if these preparations are
Culture media. Culture media are as far as possible free from issued in multidose containers, unless otherwise stated. If an
ingredients known to cause toxic, allergic or other undesirable antimicrobial preservative is used, it shall be shown that it
reactions in man; if inclusion of such ingredients is necessary, does not impair the safety or efficacy of the vaccine and its
it shall be demonstrated that the amount present in the final effectiveness throughout the period of validity shall be
lot is reduced to such a level as to render the product safe. demonstrated.
Approved animal (but not human) serum may be used in the Final lot. For vaccines for parenteral administration, the final
growth medium for cell cultures but the medium used for lot is prepared by aseptically distributing the final bulk into
maintaining cell growth during virus multiplication shall not sterile tamper-proof containers which, after freeze-drying where
contain serum, unless otherwise stated. Cell culture media applicable, are closed so as to exclude contamination. For
may contain a pH indicator such as phenol red and approved vaccines for administration by a non-parenteral route, the final
antibiotics at the lowest effective concentration although it is lot is prepared by distributing the final bulk under suitable
preferable to have a medium free from antibiotics during conditions into sterile, tamper-proof containers.
production.
Stability. Maintenance of potency of the final lot throughout
Propagation and harvest. The seed cultures are propagated the period of validity shall be demonstrated by validation
and harvested under defined conditions. The purity of the studies; the loss of potency in the recommended storage
harvest is verified by suitable tests as defined in the conditions is assessed and excessive loss even within the
monograph. limits of acceptable potency may indicate that the vaccine is
Control cells. For vaccines produced in cell cultures, control unacceptable.
cells are maintained and tested as prescribed. In order to Degree of adsorption. During development of an adsorbed
provide a valid control, these cells must be maintained in vaccine, the degree of adsorption is evaluated as part of the
conditions that are rigorously identical with those used for consistency testing. A release specification for the degree of
the production cell cultures, including use of the same batches adsorption is established in the light of results found for
of media and media changes. batches used in clinical testing. From the stability data

2
IP 2007 ADSORBED DIPHTHERIA, TETANUS AND HEPATITIS B (rDNA) VACCINE

generated for the vaccine it must be shown that at the end of Adsorbed Diphtheria, Tetanus and
the period of validity the degree of adsorption will not be less
than for batches used in clinical testing. Hepatitis B (rDNA) Vaccine
Diphtheria, Tetanus and Hepatitis B (rDNA) Vaccine
Tests
(Adsorbed) is a combined vaccine composed of: diphtheria
Vaccines, reconstituted where necessary, comply with the formol toxoid; tetanus formol toxoid; hepatitis B surface
following requirements unless otherwise stated in the antigen (HBsAg); a mineral adsorbent such as aluminium
individual monograph. hydroxide or hydrated aluminium phosphate.
Phenol (If present) (2.3.36). Not more than 0.25 per cent w/v. The formol toxoids are prepared from the toxins produced by
the growth of Corynebacterium diphtheriae and Clostridium
Thiomersal (If present) (2.3.48). Between 0.005 per cent w/v
tetani, respectively.
and 0.02 per cent w/v.
HBsAg is a component protein of hepatitis B virus; the antigen
Free formaldehyde (If present) (2.3.20). Maximum 0.02 g/l.
is obtained by recombinant DNA technology.
Aluminium (If present) (2.3.9). Not more than 1.25 mg per
dose. Production
Sterility (2.2.11). Unless otherwise stated all vaccines comply General provisions
with tests for sterility, except that for living bacterial vaccines,
growth of the organism from which the vaccine was prepared The production method shall have been shown to yield
is permitted (sterility means abesence of becterial and fungal consistently vaccines comparable with the vaccine of proven
contaminants except where specified in the individual clinical efficacy and safety in man.
monograph). The production method is validated to demonstrate that the
Abnormal toxicity (2.2.1). Unless otherwise stated, all vaccines product, if tested, would comply with the test for abnormal
comply with the test for abnormal toxicity, Method B. In toxicity for antisera and vaccines, and with the following test
vaccines containing phenol as preservative, the test in mice for specific toxicity of the diphtheria and tetanus components:
may be inappropriate. inject subcutaneously 5 times the single human dose stated
on the label into each of 5 healthy guinea-pigs, each weighing
NOTE — The statements given in this general chapter is between 250 and 350 g, that have not previously been treated
intended to be read in conjunction with the monographs on with any material that will interfere with the test. If within 42
the individual vaccine in this Pharmacopoeia which refer to days of the injection any of the animals shows signs of or dies
preparations for human use; they do not necessarily apply from diphtheria toxaemia or tetanus, the vaccine does not
to vaccines for use in veterinary practice. comply with the test. If more than 1 animal dies from non-
Storage. Liquid vaccines must be stored at a temperature specific causes, repeat the test once; if more than 1 animal
between 2o and 8o and should not be allowed to freeze unless dies in the second test, the vaccine does not comply with the
otherwise specified in the individual monograph. Freeze-dried test.
preparations must be stored at temperatures below –20o or as The content of bacterial endotoxins in the bulk purified
specified in the individual monograph. At higher temperatures diphtheria toxoid and tetanus toxoid is determined to monitor
vaccines deteriorate rapidly. the purification procedure and to limit the amount in the final
Labelling. The label states (1) for liquid vaccines, the total vaccine. For each component, the content of bacterial
number of ml in the container and, for dried vaccines, the endotoxin is less than the limit approved for the particular
number of doses in the container; (2) unless otherwise vaccine and in any case the contents are such that the final
indicated the minimum number of Units per dose or per ml or, vaccine contains less than 100 IU per single human dose.
for viral vaccines, the minimum viral titre; (3) the dose and
route of administration; (4) the name and proportion of any Reference vaccine(s)
antibacterial preservative or other auxiliary substances added Provided valid assays can be performed, monocomponent
to the vaccine; (5) the date after which the vaccine is not reference vaccines may be used for the assays on the combined
intended to be used; (6) the conditions under which it should vaccine. If this is not possible because of interaction between
be stored; (7) for dried vaccines, the liquid to be used for the components of the combined vaccine or because of the
reconstitution and its volume; (8) that the vaccine should be difference in composition between monocomponent reference
used immediately after reconstitution; (9) unless otherwise vaccine and the test vaccine, a batch of combined vaccine
directed, that the vaccine should be shaken well before use; shown to be effective in clinical trials or a batch representative
(10) any contraindication to the use of the vaccine. thereof is used as a reference vaccine. For the preparation of

3
ADSORBED DIPHTHERIA, TETANUS AND HEPATITIS B (rDNA) VACCINE IP 2007

a representative batch, strict adherence to the production vaccines, is given as an example. Dissolve in the vaccine under
process used for the batch tested in clinical trials is necessary. examination sufficient sodium citrate to give a 10 per cent
The reference vaccine may be stabilised by a method that has w/v solution. Maintain at 37° for about 16 hours and centrifuge
been shown to have no effect on the assay procedure. until a clear supernatant is obtained. The clear supernatant
liquid reacts with a suitable diphtheria antitoxin, giving a
Production of the components precipitate.
The production of the components complies with the B. Tetanus toxoid is identified by a suitable immunochemical
requirements of the monographs on Diphtheria Vaccine method (2.2.14). The following method, applicable to certain
(Adsorbed), Tetanus Vaccine (Adsorbed) and Hepatitis B vaccines, is given as an example. The clear supernatant liquid
Vaccine (rDNA). obtained during identification test A reacts with a suitable
tetanus antitoxin, giving a precipitate.
FINAL BULK VACCINE
C. The assay or, where applicable, the electrophoretic profile,
The final bulk vaccine is prepared by adsorption, separately
serves also to identify the hepatitis B component of the
or together, of suitable quantities of bulk purified diphtheria
vaccine.
toxoid, tetanus toxoid and HBsAg onto a mineral carrier such
as aluminium hydroxide or hydrated aluminium phosphate. Tests
Suitable antimicrobial preservatives may be added. Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
Only a final bulk vaccine that complies with the following if aluminium hydroxide or hydrated aluminium phosphate is
requirements may be used in the preparation of the final lot. used as the adsorbent.
Antimicrobial preservative. Where applicable, determine the Free formaldehyde (2.3.20). Maximum 0.2 g/l.
amount of antimicrobial preservative by a suitable chemical Antimicrobial preservative. Where applicable, determine the
method. The content is not less than 85.0 per cent and not amount of antimicrobial preservative by a suitable chemical
greater than 115.0 per cent of the intended amount. method. The content is not less than 85.0 per cent and not
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk greater than 115.0 per cent of the intended amount.
for each sterility medium.
Sterility (2.2.11). Complies with the test for sterility.
FINAL LOT Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
Only a final lot that is satisfactory with respect to the test for the equivalent of 1 human dose into each rabbit.
osmolality and with respect to each of the requirements given Assay
below under Identification, Tests and Assay may be released
Diphtheria component
for use.
Carry out one of the prescribed methods for the assay as
Provided the test for antimicrobial preservative and the assays
stated under Diphtheria Vaccine (Adsorbed).
for the diphtheria and tetanus components have been carried
out with satisfactory results on the final bulk vaccine, they The lower confidence limit (P = 0.95) of the estimated potency
may be omitted on the final lot. is not less than 30 IU per single human dose.
Provided the content of free formaldehyde has been Tetanus component
determined on the bulk purified antigens or on the final bulk
Carry out one of the prescribed methods for the assay as
and it has been shown that the content in the final lot will not
stated under Tetanus Vaccine (Adsorbed).
exceed 0.2 g/l, the test for free formaldehyde may be omitted
on the final lot. The lower confidence limit (P = 0.95) of the estimated potency
is not less than 40 IU per single human dose.
If an in vivo assay is used for the hepatitis B component,
provided it has been carried out with satisfactory results on Hepatitis B component
the final bulk vaccine, it may be omitted on the final lot.
It complies with the assay of Hepatitis B Vaccine.
Osmolality (2.4.23). The osmolality of the vaccine is within
the limits approved for the particular preparation. Labelling. The label states (1) the minimum number of
International Units of diphtheria and tetanus toxoid per single
Identification human dose; (2) the amount of HBsAg per single human dose;
(3) the type of cells used for production of the HBsAg
A. Diphtheria toxoid is identified by a suitable immunochemical component; (4) where applicable, that the vaccine is intended
method (2.2.14). The following method, applicable to certain for primary vaccination of children and is not necessarily

4
IP 2007 A. D. T. P. (ACELLULAR COMPONENT) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE

suitable for reinforcing doses or for administration to adults; consistently the vaccines comparable with the vaccine of
(5) the name and the amount of the adsorbent; (6) that the proven clinical efficacy and safety in man.
vaccine must be shaken before use; (7) that the vaccine is not If the vaccine is presented with the haemophilus component
to be frozen. in a separate vial, as part of consistency studies the assays of
the diphtheria, tetanus and pertussis components are carried
out on a suitable number of batches of vaccine reconstituted
Adsorbed Diphtheria, Tetanus, for use. For subsequent routine control, the assays of these
components may be carried out without mixing with the
Pertussis (Acellular Component) and haemophilus component.
Haemophilus Type B Conjugate The content of bacterial endotoxins in bulk purified diphtheria
Vaccine toxoid, tetanus toxoid, pertussis components and PRP
conjugate is determined to monitor the purification procedure
Diphtheria, Tetanus, Pertussis (Acellular Component) and and to limit the amount in the final vaccine. For each
Haemophilus Type b Conjugate Vaccine (Adsorbed) is a component, the content of bacterial endotoxins is less than
combined vaccine composed of: diphtheria formol toxoid; the limit approved for the particular vaccine; if the vaccine is
tetanus formol toxoid; individually purified antigenic presented with the haemophilus component in a separate
components of Bordetella pertussis; polyribosylribitol container, the contents of the diphtheria, tetanus and pertussis
phosphate (PRP) covalently bound to a carrier protein; a antigens are in any case such that the final vial for these
mineral absorbent such as aluminium hydroxide or hydrated components contains less than 100 IU per single human dose.
aluminium phosphate. The product may be presented with
the haemophilus type b component in a separate container, The production method is validated to demonstrate that the
the contents of which are mixed with the other components product, if tested, would comply with the test for abnormal
immediately before use. toxicity for antisera and vaccines.
The formol toxoids are prepared from the toxins produced by During development studies and wherever revalidation is
the growth of Corynebacterium diphtheriae and Clostridium necessary, it shall be demonstrated by tests in animals that
tetani respectively. the vaccine induces a T-cell dependent B-cell immune response
to PRP.
The vaccine contains either pertussis toxoid or a pertussis-
toxin-like protein free from toxic properties produced by Reference vaccine
expression of a genetically modified form of the corresponding Provided valid assays can be performed, monocomponent
gene. Pertussis toxoid is prepared from pertussis toxin by a reference vaccines may be used for the assays on the combined
method that renders the toxin harmless while maintaining vaccine. If this is not possible because of interaction between
adequate immunogenic properties and avoiding reversion to the components of the combined vaccine or because of the
toxin. The acellular pertussis component may also contain difference in composition between monocomponent reference
filamentous haemagglutinin, pertactin (a 69 kDa outer- vaccine and the test vaccine, a batch of combined vaccine
membrane protein) and other defined components of shown to be effective in clinical trials or a batch representative
B. pertussis such as fimbrial-2 and fimbrial-3 antigens. The thereof is used as a reference vaccine. For the preparation of
latter two antigens may be copurified. The antigenic a representative batch, strict adherence to the production
composition and characteristics are based on evidence of process used for the batch tested in clinical trials is necessary.
protection and freedom from unexpected reactions in the target The reference vaccine may be stabilised by a method that has
group for which the vaccine is intended. been shown to have no effect on the assay procedure.
PRP is a linear copolymer composed of repeated units of
Production of the components
3-β-D-ribofuranosyl-(1→1)-ribitol-5-phosphate
[(C10H19O12P)n], with a defined molecular size and derived from The production of the components complies with the tests of
a suitable strain of Haemophilus influenzae type b. The carrier the monographs on Diphtheria Vaccine (Adsorbed), Tetanus
protein, when conjugated to PRP, is capable of inducing a Vaccine (Adsorbed), Pertussis Vaccine (Acellular Component,
T-cell-dependent B-cell immune response to the Adsorbed) and Haemophilus Type b Conjugate Vaccine.
polysaccharide.
FINAL BULK VACCINE
Production
Different methods of preparation may be used: a final bulk
General provisions
vaccine may be prepared by adsorption, separately or together,
The production method shall have been shown to yield of suitable quantities of bulk purified diphtheria toxoid, tetanus

5
A. D. T. P. (ACELLULAR COMPONENT) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE IP 2007

toxoid, acellular pertussis components and PRP conjugate w/v solution. Maintain at 37° for about 16 h and centrifuge
onto a mineral carrier such as aluminium hydroxide or hydrated until a clear supernatant is obtained. The clear supernatant
aluminium phosphate; or 2 final bulks may be prepared and reacts with a suitable diphtheria antitoxin, giving a precipitate.
filled separately, one containing the diphtheria, tetanus and B. Tetanus toxoid is identified by a suitable immunochemical
pertussis components, the other the haemophilus component, method (2.2.14). The following method, applicable to certain
which may be freeze-dried. Suitable antimicrobial preservatives vaccines, is given as an example. The clear s u p e r n a t a n t
may be added. obtained as described in Identification test A reacts with a
Only a final bulk vaccine that complies with the following suitable tetanus antitoxin, giving a precipitate.
requirements may be used in the preparation of the final lot. C. The pertussis components are identified by a suitable
Antimicrobial preservative. Where applicable, determine the immunochemical method (2.2.14). The following method,
amount of antimicrobial preservative by a suitable chemical applicable to certain vaccines, is given as an example. The
method. The content is not less than 85.0 per cent and not clear supernatant obtained as described in Identification
greater than 115.0 per cent of the intended amount. test A reacts with a specific antisera to the pertussis
components of the vaccine.
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
for each sterility medium. D. The haemophilus component is identified by a suitable
immunochemical method (2.2.14) for PRP.
FINAL LOT
Only a final lot that is satisfactory with respect to the test for
Tests
osmolality shown below and with respect to each of the If the product is presented with the haemophilus component
requirements given below under Identification, Tests and in a separate container: the tests for absence of residual
Assay may be released for use. pertussis toxin, irreversibility of pertussis toxoid, aluminium,
Provided the tests for absence of residual pertussis toxin, free formaldehyde, antimicrobial preservative and sterility are
irreversibility of pertussis toxoid and antimicrobial preservative carried out on the container with the diphtheria, tetanus and
and the assays have been carried out with satisfactory results pertussis components; the tests for PRP content, water (where
on the final bulk vaccine, they may be omitted on the final lot. applicable), sterility and pyrogens are carried out on the
container with the haemophilus component.
Provided the free formaldehyde content has been determined
on the bulk purified antigens or the final bulk and it has been If the haemophilus component is freeze-dried, some tests may
shown that the content in the final lot will not exceed 0.2 g/l, be carried out on the freeze-dried product rather than on the
the test for free formaldehyde may be omitted on the final lot. bulk conjugate where the freeze-drying process may affect
the component under test.
Osmolality (2.4.23). The osmolality of the vaccine,
reconstituted where applicable, is within the limits approved Absence of residual pertussis toxin and irreversibility
for the particular preparation. of pertussis toxoid
pH (2.4.24). The pH of the vaccine, reconstituted if necessary, This test is not necessary for the product obtained by genetic
is within the range approved for the particular product. modification. Use 3 groups each of not less than 5 histamine-
sensitive mice. Inject intraperitoneally into each mouse of the
Free PRP. Unbound PRP is determined after removal of the first group twice the single human dose of the vaccine stored
conjugate, for example by anion-exchange, size-exclusion or at 2° to 8°. Inject intraperitoneally into each mouse of the
hydrophobic chromatography (2.4.16), ultrafiltration or other second group twice the single human dose of the vaccine
validated methods. The amount of free PRP is not greater than incubated at 37° for 4 weeks. Inject diluent into the third group
that approved for the particular product. of mice. After 5 days, inject into each mouse 2 mg of histamine
Identification base intraperitoneally in a volume not exceeding 0.5 ml and
observe for 24 h. The test is invalid if 1 or more control mice
If the vaccine is presented with the haemophilus component die following histamine challenge. The vaccine complies with
in a separate vial: identification tests A, B and C are carried the test if no animal in the first or second group dies following
out using the vial containing the diphtheria, tetanus and histamine challenge. If 1 mouse dies in either or both of the
pertussis components; identification test D is carried out on first and second groups, the test may be repeated with the
the vial containing the haemophilus components. same number of mice or with a greater number and the results
A. Diphtheria toxoid is identified by a suitable immunochemical of valid tests combined; the vaccine complies with the test if,
method (2.2.14). The following method, applicable to certain in both of the groups given the vaccine, not more than 5.0 per
vaccines, is given as an example. Dissolve in the vaccine under cent of the total number of mice die following histamine
examination sufficient sodium citrate to give a 10 per cent challenge.

6
IP 2007 A. D. T. AND P. (ACELLULAR COMPONENT) AND HEPATITIS B (rDNA) VACCINE

The histamine sensitivity of the strain of mice used is verified Labelling. The label states (1) the minimum number of
at suitable intervals as follows: inject intravenously three- International Units of diphtheria and tetanus toxoid per single
fold dilutions of a reference pertussis toxin preparation in human dose; (2) the names and amounts of the pertussis
phosphate-buffered saline solution containing 0.2 per cent components per single human dose; (3) the number of
w/v of gelatin and challenge with histamine as above; the micrograms of PRP per single human dose; (4) the type and
strain is suitable if more than 50 per cent of the animals are nominal amount of carrier protein per single human dose; (5)
sensitised by 50 ng of pertussis toxin and none of the control where applicable, that the vaccine is intended for primary
animals injected with only diluent and challenged similarly vaccination of children and is not necessarily suitable for
with histamine shows symptoms of sensitisation. reinforcing doses or for administration to adults; (6) the name
PRP. Minimum 80.0 per cent of the amount of PRP stated on and the amount of the adsorbent; (7) that the vaccine must be
the label. PRP is determined either by assay of ribose (2.7.1) or shaken before use; (8) that the vaccine is not to be frozen (9)
phosphorus (2.7.1), by an immunochemical method (2.2.14) or where applicable, that the vaccine contains a pertussis toxin-
by anion-exchange liquid chromatography with pulsed- like protein produced by genetic modification.
amperometric detection.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
if aluminium hydroxide or hydrated aluminium phosphate is Adsorbed Diphtheria, Tetanus and
used as the adsorbent. Pertussis (Acellular Component) and
Free formaldehyde (2.3.20 ). Maximum 0.2 g/l. Hepatitis B (rDNA) Vaccine
Antimicrobial preservative. Where applicable, determine the
Diphtheria, Tetanus, Pertussis (Acellular Component) and
amount of antimicrobial preservative by a suitable chemical
Hepatitis B (rDNA) Vaccine (Adsorbed) is a combined vaccine
method. The content is not less than 85.0 per cent and not
composed of: diphtheria formol toxoid; tetanus formol toxoid;
greater than 115.0 per cent of the intended amount.
individually purified antigenic components of Bordetella
Water (2.3.43). Maximum 3.0 per cent for the freeze-dried pertussis; hepatitis B surface antigen; a mineral adsorbent
haemophilus component. such as aluminium hydroxide or hydrated aluminium
Sterility (2.2.11). Complies with the test for sterility. phosphate.

Pyrogens (2.2.8). Complies with the test for pyrogens. Inject The formol toxoids are prepared from the toxins produced by
per kg of the rabbit’s mass a quantity of the vaccine equivalent the growth of Corynebacterium diphtheriae and Clostridium
to: 1 mg of PRP for a vaccine with diphtheria toxoid or CRM tetani, respectively.
197 diphtheria protein as carrier; 0.1 mg of PRP for a vaccine The vaccine contains either pertussis toxoid or a pertussis-
with tetanus toxoid as carrier; 0.025 mg of PRP for a vaccine toxin-like protein free from toxic properties, produced by
with OMP as carrier. expression of a genetically modified form of the corresponding
Assay gene. Pertussis toxoid is prepared from pertussis toxin by a
method that renders the latter harmless while maintaining
Diphtheria component adequate immunogenic properties and avoiding reversion to
Carry out one of the prescribed methods for the assay as toxin. The vaccine may also contain filamentous
stated under Diphtheria Vaccine (Adsorbed). haemagglutinin, pertactin (a 69 kDa outer-membrane protein)
The lower confidence limit (P = 0.95) of the estimated potency and other defined components of B. pertussis such as fimbrial-
is not less than the minimum potency stated on the label. 2 and fimbrial-3 antigens. The latter 2 antigens may be
copurified. The antigenic composition and characteristics are
Unless otherwise justified and authorised, the minimum based on evidence of protection and freedom from unexpected
potency stated on the label is 30 IU per single human dose. reactions in the target group for which the vaccine is intended.
Tetanus component Hepatitis B surface antigen is a component protein of hepatitis
Carry out one of the prescribed methods for the assay as B virus; the antigen is obtained by recombinant DNA
stated under Tetanus Vaccine (Adsorbed). technology.
The lower confidence limit (P = 0.95) of the estimated potency Production
is not less than 40 IU per single human dose.
Pertussis component General provisions
The vaccine complies with the assay as the stated Adsorbed The production method shall have been shown to yield
Pertussis Vaccine (Acellular Component). consistently vaccines comparable with the vaccine of proven

7
A. D. T. AND P. (ACELLULAR COMPONENT) AND HEPATITIS B (rDNA) VACCINE IP 2007

clinical efficacy and safety in man. Provided the tests for absence of residual pertussis toxin,
The content of bacterial endotoxins in the bulk purified irreversibility of pertussis toxoid and antimicrobial preservative
diphtheria toxoid, tetanus toxoid and pertussis components and the assays for the diphtheria, tetanus and pertussis
is determined to monitor the purification procedure and to components have been carried out with satisfactory results
limit the amount in the final vaccine. For each component, the on the final bulk vaccine, they may be omitted on the final lot.
content of bacterial endotoxins is less than the limit approved Provided the content of free formaldehyde has been
for the particular vaccine. determined on the bulk purified antigens or on the final bulk
and it has been shown that the content in the final lot will not
Reference vaccine(s) exceed 0.2 g/l, the test for free formaldehyde may be omitted
Provided valid assays can be performed, monocomponent on the final lot.
reference vaccines may be used for the assays on the combined If an in vivo assay is used for the hepatitis B component,
vaccine. If this is not possible because of interaction between provided it has been carried out with satisfactory results on
the components of the combined vaccine or because of the the final bulk vaccine, it may be omitted on the final lot.
difference in composition between monocomponent reference
vaccine and the test vaccine, a batch of combined vaccine Osmolality (2.2.23). The osmolality of the vaccine is within
shown to be effective in clinical trials or a batch representative the limits approved for the particular preparation.
thereof is used as a reference vaccine. For the preparation of Identification
a representative batch, strict adherence to the production
process used for the batch tested in clinical trials is necessary. A. Diphtheria toxoid is identified by a suitable immunochemical
The reference vaccine may be stabilised by a method that has method (2.2.14). The following method, applicable to certain
been shown to have no effect on the assay procedure. vaccines, is given as an example. Dissolve in the vaccine under
examination sufficient sodium citrate to give a 10 per cent
Production of the components w/v solution. Maintain at 37° for about 16 h and centrifuge
until a clear supernatant is obtained. The clear supernatant
The production of the components complies with the reacts with a suitable diphtheria antitoxin, giving a precipitate.
requirements of the monographs on Diphtheria Vaccine
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine B. Tetanus toxoid is identified by a suitable immunochemical
(Acellular Component, Adsorbed) and Hepatitis B Vaccine method (2.2.14). The following method, applicable to certain
(rDNA). vaccines, is given as an example. The clear supernatant
obtained as described in identification test A reacts with a
FINAL BULK VACCINE suitable tetanus antitoxin, giving a precipitate.
The final bulk vaccine is prepared by adsorption, separately C. The pertussis components are identified by a suitable
or together, of suitable quantities of bulk purified diphtheria immunochemical method (2.2.14). The following method,
toxoid, tetanus toxoid, acellular pertussis components and applicable to certain vaccines, is given as an example. The
hepatitis B surface antigen onto a mineral carrier such as clear supernatant obtained as described in identification test
aluminium hydroxide or hydrated aluminium phosphate. A reacts with a specific antisera to the pertussis components
Suitable antimicrobial preservatives may be added. of the vaccine.
Only a final bulk vaccine that complies with the following D. The assay or, where applicable, the electrophoretic profile,
requirements may be used in the preparation of the final lot. serves also to identify the hepatitis B component of the
vaccine.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical Tests
method. The content is not less than 85.0 per cent and not Absence of residual pertussis toxin and irreversibility
greater than 115.0 per cent of the intended amount. of pertussis toxoid
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk This test is not necessary for the product obtained by genetic
for each sterility medium. modification. Use 3 groups each of not less than 5 histamine-
sensitive mice. Inject intraperitoneally into each mouse of the
FINAL LOT
first group twice the single human dose of the vaccine stored
Only a final lot that is satisfactory with respect to the test for at 2° to 8°. Inject intraperitoneally into each mouse of the
osmolality and with respect to each of the requirements given second group twice the single human dose of the vaccine
below under Identification, Tests and Assay may be released incubated at 37° for 4 weeks. Inject diluent into the third group
for use. of mice. After 5 days, inject into each mouse 2 mg of histamine

8
IP 2007 A. D. T. P. (ACELL. COMPONENT), INACTIVATED POLIOMYLITIS VACCINE AND HAEMOPHILUS TYPE B CONJUGATE

base intraperitoneally in a volume not exceeding 0.5 ml and The vaccine complies with the assay as stated under Adsorbed
observe for 24 h. The test is invalid if 1 or more control mice Pertussis Vaccine (Acellular Component).
die following histamine challenge. The vaccine complies with
the test if no animal in the first or second group dies following Hepatitis B component
histamine challenge. If 1 mouse dies in either or both of the The vaccine complies with the assay as stated under Hepatitis
first and second groups, the test may be repeated with the B Vaccine (rDNA).
same number of mice or with a greater number and the results
of valid tests combined; the vaccine complies with the test if, Labelling. The label states (1) the minimum number of
in both of the groups given the vaccine, not more than International Units of diphtheria and tetanus toxoid per single
5 per cent of the total number of mice die following histamine human dose; (2) the names and amounts of the pertussis
challenge. components per single human dose; (3) the amount of HBsAg
per single human dose; (4) the type of cells used for production
The histamine sensitivity of the strain of mice used is verified of the hepatitis B component; (5) where applicable, that the
at suitable intervals as follows: inject intravenously threefold vaccine is intended for primary vaccination of children and is
dilutions of a reference pertussis toxin preparation in not necessarily suitable for reinforcing doses or for
phosphate-buffered saline solution containing 0.2 per cent administration to adults; (6) the name and the amount of the
w/v of gelatin and challenge with histamine as above; the adsorbent; (7) that the vaccine must be shaken before use; (8)
strain is suitable if more than 50.0 per cent of the animals are that the vaccine is not to be frozen (9) where applicable, that
sensitised by 50 ng of pertussis toxin and none of the control the vaccine contains a pertussis toxin-like protein produced
animals injected with only diluent and challenged similarly by genetic modification.
with histamine shows symptoms of sensitisation.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
if aluminium hydroxide or hydrated aluminium phosphate is Adsorbed Diphtheria, Tetanus,
used as the adsorbent.
Pertussis (Acellular Component),
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
Inactivated Poliomyelitis Vaccine and
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical Haemophilus Type b Conjugate
method. The content is not less than 85.0 per cent and not Vaccine
greater than 115.0 per cent of the intended amount.
Diphtheria, Tetanus, Pertussis (Acellular Component) and
Sterility ( 2.2.11). Complies with the test for sterility. Haemophilus Type b Conjugate Vaccine (Adsorbed) is a
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject combined vaccine composed of: diphtheria formol toxoid;
the equivalent of 1 human dose into each rabbit. tetanus formol toxoid; individually purified antigenic
components of Bordetella pertussis; polyribosylribitol
Assay phosphate (PRP) covalently bound to a carrier protein; a
mineral absorbent such as aluminium hydroxide or hydrated
Diphtheria component aluminium phosphate. The product may be presented with
Carry out one of the prescribed methods for the assay as the haemophilus type b component in a separate container,
stated under Diphtheria Vaccine (Adsorbed). the contents of which are mixed with the other components
immediately before use.
The lower confidence limit (P = 0.95) of the estimated potency
is not less than the minimum potency stated on the label. The formol toxoids are prepared from the toxins produced by
the growth of Corynebacterium diphtheriae and Clostridium
Unless otherwise justified and authorised, the minimum tetani respectively.
potency stated on the label is 30 IU per single human dose.
The vaccine contains either pertussis toxoid or a pertussis-
Tetanus component toxin-like protein free from toxic properties produced by
Carry out one of the prescribed methods for the assay as expression of a genetically modified form of the corresponding
stated under Tetanus Vaccine (Adsorbed). gene. Pertussis toxoid is prepared from pertussis toxin by a
method that renders the toxin harmless while maintaining
The lower confidence limit (P = 0.95) of the estimated potency adequate immunogenic properties and avoiding reversion to
is not less than 40 IU per single human dose. toxin. The acellular pertussis component may also contain
Pertussis component filamentous haemagglutinin, pertactin (a 69 kDa outer-

9
A. D. T. P. (ACELL. COMPONENT), INACTIVATED POLIOMYLITIS VACCINE AND HAEMOPHILUS TYPE B CONJUGATE IP 2007

membrane protein) and other defined components of B. Reference vaccine(s)


pertussis such as fimbrial-2 and fimbrial-3 antigens. The latter Provided valid assays can be performed, monocomponent
2 antigens may be co-purified. The antigenic composition and reference vaccines may be used for the assays on the combined
characteristics are based on evidence of protection and vaccine. If this is not possible because of interaction between
freedom from unexpected reactions in the target group for the components of the combined vaccine or because of the
which the vaccine is intended. difference in composition between monocomponent reference
PRP is a linear copolymer composed of repeated units of vaccine and the test vaccine, a batch of combined vaccine
3-β-D-ribofuranosyl-(1→1)-ribitol-5-phosphate shown to be effective in clinical trials or a batch representative
[(C10H19O12P)n], with a defined molecular size and derived from thereof is used as a reference vaccine. For the preparation of
a suitable strain of Haemophilus influenzae type b. The carrier a representative batch, strict adherence to the production
protein, when conjugated to PRP, is capable of inducing a T- process used for the batch tested in clinical trials is necessary.
cell-dependent B-cell immune response to the polysaccharide. The reference vaccine may be stabilised by a method that has
been shown to have no effect on the assay procedure.
Production
Production of the components
General provisions
The production of the components complies with the
The production method shall have been shown to yield requirements of the monographs on Diphtheria Vaccine
consistently vaccines comparable with the vaccine of proven (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
clinical efficacy and safety in man. (Acellular Component, Adsorbed), Poliomyelitis Vaccine
(Inactivated) and Haemophilus Type b Conjugate Vaccine.
The content of bacterial endotoxins in bulk purified diphtheria
toxoid, tetanus toxoid, pertussis components, purified, FINAL BULK VACCINE
inactivated monovalent poliovirus harvests and bulk PRP
conjugate is determined to monitor the purification procedure The final bulk of the diphtheria, tetanus, pertussis and
and to limit the amount in the final vaccine. For each poliomyelitis components is prepared by adsorption,
component, the content of bacterial endotoxins is less than separately or together, of suitable quantities of bulk purified
the limit approved for the particular vaccine and, in any case, diphtheria toxoid, bulk purified tetanus toxoid and bulk purified
the contents are such that the final vaccine contains less than acellular pertussis components onto a mineral carrier such as
100 IU per single human dose. aluminium hydroxide or hydrated aluminium phosphate and
admixture of suitable quantities of purified, monovalent
The production method is validated to demonstrate that the
harvests of human polioviruses 1, 2 and 3 or a suitable quantity
product, if tested, would comply with the following test. Inject
of a trivalent pool of such monovalent harvests. Suitable
subcutaneously 5 times the single human dose stated on the
antimicrobial preservatives may be added.
label into each of 5 healthy guinea-pigs, each weighing
between 250 and 350 g, that have not previously been treated The final bulk of the haemophilus component is prepared by
with any material that will interfere with the test. If within 42 dilution of the bulk conjugate to the final concentration with a
days of the injection any of the animals shows signs of or dies suitable diluent. A stabiliser may be added.
from diphtheria, toxaemia or tetanus, the vaccine does not Only a final bulk vaccine that complies with the following
comply with the test. If more than 1 animal dies from non- requirements may be used in the preparation of the final lot.
specific causes, repeat the test once; if more than 1 animal
dies in the second test, the vaccine does not comply with the Bovine serum albumin. Determine on the poliomyelitis
test. components by a suitable immunochemical method (2.2.14)
during preparation of the final bulk vaccine, before addition
During development studies and wherever revalidation is of the adsorbent, the amount of bovine serum albumin is such
necessary, it shall be demonstrated by tests in animals that that the content in the final vaccine will not be more than 50
the vaccine induces a T-cell dependent B-cell immune response ng per single human dose.
to PRP.
Antimicrobial preservative. Where applicable, determine the
As part of consistency studies the assays of the diphtheria, amount of antimicrobial preservative by a suitable chemical
tetanus, pertussis and poliomyelitis components are carried method. The content is not less than 85.0 per cent and not
out on a suitable number of batches of vaccine reconstituted greater than 115.0 per cent of the intended amount.
for use. For subsequent routine control, the assays of these
components may be carried out without mixing with the Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
haemophilus component. for each sterility medium.

10
IP 2007 A. D. T. P. (ACELL. COMPONENT), INACTIVATED POLIOMYLITIS VACCINE AND HAEMOPHILUS TYPE B CONJUGATE

FINAL LOT immunochemical methods (2.2.14). The following method,


applicable to certain vaccines, is given as an example. The
The final bulk of the haemophilus component is freeze-dried.
clear supernatant obtained during identification test A reacts
Only a final lot that is satisfactory with respect to the test for with specific antisera to the pertussis components of the
osmolality shown below and with respect to each of the vaccine.
requirements given below under Identification, Tests and D. The vaccine is shown to contain human polioviruses 1, 2
Assay may be released for use. and 3 by a suitable immunochemical method (2.2.14), such as
Provided that the test for absence of residual pertussis toxin determination of D-antigen by enzyme-linked mmunosorbent
and irreversibility of pertussis toxoid, the test for antimicrobial assay (ELISA).
preservative and the assay have been carried out with
E. The haemophilus component is identified by a suitable
satisfactory results on the final bulk vaccine, they may be
immunochemical method (2.2.14) for PRP.
omitted on the final lot.
Provided that the free formaldehyde content has been Tests
determined on the bulk purified antigens and the purified
The tests for absence of residual pertussis toxin, irreversibility
monovalent harvests or the trivalent pool of polioviruses or
of pertussis toxoid, aluminium, free formaldehyde,
the final bulk and it has been shown that the content in the
antimicrobial preservative and sterility are carried out on
final lot will not exceed 0.2 g/l, the test for free formaldehyde
the container with the diphtheria, tetanus, pertussis and
may be omitted on the final lot.
poliomyelitis components; the tests for PRP content, water,
Provided that the in vivo assay for the poliomyelitis component sterility and pyrogens are carried out on the container with
has been carried out with satisfactory results on the final bulk the haemophilus component.
vaccine, it may be omitted on the final lot. Some tests for the haemophilus component may be carried
Osmolality (2.4.23). The osmolality of the vaccine, out on the freeze-dried product rather than on the bulk
reconstituted where applicable, is within the limits approved conjugate where the freeze-drying process may affect the
for the particular preparation. component under test.
Free PRP Absence of residual pertussis toxin and irreversibility
of pertussis toxoid
Unbound PRP is determined on the haemophilus component
after removal of the conjugate, for example by anion-exchange, This test is not necessary for the product obtained by genetic
size-exclusion or hydrophobic chromatography (2.4.16), modification. Use 3 groups each of not fewer than 5 histamine-
ultrafiltration or other validated methods. The amount of free sensitive mice. Inject intraperitoneally into each mouse of the
PRP is not greater than that approved for the particular product. first group twice the single human dose of the vaccine stored
at 2° to 8°. Inject intraperitoneally into each mouse of the
Identification second group twice the single human dose of the vaccine
Identification tests A, B, C and D are carried out using the vial incubated at 37° for 4 weeks. Inject diluent into the third group
containing the diphtheria, tetanus, pertussis and poliomyelitis of mice. After 5 days, inject into each mouse 2 mg of histamine
components; identification test E is carried out on the vial base intraperitoneally in a volume not exceeding 0.5 ml and
containing the haemophilus component. observe for 24 hours. The test is invalid if 1 or more control
mice die following histamine challenge. The vaccine complies
A. Diphtheria toxoid is identified by a suitable immunochemical
with the test if no animal in the first or second group dies
method (2.2.14). The following method, applicable to certain
following histamine challenge. If 1 mouse dies in either or
vaccines, is given as an example. Dissolve in the vaccine under
both of the first and second groups, the test may be repeated
examination sufficient sodium citrate to give a 10 per cent
with the same number of mice or with a greater number and the
w/v solution. Maintain at 37° for about 16 hours and centrifuge
results of valid tests combined; the vaccine complies with the
until a clear supernatant is obtained. The clear supernatant
test if, in both of the groups given the vaccine, not more than
reacts with a suitable diphtheria antitoxin, giving a precipitate.
5 per cent of the total number of mice die following histamine
B. Tetanus toxoid is identified by a suitable immunochemical challenge.
method (2.2.14). The following method, applicable to certain
The histamine sensitivity of the strain of mice used is verified
vaccines, is given as an example. The clear supernatant
at suitable intervals as follows: inject intravenously threefold
obtained during identification test A reacts with a suitable
dilutions of a reference pertussis toxin preparation in
tetanus antitoxin, giving a precipitate.
phosphate-buffered saline solution containing 0.2 per cent
C. The pertussis components are identified by suitable w/v of gelatin and challenge with histamine as above; the

11
A. D. T. P. (ACELLULAR. COMPONENT) AND INACTIVATED POLIOMYLITIS VACCINE IP 2007

strain is suitable if more than 50.0 per cent of the animals are immunochemical method (2.2.14) using a reference preparation
sensitised by 50 ng of pertussis toxin and none of the control calibrated in units of D-antigen. For each type, the content,
animals injected with only diluent and challenged similarly expressed with reference to the amount of D-antigen stated
with histamine show symptoms of sensitisation. on the label, is within the limits approved for the particular
PRP. Minimum 80.0 per cent of the amount of PRP stated on product. Poliomyelitis vaccine (inactivated) reference
the label. PRP is determined either by assay of ribose ( 2.7.1) preparation is calibrated in Units and intended for use in the
or phosphorus ( 2.7.1), by an immunochemical method ( 2.2.14) assay of D-antigen. The Unit and the International Unit are
or by anion-exchange liquid chromatography with pulsed- equivalent.
amperometric detection.
In vivo test
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
if aluminium hydroxide or hydrated aluminium phosphate is The vaccine complies with the in vivo assay as stated under
used as the adsorbent. Inactivated Poliomyelitis Vaccine.

Free formaldehyde (2.3.20). Maximum 0.2 g/l. Labelling. The label states (1) the minimum number of
International Units of diphtheria and tetanus toxoid per single
Antimicrobial preservative. Where applicable, determine the
human dose (2) the names and amounts of the pertussis
amount of antimicrobial preservative by a suitable chemical
components per single human dose; (3) the nominal amount
method. The content is not less than 85.0 per cent and not
of poliovirus of each type (1, 2 and 3), expressed in units of
greater than 115.0 per cent of the intended amount.
D-antigen per single human dose; (4) the type of cells used
Water (2.3.43). Maximum 3.0 per cent for the haemophilus for production of the poliomyelitis component; (5) the number
component. of micrograms of PRP per single human dose; (6) the type and
Sterility (2.2.11 ). Complies with the test for sterility. nominal amount of carrier protein per single human dose; (7)
where applicable, that the vaccine is intended for primary
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject vaccination of children and is not necessarily suitable for
per kg of the rabbit’s mass a quantity of the vaccine equivalent reinforcing doses or for administration to adults; (8) the name
to: 1 mg of PRP for a vaccine with diphtheria toxoid or CRM and the amount of the adsorbent; (9) that the vaccine must be
197 diphtheria protein as carrier; 0.1 mg of PRP for a vaccine shaken before use; (10) that the vaccine is not to be frozen;
with tetanus toxoid as carrier; 0.025 mg of PRP for a vaccine (11) where applicable, that the vaccine contains a pertussis
with OMP as a carrier. toxin-like protein produced by genetic modification.
Assay
Diphtheria component
Carry out one of the prescribed methods for the assay as Adsorbed Diphtheria, Tetanus,
stated under Diphtheria Vaccine (Adsorbed). Pertussis (Acellular Component) and
Unless otherwise justified and authorised, the lower Inactivated Poliomyelitis Vaccine
confidence limit (P = 0.95) of the estimated potency is not less
than 30 IU per single human dose. Diphtheria, Tetanus, Pertussis (Acellular Component) and
Poliomyelitis (Inactivated) Vaccine is a combined vaccine
Tetanus component
containing: diphtheria formol toxoid; tetanus formol toxoid;
Carry out one of the prescribed methods for the assay as individually purified antigenic components of Bordetella
stated under Tetanus Vaccine (Adsorbed). pertussis; suitable strains of human polioviruses 1, 2 and 3
The lower confidence limit (P = 0.95) of the estimated potency grown in suitable cell cultures and inactivated by a validated
is not less than 40 IU per single human dose. method; a mineral adsorbent such as aluminium hydroxide or
hydrated aluminium phosphate.
Pertussis component
The formol toxoids are prepared from the toxins produced by
It complies with the assay as stated under Adsorbed Pertussis the growth of Corynebacterium diphtheriae and Clostridium
Vaccine (Acellular Component). tetani respectively.
Poliomyelitis component The vaccine contains either pertussis toxoid or a pertussis-
toxin-like protein free from toxic properties produced by
D-antigen content expression of a genetically modified form of the corresponding
As a measure of consistency of production, determine the D- gene. Pertussis toxoid is prepared from pertussis toxin by a
antigen content for human polioviruses 1, 2 and 3 by a suitable method that renders the toxin harmless while maintaining

12
IP 2007 A. D. T. P. (ACELLULAR. COMPONENT) AND INACTIVATED POLIOMYLITIS VACCINE

adequate immunogenic properties and avoiding reversion to bulk purified diphtheria toxoid, tetanus toxoid, acellular
toxin. The vaccine may also contain filamentous pertussis components and admixture of suitable quantities of
haemagglutinin, pertactin (a 69 kDa outer-membrane protein) purified monovalent harvests of human polioviruses 1, 2 and
and other defined components of B. pertussis such as fimbrial- 3 or a suitable quantity of a trivalent pool of such purified
2 and fimbrial-3 antigens. The latter 2 antigens may be monovalent harvests. Suitable antimicrobial preservatives may
copurified. The antigenic composition and characteristics are be added.
based on evidence of protection and freedom from unexpected Only a final bulk vaccine that complies with the following
reactions in the target group for which the vaccine is intended. requirements may be used in the preparation of the final lot.
Production Bovine serum albumin. Determine on the poliomyelitis
General provisions components by a suitable immunochemical method (2.2.14)
after virus harvest and before addition of the adsorbent in the
The production method shall have been shown to yield preparation of the final bulk vaccine, the amount of bovine
consistently vaccines comparable with the vaccine of proven serum albumin is such that the content in the final vaccine will
clinical efficacy and safety in man. be not more than 50 ng per single human dose.
The production method is validated to demonstrate that the Antimicrobial preservative. Where applicable, determine the
product, if tested, would comply with the test for abnormal amount of antimicrobial preservative by a suitable chemical
toxicity for antisera and vaccines. method. The content is not less than 85.0 per cent and not
The content of bacterial endotoxins in bulk purified diphtheria greater than 115.0 per cent of the intended amount.
toxoid, tetanus toxoid, pertussis components and purified, Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
inactivated monovalent poliovirus harvests is determined to for each sterility medium.
monitor the purification procedure and to limit the amount in
the final vaccine. For each component, the content of bacterial FINAL LOT
endotoxins is less than the limit approved for the particular
vaccine and, in any case, the contents are such that the final Only a final lot that is satisfactory with respect to the test for
vaccine contains less than 100 IU per single human dose. osmolality and with respect to each of the requirements given
below under Identification, Tests and Assay may be released
Reference vaccine(s) for use.
Provided valid assays can be performed, monocomponent Provided the tests for absence of residual pertussis toxin,
reference vaccines may be used for the assays on the combined irreversibility of pertussis toxoid and antimicrobial preservative
vaccine. If this is not possible because of interaction between and the assays for the diphtheria, tetanus and pertussis
the components of the combined vaccine or because of the components have been carried out with satisfactory results
difference in composition between monocomponent reference on the final bulk vaccine, they may be omitted on the final lot.
vaccine and the test vaccine, a batch of combined vaccine
shown to be effective in clinical trials or a batch representative Provided the free formaldehyde content has been determined
thereof is used as a reference vaccine. For the preparation of on the bulk purified antigens or on the final bulk and it has
a representative batch, strict adherence to the production been shown that the content in the final lot will not exceed
process used for the batch tested in clinical trials is necessary. 0.2 g/l, the test for free formaldehyde may be omitted on the
The reference vaccine may be stabilised by a method that has final lot.
been shown to have no effect on the assay procedure. Provided that the determination of D-antigen content has been
carried out with satisfactory results during preparation of the
Production of the components final bulk before addition of the adsorbent, it may be omitted
The production of the components complies with the on the final lot.
requirements of the monographs on Diphtheria Vaccine Provided that the in vivo assay for the poliomyelitis component
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine has been carried out with satisfactory results on the final bulk
(Acellular Component, Adsorbed) and Poliomyelitis Vaccine vaccine, it may be omitted on the final lot.
(Inactivated).
Osmolality (2.4.23). The osmolality of the vaccine is within
FINAL BULK VACCINE the limits approved for the particular preparation.

The final bulk vaccine is prepared by adsorption onto a mineral Identification


carrier such as aluminium hydroxide or hydrated aluminium
phosphate, separately or together, of suitable quantities of A. Diphtheria toxoid is identified by a suitable immunochemical

13
A. D. T. P. (ACELLULAR. COMPONENT) AND INACTIVATED POLIOMYLITIS VACCINE IP 2007

method (2.2.14). The following method, applicable to certain sensitised by 50 ng of pertussis toxin and none of the control
vaccines, is given as an example. Dissolve in the vaccine under animals injected with only diluent and challenged similarly
examination sufficient sodium citrate to give a 10 per cent with histamine shows symptoms of sensitisation.
w/v solution. Maintain at 37° for about 16 h and centrifuge Aluminium (2.3.9). Maximum 1.25 mg per single human dose
until a clear supernatant is obtained. The clear supernatant if aluminium hydroxide or hydrated aluminium phosphate is
reacts with a suitable diphtheria antitoxin, giving a precipitate. used as the adsorbent.
B. Tetanus toxoid is identified by a suitable immunochemical
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
method (2.2.14). The following method, applicable to certain
vaccines, is given as an example. The clear supernatant Antimicrobial preservative. Where applicable, determine the
obtained as described in Identification test A reacts with a amount of antimicrobial preservative by a suitable chemical
suitable tetanus antitoxin, giving a precipitate. method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount.
C. The pertussis components are identified by a suitable
immunochemical method (2.2.14). The following method, Sterility (2.2.11). Complies with the test for sterility.
applicable to certain vaccines, is given as an example. The
Assay
clear supernatant obtained as described in Identification test
A reacts with a specific antisera to the pertussis components
Diphtheria component
of the vaccine.
Carry out one of the prescribed methods for the assay as
D. The vaccine is shown to contain human polioviruses 1, 2 stated under Diphtheria Vaccine (Adsorbed).
and 3 by a suitable immunochemical method (2.2.14) such as
the determination of D-antigen by enzyme-linked immuno- The lower confidence limit (P = 0.95) of the estimated potency
sorbent assay (ELISA). is not less than the minimum potency stated on the label.
Unless otherwise justified and authorised, the minimum
Tests potency stated on the label is 30 IU per single human dose.
Absence of residual pertussis toxin and irreversibility Tetanus component
of pertussis toxoid
Carry out one of the prescribed methods for the assay as
This test is not necessary for the product obtained by genetic stated under Tetanus Vaccine (Adsorbed).
modification. Use 3 groups each of not less than 5 histamine- The lower confidence limit (P = 0.95) of the estimated potency
sensitive mice. Inject intraperitoneally into each mouse of the is not less than 40 IU per single human dose.
first group twice the single human dose of the vaccine stored
at 2° to 8°. Inject intraperitoneally into each mouse of the Pertussis component
second group twice the single human dose of the vaccine
The vaccine complies with the assay as stated under Adsorbed
incubated at 37° for 4 weeks. Inject diluent into the third group
Pertussis Vaccine (Acellular Component).
of mice. After 5 days, inject into each mouse 2 mg of histamine
base intraperitoneally in a volume not exceeding 0.5 ml and Poliomyelitis component
observe for 24 hours. The test is invalid if 1 or more control
mice die following histamine challenge. The vaccine complies D-antigen content
with the test if no animal in the first or second group dies As a measure of consistency of production, determine the D-
following histamine challenge. If 1 mouse dies in either or antigen content for human polioviruses 1, 2 and 3 by a suitable
both of the first and second groups, the test may be repeated immunochemical method (2.2.14) following desorption using
with the same number of mice or with a greater number and the
a reference preparation calibrated in units of D-antigen. For
results of valid tests combined; the vaccine complies with the
each type, the content, expressed with reference to the amount
test if, in both of the groups given the vaccine, not more than
of D-antigen stated on the label, is within the limits approved
5.0 per cent of the total number of mice die following histamine
for the particular product. Poliomyelitis vaccine (inactivated)
challenge.
reference preparation is calibrated in Units and intended for
The histamine sensitivity of the strain of mice used is verified use in the assay of D-antigen. The Unit and the International
at suitable intervals as follows: inject intravenously three- Unit are equivalent.
fold dilutions of a reference pertussis toxin preparation in
In vivo test
phosphate-buffered saline solution containing 0.2 per cent
w/v of gelatin and challenge with histamine as above; the The vaccine complies with the in vivo assay as stated under
strain is suitable if more than 50.0 per cent of the animals are Inactivated Poliomyelitis Vaccine.

14
IP 2007 ADSORBED DIPHTHERIA TETANUS PERTUSSIS AND POLIOMYLITIS (INACTIVATED) VACCINE

Labelling. The label complies with the requirements stated specific causes, repeat the test once; if more than 1 animal
under Vaccine and also states (1) the minimum number of dies in the second test, the vaccine does not comply with the
International Units of diphtheria and tetanus toxoid per single test.
human dose (2) the names and amounts of the pertussis
components per single human dose; (3) the nominal amount Reference vaccine(s)
of poliovirus of each type (1, 2 and 3), expressed in units of D- Provided valid assays can be performed, monocomponent
antigen per single human dose; (4) the type of cells used for reference vaccines may be used for the assays on the combined
production of the poliomyelitis component; (5) the number of vaccine. If this is not possible because of interaction between
micrograms of PRP per single human dose; (6) the type and the components of the combined vaccine or because of the
nominal amount of carrier protein per single human dose; (7) difference in composition between monocomponent reference
where applicable, that the vaccine is intended for primary vaccine and the test vaccine, a batch of combined vaccine
vaccination of children and is not necessarily suitable for shown to be effective in clinical trials or a batch representative
reinforcing doses or for administration to adults; (8) the name thereof is used as a reference vaccine. For the preparation of
and the amount of the adsorbent; (9) that the vaccine must be a representative batch, strict adherence to the production
shaken before use; (10) that the vaccine is not to be frozen; process used for the batch tested in clinical trials is necessary.
(11) where applicable, that the vaccine contains a pertussis The reference vaccine may be stabilised by a method that has
toxin-like protein produced by genetic modification. been shown to have no effect on the assay procedure.

Production of the components


Adsorbed Diphtheria, Tetanus, The production of the components complies with the
Pertussis and Poliomyelitis requirements of the monographs on Diphtheria Vaccine
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
(Inactivated) Vaccine (Adsorbed) and Poliomyelitis Vaccine (Inactivated).
Diphtheria, Tetanus, Pertussis and Poliomyelitis (Inactivated)
Vaccine (Adsorbed) is a combined vaccine containing: FINAL BULK VACCINE
diphtheria formol toxoid; tetanus formol toxoid; an inactivated The final bulk vaccine is prepared by adsorption onto a mineral
suspension of Bordetella pertussis; suitable strains of human carrier such as aluminium hydroxide or hydrated aluminium
polioviruses 1, 2 and 3 grown in suitable cell cultures and phosphate, separately or together, of suitable quantities of
inactivated by a validated method; a mineral adsorbent such bulk purified diphtheria toxoid and bulk purified tetanus toxoid
as aluminium hydroxide or hydrated aluminium phosphate. and admixture of suitable quantities of an inactivated
The formol toxoids are prepared from the toxins produced by suspension of B. pertussis and purified monovalent harvests
the growth of Corynebacterium diphtheriae and Clostridium of human polioviruses 1, 2 and 3 or a suitable quantity of a
tetani respectively. trivalent pool of such purified monovalent harvests. Suitable
antimicrobial preservatives may be added.
Production
Only a final bulk vaccine that complies with the following
General provisions requirements may be used in the preparation of the final lot.

The production method shall have been shown to yield Specific toxicity
consistently the vaccines comparable with the vaccine of
Use not less than 5 healthy mice each weighing between
proven clinical efficacy and safety in man.
14 and 16 g for the vaccine group and for the saline control.
The production method is validated to demonstrate that the Use mice of the same sex or distribute males and females equally
product, if tested, would comply with the test for abnormal between the groups. Allow the animals access to food and
toxicity for antisera and vaccines, and with the following test water for at least 2 hours before injection and during the test.
for specific toxicity of the diphteria and tetanus components : Inject each mouse of the vaccine group intraperitoneally with
inject subcutaneously 5 times the single human dose stated 0.5 ml, containing a quantity of the vaccine equivalent to not
on the label into each of 5 healthy guinea-pigs, each weighing less than half the single human dose. Inject each mouse of the
between 250 and 350 g, that have not previously been treated control group with 0.5 ml of a 0.9 per cent sterile solution of
with any material that will interfere with the test. If within 42 sodium chloride, preferably containing the same amount of
days of the injection any of the animals shows signs of or dies antimicrobial preservative as that injected with the vaccine.
from diphtheria toxaemia or tetanus, the vaccine does not Weigh the groups of mice immediately before the injection
comply with the test. If more than 1 animal dies from non- and 72 hours and 7 days after the injection. The vaccine

15
ADSORBED DIPHTHERIA TETANUS PERTUSSIS AND POLIOMYLITIS (INACTIVATED) VACCINE IP 2007

complies with the test if: (a) at the end of 72 hours the total reacts with a suitable diphtheria antitoxin, giving a precipitate.
mass of the group of vaccinated mice is not less than that
B. Tetanus toxoid is identified by a suitable immunochemical
preceding the injection; (b) at the end of 7 days the average
method (2.2.14). The following method, applicable to certain
increase in mass per vaccinated mouse is not less than
vaccines, is given as an example. The clear s u p e r n a t a n t
60 per cent of that per control mouse; and (c) not more than
obtained during identification test A reacts with a suitable
5 per cent of the vaccinated mice die during the test. The test
tetanus antitoxin, giving a precipitate.
may be repeated and the results of the tests combined.
Bovine serum albumin. Determine on the poliomyelitis C. The centrifugation residue obtained in identification A may
components by a suitable immunochemical method (2.2.14) be used. Other suitable methods for separating the bacteria
during preparation of the final bulk vaccine; before addition from the adsorbent may also be used. Identify pertussis
of the adsorbent, the amount of bovine serum albumin is such vaccine by agglutination of the bacteria from the resuspended
that the content in the final vaccine will be not more than 50 precipitate by antisera specific to B. pertussis or by the assay
ng per single human dose. of the pertussis component prescribed under Assay.

Antimicrobial preservative. Where applicable, determine the D. The vaccine is shown to contain human polioviruses 1, 2
amount of antimicrobial preservative by a suitable chemical and 3 by a suitable immunochemical method (2.2.14) such as
method. The content is not less than 85.0 per cent and not the determination of D-antigen by enzyme-linked immuno-
greater than 115.0 per cent of the intended amount. sorbent assay (ELISA).

Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk Tests
for each sterility medium.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
FINAL LOT if aluminium hydroxide or hydrated aluminium phosphate is
Only a final lot that is satisfactory with respect to the test for used as the adsorbent.
osmolality and with respect to each of the requirements given Free formaldehyde (2.3.20). Maximum 0.2 g/l.
below under Identification, Tests and Assay may be released
for use. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Provided that the tests for specific toxicity and antimicrobial method. The content is not less than 85.0 per cent and not
preservative, and the assays for the diphtheria, tetanus and greater than 115.0 per cent of the intended amount.
pertussis components have been carried out with satisfactory
results on the final bulk vaccine, they may be omitted on the Sterility (2.2.11). Complies with the test for sterility.
final lot. Assay
Provided that the free formaldehyde content has been
determined on the bulk purified antigens, the inactivated B. Diphtheria component
pertussis suspension and the purified monovalent harvests Carry out one of the prescribed methods for the assay as
or the trivalent pool of polioviruses or on the final bulk and it stated under Diphtheria Vaccine (Adsorbed).
has been shown that the content in the final lot will not exceed
0.2 g/l, the test for free formaldehyde may be omitted on the The lower confidence limit (P = 0.95) of the estimated potency
final lot. is not less than 30 IU per single human dose.

Provided that the in vivo assay for the poliomyelitis component Tetanus component
has been carried out with satisfactory results on the final bulk Carry out one of the prescribed methods for the assay as
vaccine, it may be omitted on the final lot. stated under Tetanus Vaccine (Adsorbed).
Osmolality (2.4.23). The osmolality of the vaccine is within If the test is carried out in guinea pigs, the lower confidence
the limits approved for the particular preparation. limit (P = 0.95) of the estimated potency is not less than 40 IU
Identification per single human dose; if the test is carried out in mice, the
lower confidence limit (P = 0.95) of the estimated potency is
A. Diphtheria toxoid is identified by a suitable immunochemical not less than 60 IU per single human dose.
method (2.2.14). The following method, applicable to certain
vaccines, is given as an example. Dissolve in the vaccine under Pertussis component
examination sufficient sodium citrate to give a 10 per cent
Carry out the assay as stated under Pertussis Vaccine.
w/v solution. Maintain at 37° for about 16 hours and centrifuge
until a clear supernatant is obtained. The clear supernatant The estimated potency is not less than 4 IU per single human

16
IP 2007 A. D., T., P., POLIOMYLITIS (INACTIVATED) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE

dose and the lower confidence limit (P = 0.95) of the estimated The formol toxoids are prepared from the toxins produced by
potency is not less than 2 IU per single human dose. the growth of Corynebacterium diphtheriae and Clostridium
tetani respectively.
Poliomyelitis component
PRP is a linear copolymer composed of repeated units of
D-antigen content 3-β-D-ribofuranosyl-(1→1)-ribitol-5-phosphate
[(C10H19O12P)n], with a defined molecular size and derived from
As a measure of consistency of production, determine the D- a suitable strain of Haemophilus influenzae type b. The carrier
antigen content for human polioviruses 1, 2 and 3 by a suitable protein, when conjugated to PRP, is capable of inducing a T-
immunochemical method (2.2.14) using a reference preparation cell-dependent B-cell immune response to the polysaccharide.
calibrated in Units of D-antigen. For each type, the content,
expressed with reference to the amount of D-antigen stated Production
on the label, is within the limits approved for the particular
product. Poliomyelitis vaccine (inactivated) reference General provisions
preparation is calibrated in Units and is intended for use in The production method shall have been shown to yield
the assay of D-antigen. The Unit and the IU are equivalent. consistently vaccines comparable with the vaccine of proven
clinical efficacy and safety in man.
In vivo test
The production method is validated to demonstrate that the
The vaccine complies with the in vivo assay as stated under product, if tested, would comply with the test for abnormal
Poliomyelitis Vaccine (Inactivated). toxicity for antisera and vaccines, and with the following test
Labelling. The label states (1) the minimum number of for specific toxicity of the diphtheria and tetanus components:
International Units of diphtheria and tetanus toxoid per single inject subcutaneously 5 times the single human dose stated
human dose; (2) the minimum number of International Units on the label into each of 5 healthy guinea-pigs, each weighing
of pertussis vaccine per single human dose; (3) the nominal between 250 and 350 g, that have not previously been treated
amount of poliovirus of each type (1, 2 and 3), expressed in with any material that will interfere with the test. If within 42
units of D-antigen per single human dose; (4) the type of cells days of the injection any of the animals shows signs of or dies
used for production of the poliomyelitis component; (5) where from diphtheria toxaemia or tetanus, the vaccine does not
applicable, that the vaccine is intended for primary vaccination comply with the test. If more than 1 animal dies from non-
of children and is not necessarily suitable for reinforcing doses specific causes, repeat the test once; if more than 1 animal
or for administration to adults; (6) the name and the amount of dies in the second test, the vaccine does not comply with the
the adsorbent; (7) that the vaccine must be shaken before test.
use; (8) that the vaccine is not to be frozen. During development studies and wherever revalidation is
necessary, it shall be demonstrated by tests in animals that
the vaccine induces a T-cell dependent B-cell immune response
to PRP.
Adsorbed Diphtheria, Tetanus, As part of consistency studies the assays of the diphtheria,
tetanus, pertussis and poliomyelitis components are carried
Pertussis, Poliomyelitis (Inactivated) out on a suitable number of batches of vaccine reconstituted
and Haemophilus Type b Conjugate for use. For subsequent routine control, the assays of these
Vaccine components may be carried out without mixing with the
haemophilus component.
Diphtheria, Tetanus, Pertussis, Poliomyelitis (Inactivated) and
Reference vaccine(s)
Haemophilus Type b Conjugate Vaccine (Adsorbed) is a
combined vaccine composed of: diphtheria formol toxoid; Provided valid assays can be performed, monocomponent
tetanus formol toxoid; an inactivated suspension of Bordetella reference vaccines may be used for the assays on the combined
pertussis; suitable strains of human polioviruses 1, 2 and 3 vaccine. If this is not possible because of interaction between
grown in suitable cell cultures and inactivated by a suitable the components of the combined vaccine or because of the
method; polyribosylribitol phosphate (PRP) covalently bound difference in composition between monocomponent reference
to a carrier protein; a mineral adsorbent such as aluminium vaccine and the test vaccine, a batch of combined vaccine
hydroxide or hydrated aluminium phosphate. The product is shown to be effective in clinical trials or a batch representative
presented with the haemophilus component in a separate thereof is used as a reference vaccine. For the preparation of
container, the contents of which are mixed with the other a representative batch, strict adherence to the production
components immediately before use. process used for the batch tested in clinical trials is necessary.

17
A. D., T., P., POLIOMYLITIS (INACTIVATED) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE IP 2007

The reference vaccine may be stabilised by a method that has of the adsorbent, the amount of bovine serum albumin is such
been shown to have no effect on the assay procedure. that the content in the final vaccine will not be more than 50
ng per single human dose.
Production of components
Antimicrobial preservative. Where applicable, determine the
The production of the components complies with the amount of antimicrobial preservative by a suitable chemical
requirements of the monographs on Diphtheria Vaccine method. The content is not less than 85.0 per cent and not
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine greater than 115.0 per cent of the intended amount.
(Adsorbed), Poliomyelitis Vaccine (Inactivated) and
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
Haemophilus Type b Conjugate Vaccine.
for each sterility medium.
FINAL BULK VACCINE
FINAL LOT
The final bulk of the diphtheria, tetanus, pertussis and
The final bulk of the haemophilus component is freeze-dried.
poliomyelitis components is prepared by adsorption,
separately or together, of suitable quantities of bulk purified Only a final lot that is satisfactory with respect to the test for
diphtheria toxoid, and bulk purified tetanus toxoid onto a osmolality shown below and with respect to each of the
mineral carrier such as aluminium hydroxide or hydrated requirements given below under Identification, Tests and
aluminium phosphate and admixture of suitable quantities of Assay may be released for use.
an inactivated suspension of B. pertussis and of purified, Provided that the tests for specific toxicity and antimicrobial
monovalent harvests of human polioviruses 1, 2 and 3 or a preservative, and the assays for the diphtheria, tetanus and
suitable quantity of a trivalent pool of such monovalent pertussis components have been carried out with satisfactory
harvests. Suitable antimicrobial preservatives may be added. results on the final bulk vaccine, they may be omitted on the
The final bulk of the haemophilus component is prepared by final lot.
dilution of the bulk conjugate to the final concentration with a Provided that the free formaldehyde content has been
suitable diluent. A stabiliser may be added. determined on the bulk purified antigens, the inactivated B.
Only final bulk that complies with the following requirements pertussis suspension and the purified monovalent harvests
may be used in the preparation of the final lot. or the trivalent pool of polioviruses or on the final bulk and it
has been shown that the content in the final lot will not exceed
Specific toxicity 0.2 g/l, the test for free formaldehyde may be omitted on the
Use not less than 5 healthy mice each weighing between 14 final lot.
and 16 g, for the vaccine group and for the saline control. Use Provided that the in vivo assay for the poliomyelitis component
mice of the same sex or distribute males and females equally has been carried out with satisfactory results on the final bulk
between the groups. Allow the animals access to food and vaccine, it may be omitted on the final lot.
water for at least 2 hours before injection and during the test.
Osmolality (2.4.23). The osmolality of the vaccine,
Inject each mouse of the vaccine group intraperitoneally with
reconstituted where applicable, is within the limits approved
0.5 ml, containing a quantity of the vaccine equivalent to not
for the particular preparation.
less than half the single human dose. Inject each mouse of the
control group with 0.5 ml of a 0.9 per cent sterile solution of Free PRP
sodium chloride, preferably containing the same amount of
antimicrobial preservative as that injected with the vaccine. Unbound PRP is determined on the haemophilus component
Weigh the groups of mice immediately before the injection after removal of the conjugate, for example by anion-exchange,
and 72 hours and 7 days after the injection. The vaccine size-exclusion or hydrophobic chromatography (2.4.16),
complies with the test if (a) at the end of 72 hours the total ultrafiltration or other validated methods. The amount of free
mass of the group of vaccinated mice is not less than that PRP is not greater than that approved for the particular product.
preceding the injection; (b) at the end of 7 days the average
Identification
increase in mass per vaccinated mouse is not less than
60 per cent of that per control mouse; and (c) not more than Identification tests A, B, C and D are carried out using the
5 per cent of the vaccinated mice die during the test. The test vial containing the diphtheria, tetanus, pertussis and
may be repeated and the results of the tests combined. poliomyelitis components; identification test E is carried
out on the vial containing the haemophilus component.
Bovine serum albumin. Determine on the poliomyelitis
components by a suitable immunochemical method (2.2.14) A. Diphtheria toxoid is identified by a suitable immunochemical
during preparation of the final bulk vaccine, before addition method (2.2.14). The following method, applicable to certain

18
IP 2007 A. D., T., P., POLIOMYLITIS (INACTIVATED) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE

vaccines, is given as an example. Dissolve in the vaccine under Sterility (2.2.11). Complies with the test for sterility.
examination sufficient sodium citrate to give a 10 per cent Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
w/v solution. Maintain at 37° for about 16 hours and centrifuge per kg of the rabbit’s mass a quantity of the vaccine equivalent
until a clear supernatant is obtained. The clear supernatant to 1 mg of PRP for a vaccine with diphtheria toxoid or CRM
reacts with a suitable diphtheria antitoxin, giving a precipitate. 197 diphtheria protein as carrier; 0.1 mg of PRP for a vaccine
B. Tetanus toxoid is identified by a suitable immunochemical with tetanus toxoid as carrier; 0.025 mg of PRP for a vaccine
method (2.2.14). The following method, applicable to certain with OMP as carrier.
vaccines, is given as an example. The clear s u p e r n a t a n t
Assay
obtained during identification test A reacts with a suitable
tetanus antitoxin, giving a precipitate. Diphtheria component
C. The centrifugation residue obtained in identification A may Carry out one of the prescribed methods for the assay as
be used. Other suitable methods for separating the bacteria stated under Diphtheria Vaccine (Adsorbed).
from the adsorbent may also be used. Identify pertussis The lower confidence limit (P = 0.95) of the estimated potency
vaccine by agglutination of the bacteria from the resuspended is not less than 30 IU per single human dose.
precipitate by antisera specific to B. pertussis or by the assay
of the pertussis component prescribed under Assay. Tetanus component
D. The vaccine is shown to contain human polioviruses 1, 2 Carry out one of the prescribed methods for the assay as
and 3 by a suitable immunochemical method (2.2.14), such as stated under Tetanus Vaccine (Adsorbed).
determination of D-antigen by enzymelinked immunosorbent If the test is carried out in guinea-pigs, the lower confidence
assay (ELISA). limit (P = 0.95) of the estimated potency is not less than 40 IU
E. The haemophilus component is identified by a suitable per single human dose; if the test is carried out in mice, the
immunochemical method (2.2.14) for PRP. lower confidence limit (P = 0.95) of the estimated potency is
not less than 60 IU per single human dose.
Tests
Pertussis component
The tests for specific toxicity, aluminium, free formaldehyde,
antimicrobial preservative and sterility are carried out on Carry out the assay as stated under Pertussis Vaccine.
the container with diphtheria, tetanus, pertussis and The estimated potency is not less than 4 IU per single human
poliomyelitis components; the tests for PRP content, water, dose and the lower confidence limit (P = 0.95) of the estimated
sterility and pyrogens are carried out on the container with potency is not less than 2 IU per single human dose.
the haemophilus component.
Poliomyelitis component
Some tests for the haemophilus component may be carried
out on the freeze-dried product rather than on the bulk D-antigen content
conjugate where the freeze-drying process may affect the
component under test. As a measure of consistency of production, determine the D-
antigen content for human polioviruses 1, 2 and 3 by a suitable
PRP. Minimum 80.0 per cent of the amount of PRP stated on immunochemical method (2.2.14) using a reference preparation
the label. PRP is determined either by assay of ribose (2.7.1) or calibrated in Units of D-antigen. For each type, the content,
phosphorus (2.7.1), by an immunochemical method (2.2.14) or expressed with reference to the amount of D-antigen stated
by anion-exchange liquid chromatography with pulsed- on the label, is within the limits approved for the particular
amperometric detection. product. Poliomyelitis vaccine (inactivated) reference
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, preparation is calibrated in Units and intended for use in the
if aluminium hydroxide or hydrated aluminium phosphate is assay of D-antigen. The Unit and the IU are equivalent.
used as the adsorbent. In vivo test
Free formaldehyde (2.3.20). Maximum 0.2 g/l. The vaccine complies with the in vivo assay as stated under
Antimicrobial preservative. Where applicable, determine the Poliomyelitis Vaccine (Inactivated).
amount of antimicrobial preservative by a suitable chemical Labelling. The label states (1) the minimum number of
method. The content is not less than 85.0 per cent and not International Units of diphtheria and tetanus toxoid per single
greater than 115.0 per cent of the intended amount. human dose; (2) the minimum number of International Units
Water (2.3.43). Maximum 3.0 per cent for the haemophilus of pertussis vaccine per single human dose; (3) the nominal
component. amount of poliovirus of each type (1, 2 and 3), expressed in

19
ADSORBED PERTUSSIS VACCINE (ACELLULAR COMPONENT) IP 2007

Units of D-antigen per single human dose; (4) the type of cells individual components that comply with the following
used for production of the poliomyelitis component; (5) the requirements; after demonstration of consistency, the tests
number of micrograms of PRP per single human dose; (6) the need not be applied routinely to each batch.
type and nominal amount of carrier protein per single human Adenylate cyclase. Not more than 500 ng in the equivalent of
dose; (7) where applicable, that the vaccine is intended for 1 dose of the final vaccine, determined by immunoblot analysis
primary vaccination of children and is not necessarily suitable or another suitable method.
for reinforcing doses or for administration to adults; (8) the
name and the amount of the adsorbent; (9) that the vaccine Tracheal cytotoxin. Not more than 2 pmol in the equivalent of
must be shaken before use; (9) that the vaccine is not to be 1 dose of the final vaccine, determined by a suitable method
frozen. such as a biological assay or liquid chromatography (2.4.14).
Absence of residual dermonecrotic toxin. Inject intradermally
into each of 3 unweaned mice, in a volume of 0.1 ml, the
Adsorbed Pertussis Vaccine (Acellular amount of component or antigenic fraction equivalent to 1
Component) dose of the final vaccine. Observe for 48 hours. No
dermonecrotic reaction is demonstrable.
Pertussis Vaccine (Acellular Component, Adsorbed) is a
Specific properties. The components of the vaccine are
preparation of individually prepared and purified antigenic
analysed by one or more of the methods shown below in
components of Bordetella pertussis adsorbed on a mineral
order to determine their identity and specific properties (activity
carrier such as aluminium hydroxide or hydrated aluminium
per unit amount of protein) in comparison with reference
phosphate.
preparations.
The vaccine contains either pertussis toxoid or a pertussis
toxin, like protein free from toxic properties, produced by Pertussis toxin
expression of a genetically modified form of the corresponding Chinese hamster ovary (CHO) cell-clustering effect and
gene. Pertussis toxoid is prepared from pertussis toxin by a haemagglutination as in vitro methods; lymphocytosis-
method that renders the latter harmless while maintaining promoting activity, histamine-sensitising activity and insulin
adequate immunogenic properties and avoiding reversion to secretory activity as in vivo methods. The toxin shows ADP-
toxin. The vaccine may also contain filamentous ribosyl transferase activity using transducin as the acceptor.
haemagglutinin, pertactin (a 69 kDa outer-membrane protein)
and other defined components of B. pertussis such as fimbrial- Filamentous haemagglutinin
2 and fimbrial-3 antigens. The latter 2 antigens may be Haemagglutination and inhibition by specific antibody.
copurified. The antigenic composition and characteristics are Pertactin, fimbrial-2 and fimbrial-3 antigens. Reactivity with
based on evidence of protection and freedom from unexpected specific antibody.
reactions in the target group for which the vaccine is intended.
Pertussis toxoid
Production
The toxoid induces in animals production of antibodies
General provisions capable of inhibiting all the properties of pertussis toxin.
The production method shall have been shown to yield
PURIFIED COMPONENTS
consistently the vaccines comparable with the vaccine of
proven clinical efficacy and safety in man. Production of each component is based on a seed-lot system.
The seed cultures from which toxin is prepared are managed
Reference vaccine
to conserve or where necessary restore toxinogenicity by
A batch of vaccine shown to be effective in clinical trials or a deliberate selection.
batch representative thereof is used as a reference vaccine. None of the media used at any stage contains blood or blood
For the preparation of a representative batch, strict adherence products of human origin. Media used for the preparation of
to the production process used for the batch tested in clinical seed lots and inocula may contain blood or blood products of
trials is necessary. The reference vaccine is preferably animal origin.
stabilised by a method that has been shown to have no
significant effect on the assay procedure when the stabilised Pertussis toxin and, where applicable, filamentous
and non-stabilised batches are compared. haemagglutinin and pertactin are purified and, after appropriate
characterisation, detoxified using suitable chemical reagents,
CHARACTERISATION OF COMPONENTS by a method that avoids reversion of the toxoid to toxin,
During development of the vaccine, the production process particularly on storage or exposure to heat. Other components
shall be validated to demonstrate that it yields consistently such as fimbrial-2 and fimbrial-3 antigens are purified either

20
IP 2007 ADSORBED PERTUSSIS VACCINE (ACELLULAR COMPONENT)

separately or together, characterised and shown to be free validation of the process has demonstrated acceptable
from toxic substances. The purification procedure is validated clearance.
to demonstrate appropriate clearance of substances used Antigen content
during culture or purification.
Determine the antigen content by a suitable immunochemical
The content of bacterial endotoxins is determined to monitor
method (2.2.14) and protein nitrogen by sulphuric acid
the purification procedure and to limit the amount in the final
digestion (2.2.30) or another suitable method. The ratio of
vaccine. The limits applied for the individual components are
antigen content to protein nitrogen is within the limits
such that the final vaccine contains less than 100 IU per single
established for the product.
human dose.
Before detoxification, the purity of the components is FINAL BULK VACCINE
determined by a suitable method such as polyacrylamide gel
electrophoresis (PAGE) or liquid chromatography. SDS-PAGE The vaccine is prepared by adsorption of suitable quantities
or immunoblot analysis with specific monoclonal or polyclonal of purified components, separately or together, onto aluminium
antibodies may be used to characterise subunits. Requirements hydroxide or hydrated aluminium phosphate. A suitable
are established for each individual product. antimicrobial preservative may be added.

Only purified components that comply with the following Only a final bulk vaccine that complies with the following
requirements may be used in the preparation of the final bulk requirements may be used in the preparation of the final lot.
vaccine. Antimicrobial preservative. Where applicable, determine the
Sterility (2.2.11). Carry out the test for sterility using for each amount of antimicrobial preservative by a suitable chemical
medium a quantity of purified component equivalent to not method. The content is not less than 85.0 per cent and not
less than 100 doses. greater than 115.0 per cent of the intended amount.
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
Absence of residual pertussis toxin for each sterility medium.
This test is not necessary for the product obtained by genetic
FINAL LOT
modification. Use a group of not fewer than 5 histamine-
sensitive mice each weighing between 18 and 26 g. Inject into Only a final lot that is satisfactory with respect to each of the
each mouse the equivalent of 1 human dose intravenously or requirements given below under Identification, Tests and
twice the human dose intraperitoneally, diluted to not more Assay may be released for use. Provided that the tests for
than 0.5 ml with phosphate-buffered saline solution containing absence of residual pertussis toxin and irreversibility of
0.2 per cent w/v of gelatin. Inject diluent into a second group pertussis toxoid, antimicrobial preservative, free formaldehyde
of control mice. After 5 days, inject 2 mg of histamine base and the assay have been carried out with satisfactory results
intraperitoneally in a volume not exceeding 0.5 ml and observe on the final bulk vaccine, these tests may be omitted on the
for 24 hours. If no animal dies, the preparation complies with final lot.
the test.
Identification
The histamine sensitivity of the strain of mice used is verified
at suitable intervals as follows: inject three-fold dilutions of a Subject the vaccine to a suitable desorption procedure such
reference pertussis toxin preparation in phosphate-buffered as the following: dissolve in the vaccine under examination
saline solution containing 0.2 per cent w/v of gelatin and sufficient sodium citrate to give a 10 per cent w/v solution;
challenge with histamine as above; the strain is suitable if maintain at 37° for about 16 h and centrifuge until a clear
more than 50 per cent of the animals are sensitised by 50 ng of supernatant liquid is obtained. Examined by a suitable
pertussis toxin and none of the control animals injected with immunochemical method (2.2.14), the clear supernatant liquid
only diluent and challenged similarly with histamine show reacts with specific antisera to the components stated on the
symptoms of sensitisation. label.
A validated test based on the clustering effect of the toxin for Tests
Chinese hamster ovary (CHO) cells may be used instead of
the test on mice. Absence of residual pertussis toxin and irreversibility of
pertussis toxoid
Residual detoxifying agents and other reagents
This test is not necessary for the product obtained by genetic
The content of residual detoxifying agents and other reagents modification. Use 3 groups each of not fewer than 5 histamine-
is determined and shown to be below approved limits unless sensitive mice. Inject intraperitoneally into the first group twice

21
IP 2007

the single human dose of the vaccine stored at 2° to 8°. Inject Selection and distribution of test animals
intraperitoneally into the second group twice the single human Use in the test healthy mice (for example, CD1 strain) of the
dose of the vaccine incubated at 37° for 4 weeks. Inject diluent same stock 4 to 8 weeks old. Distribute the animals in 6 groups
into the third group of mice. After 5 days, inject into each of a number appropriate to the requirements of the assay. Use
mouse 2 mg of histamine base intraperitoneally in a volume 3 dilutions of the vaccine under examination and 3 dilutions of
not exceeding 0.5 ml and observe for 24 hours. The test is a reference preparation and attribute each dilution to a group
invalid if 1 or more control mice die following histamine of mice. Inject intraperitoneally or subcutaneously into each
challenge. The vaccine complies with the test if no animal in mouse 0.5 ml of the dilution attributed to its group.
the first or second group dies following histamine challenge.
If 1 mouse dies in either or both of the first and second groups, Collection of serum samples
the test may be repeated with the same number of mice or with 4 to 5 weeks after vaccination, bleed the mice individually
a greater number and the results of valid tests combined; the under anaesthesia. Store the sera at -20° until tested for
vaccine complies with the test if, in both of the groups given antibody content.
the vaccine, not more than 5.0 per cent of the total number of
Antibody determination
mice die following histamine challenge.
The histamine sensitivity of the strain of mice used is verified Assay the individual sera for content of specific antibodies to
at suitable intervals as follows: inject intravenously threefold each component using a validated method such as the ELISA
dilutions of a reference pertussis toxin preparation in test shown below.
phosphate-buffered saline solution containing 0.2 per cent ELISA
w/v of gelatin and challenge with histamine as above; the
Microtitre plates (poly(vinyl chloride) or polystyrene as
strain is suitable if more than 50.0 per cent of the animals are
appropriate for the specific antigen) are coated with the purified
sensitised by 50 ng of pertussis toxin and none of the control
antigen at a concentration of 100 ng per well. After washing,
animals injected with only diluent and challenged similarly
unreacted sites are blocked by incubating with a solution of
with histamine show symptoms of sensitisation.
bovine serum albumin and then washed. Two-fold dilutions
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, of sera from mice immunised with test or reference vaccines
if aluminium hydroxide or hydrated aluminium phosphate is are made on the plates. After incubation at 22° to 25° for 1 h,
used as the adsorbent. the plates are washed. A suitable solution of anti-mouse IgG
enzyme conjugate is added to each well and incubated at 22°
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
to 25° for 1 h. After washing, a substrate is added from which
Antimicrobial preservative. Where applicable, determine the the bound enzyme conjugate liberates a chromophore which
amount of antimicrobial preservative by a suitable chemical can be quantified by measurement of absorbance. The test
method. The content is not less than 85.0 per cent and not conditions are designed to obtain a linear response for
greater than 115.0 per cent of the intended amount. absorbance with respect to antibody content over the range
of measurement used and absorbance values within the range
Sterility (2.2.11). Complies with the test for sterility.
0.1 to 2.0.
Assay A reference antiserum of assigned potency is used in the test
and serves as the basis for calculation of the antibody levels
The capacity of the vaccine to induce the formation of specific in test sera. A standardised control serum is also included in
antibodies is compared with the same capacity of a reference the test.
preparation examined in parallel; antibodies are determined
using suitable immunochemical methods (2.2.14) such as The test is not valid if (a) the value found for the control
enzyme-linked immunosorbent assay (ELISA). The test on serum differs by more than 2 standard deviations from the
mice shown below uses a three-point model but, after assigned value; (b) the confidence interval of the potency
validation, for routine testing a single-dilution method may be estimate is greater than 50.0 per cent to 200.0 per cent.
used. Calculation
The antibody titres in the sera of mice immunised with
Requirement
reference and test vaccines are calculated and from the values
The capacity to induce antibodies is not significantly (P = obtained the potency of the test vaccine in relation to the
0.95) less than that of the reference vaccine. reference vaccine is calculated by the usual statistical methods.
The following test model is given as an example of a method Labelling. The label states (1) the names and amounts of the
that has been found to be satisfactory. components present in the vaccine; (2) where applicable, that

22
IP 2007 ADSORBED PERTUSSIS VACCINE (ACELLULAR CO-PURIFIED)

the vaccine contains a pertussis toxin-like protein produced Tracheal cytotoxin. Not more than 2 pmol in the equivalent of
by genetic modification; (3) the name and amount of the 1 dose of the final vaccine, determined by a suitable method
adsorbent; (4) that the vaccine must be shaken before use; (5) such as a biological assay or liquid chromatography (2.4.14).
that the vaccine is not to be frozen. Absence of residual dermonecrotic toxin. Inject intradermally
into each of 3 unweaned mice, in a volume of 0.1 ml, the amount
of antigenic fraction equivalent to 1 dose of the final vaccine.
Adsorbed Pertussis Vaccine (Acellular, Observe for 48 hours. No dermonecrotic reaction is
Co-Purified) demonstrable.
Specific properties. The antigenic fraction is analyzed by one
Pertussis Vaccine (Acellular, Co-Purified, Adsorbed) is a
or more of the methods shown below in order to determine the
preparation of antigenic components of Bordetella pertussis
identity and specific properties (activity per unit amount of
adsorbed on a mineral carrier such as aluminium hydroxide or
protein) of its components in comparison with reference
hydrated aluminium phosphate.
preparations.
The vaccine contains an antigenic fraction purified without
Pertussis toxin
separation of the individual components. The antigenic fraction
is treated by a method that transforms pertussis toxin to toxoid, Chinese hamster ovary (CHO) cell-clustering effect and
rendering it harmless while maintaining adequate immunogenic haemagglutination as in vitro methods; lymphocytosis-
properties of all the components and avoiding reversion to promoting activity, histamine-sensitising activity and insulin
toxin. The antigenic fraction is composed of pertussis toxoid, secretory activity as in vivo methods. The toxin shows
filamentous haemagglutinin, pertactin (a 69 kDa outer- ADP-ribosyl transferase activity using transducin as the
membrane protein) and other defined components of B. acceptor.
pertussis such as fimbrial-2 and fimbrial-3 antigens. It may Filamentous haemagglutinin
contain residual pertussis toxin up to a maximum level
approved by the competent authority. The antigenic Haemagglutination and inhibition by specific antibody.
composition and characteristics are based on evidence of Pertactin, fimbrial-2 and fimbrial-3 antigens. Reactivity with
protection and freedom from unexpected reactions in the target specific antibody.
group for which the vaccine is intended.
Pertussis toxoid
Production The toxoid induces in animals the production of antibodies
capable of inhibiting all the properties of pertussis toxin.
General provisions. The production method shall have been
shown to yield consistently vaccines comparable with the PURIFIED ANTIGENIC FRACTION
vaccine of proven clinical efficacy and safety in man.
Production of the antigenic fraction is based on a seed-lot
Reference vaccine. A batch of vaccine shown to be effective system. The seed cultures are managed to conserve or, where
in clinical trials or a batch representative thereof is used as a necessary, restore toxinogenicity by deliberate selection.
reference vaccine. For the preparation of a representative
None of the media used at any stage contains blood or blood
batch, strict adherence to the production process used for the
products of human origin. Media used for the preparation of
batch tested in clinical trials is necessary. The reference vaccine
seed batches and inocula may contain blood or blood products
is preferably stabilised, by a method that has been shown to
of animal origin.
have no significant effect on the assay procedure when the
stabilised and non-stabilised batches are compared. The antigenic fraction is purified and, after appropriate
characterisation, detoxified using suitable reagents by a
CHARACTERISATION OF COMPONENTS method that ensures minimal reversion of toxoid to toxin,
particularly on or exposure to heat. The purification procedure
During development of the vaccine, the production process
is validated to demonstrate appropriate clearance of
shall be validated to demonstrate that it yields consistently
substances used during culture or purification.
an antigenic fraction that complies with the following
requirements; after demonstration of consistency, the tests The content of bacterial endotoxins is determined to monitor
need not be applied routinely to each batch. the purification procedure and to limit the amount in the final
vaccine. The limits applied are such that the final vaccine
Adenylate cyclase. Not more than 500 ng in the equivalent of
contains not more than 100 IU per single human dose.
1 dose of the final vaccine, determined by immunoblot analysis
or another suitable method. Before detoxification, the purity of the antigenic fraction is

23
ADSORBED PERTUSSIS VACCINE (ACELLULAR CO-PURIFIED) IP 2007

determined by a suitable method such as polyacrylamide gel FINAL BULK VACCINE


electrophoresis (PAGE) (2.4.12) or liquid chromatography The vaccine is prepared by adsorption of a suitable quantity
(2.4.14). SDS-PAGE or immunoblot analysis with specific of the antigenic fraction onto aluminium hydroxide or hydrated
monoclonal or polyclonal antibodies may be used to aluminium phosphate. A suitable antimicrobial preservative
characterise subunits. Requirements are established for each may be added.
individual product.
Only a final bulk vaccine that complies with the following
Only a purified antigenic fraction that complies with the
requirements may be used in the preparation of the final lot.
following requirements may be used in the preparation of the
final bulk vaccine. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Sterility (2.2.11). Carry out the test for sterility using for each
method. The content is not less than 85.0 per cent and not
medium a quantity of purified antigenic fraction equivalent to
greater than 115.0 per cent of the intended amount.
not less than 100 doses of the final vaccine.
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
Test for residual pertussis toxin. Use 3 groups of not fewer for each sterility medium.
than 5 histamine-sensitive mice each weighing between 18
and 26 g. Using phosphate-buffered saline containing 0.2 per FINAL LOT
cent of gelatin, prepare a series of dilutions of the purified
Only a final lot that is satisfactory with respect to each of the
antigenic fraction that have been shown to yield a graded
requirements given below under Identification, Tests and
response and attribute each dilution to a separate group of
Assay may be released for use.
mice. Inject intraperitoneally into each mouse the dilution
attributed to its group. Inject diluent into a fourth group of Provided that the tests for residual pertussis toxin, reversibility
control mice. After 5 days, inject intraperitoneally into each of toxoid, antimicrobial preservative, free formaldehyde and
mouse 1 mg of histamine base in a volume not exceeding 0.5 the assay have been carried out with satisfactory results on
ml. Record the number of animals that die within 24 h of the final bulk vaccine, these tests may be omitted on the final
histamine challenge. Calculate the weight or volume of a lot.
preparation that sensitises 50.0 per cent of the mice injected
using a suitable statistical method such as probit analysis. Identification
The residual activity of pertussis toxin does not exceed that Subject the vaccine to a suitable desorption procedure such
of batches shown to be safe in clinical studies. as the following: dissolve in the vaccine under examination
The histamine sensitivity of the strain of mice used is verified sufficient sodium citrate to give a 10 per cent w/v solution;
at suitable intervals as follows: inject threefold dilutions of a maintain at 37° for about 16 h and centrifuge until a clear
reference pertussis toxin preparation in phosphate-buffered supernatant is obtained. Examine by a suitable immunochemical
saline solution containing 0.2 per cent w/v of gelatin and method (2.2.14), the clear supernatant reacts with specific
challenge with histamine as described above; the strain is antisera to the components in the vaccine.
suitable if more than 50 per cent of the animals are sensitised
by 50 ng of pertussis toxin and none of the control animals Tests
injected with only diluent and challenged similarly with Test for residual pertussis toxin. Use 3 groups of not fewer
histamine show symptoms of sensitisation. than 5 histamine-sensitive mice (see under Production) each
A validated test based on the clustering effect of the toxin for weighing between 18 and 26 g. Using phosphate-buffered
Chinese hamster ovary (CHO) cells may be used instead of saline containing 0.2 per cent w/v of gelatin, prepare a series
the test on mice. of dilutions of the vaccine under examination that have been
shown to yield a graded response and attribute each dilution
Residual detoxifying agents and other reagents. The content to a separate group of mice. Inject intraperitoneally into each
of residual detoxifying agents and other reagents is determined mouse the dilution attributed to its group. Inject diluent into
and shown to be below approved limits unless validation of a fourth group of control mice. After 5 days, inject
the process has demonstrated acceptable clearance. intraperitoneally into each mouse 1 mg of histamine base in a
Antigen content. Determine the complete quantitative antigen volume not exceeding 0.5 ml. Note the number of animals that
composition of the antigenic fraction by suitable die within 24 h of histamine challenge. Calculate the weight or
immunochemical methods (2.2.14) and protein nitrogen by volume of a preparation that sensitises 50 per cent of the mice
sulphuric acid digestion or another suitable method. The ratio injected using a suitable statistical method such as probit
of total antigen content to protein nitrogen is within the limits analysis. The residual activity of pertussis toxin does not
established for the product. exceed that of batches shown to be safe in clinical studies.

24
IP 2007 BACILLUS CALMETTE-GUERIN VACCINE (FREEZE-DRIED)

Reversibility of toxoid. Carry out the test for residual pertussis designed that all cultures and vaccines are protected from
toxin described above using the vaccine incubated at 37° for direct sunlight and from ultraviolet light at all stages of
4 weeks in parallel with a sample stored at 2° to 8°. The degree manufacture, testing and storage.
of reversibility does not exceed that of batches shown to be Production of the vaccine is based on a seed-lot system. The
safe in clinical studies. production method shall have been shown to yield
Antimicrobial preservative. Where applicable, determine the consistently BCG vaccines that induce adequate sensitivity
amount of antimicrobial preservative by a suitable chemical to tuberculin in man, that have acceptable protective potency
method. The content is not less than 85.0 per cent and not in animals and are safe. The vaccine is prepared from cultures
greater than 115.0 per cent of the intended amount. which are derived from the master seed lot by as few
subcultures as possible and in any case not more than 12
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
subcultures e.g. If the secondary seed lot is 4 culture passages
if aluminium hydroxide or hydrated aluminium phosphate is
removed from the primary seed lot, the no. of passages from
used as the adsorbent.
the secondary seed lot must not exceed 8.
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
The capacity of the working seed lot to induce sensitivity to
Sterility (2.2.11). Complies with the test for sterility. tuberculin in guinea-pigs is demonstrated.
Assay If a bioluminescence test or other biochemical method is used
instead of viable count, the method is validated against the
The vaccine complies with the assay as stated under Adsorbed
viable count for each stage of the process at which it is used.
Pertussis Vaccine (Acellular Component).
SEED LOT
Labelling. The label states (1) the names and amounts of the
antigenic components present in the vaccine; (2) the maximum The strain used to establish the master seed lot is chosen for
amount of residual pertussis toxin present in the vaccine; (3) and maintained to preserve its stability, its capacity to sensitise
the maximum degree of reversion of toxoid to toxin during the man and guinea-pigs to tuberculin and to protect animals
period of validity; (4) the name and amount of the adsorbent; against tuberculosis, and its relative absence of pathogenicity
(5) that the vaccine must be shaken before use; (6) that the for man and laboratory animals. The strain used shall be
vaccine is not to be frozen. identified by historical records that include information on its
origin and subsequent manipulation.
A suitable batch of vaccine is prepared from the first working
Bacillus Calmette-Guerin Vaccine seed lot and is reserved for use as the comparison/ in-house
reference vaccine. When a new working seed lot is
(Freeze-Dried) established, a suitable test for delayed hypersensitivity in
Freeze-dried BCG Vaccine is a preparation of live bacteria guinea-pigs is carried out on a batch of vaccine prepared from
derived from a culture of the bacillus of Calmette and Guérin the new working seed lot; the vaccine is shown to be not
(Mycobacterium bovis BCG) capacity of which to protect significantly different in activity from the comparison vaccine.
against tuberculosis has been established. Only a working seed lot that complies with the following
Vaccine complies with the requirements stated under Vaccines requirements may be used for propagation.
with the following modifications. Identification
Production The bacteria in the working seed lot are identified as
Mycobacterium bovis BCG using microbiological techniques,
General provisions
which may be supplemented by molecular biology techniques
The production method is validated to demonstrate that the (for example, nucleic acid amplification and restriction-
product, if tested, would comply with the tests for safety and fragment-length polymorphism).
efficacy.
Sterility (2.2.11). Complies with the test for sterility, carried
BCG vaccine shall be produced by a staff consisting of healthy out using 10 ml for each medium. The working seed lot
persons who do not work with other infectious agents; in complies with the test for sterility except for the presence of
particular they shall not work with virulent strains of mycobacteria.
Mycobacterium tuberculosis, during the course of production
Virulent mycobacteria
cycle nor shall they be exposed to a known risk of tuberculosis
infection. BCG vaccine is susceptible to sunlight: the Examine the working seed lot as prescribed under Tests, using
procedures for the preparation of the vaccine shall be so 10 guinea pigs.

25
BACILLUS CALMETTE-GUERIN VACCINE (FREEZE-DRIED) IP 2007

PROPAGATION AND HARVEST Except where the filled and closed containers are stored at a
temperature of -20° or lower, the expiry date is not later than 4
The bacteria are grown in a suitable medium for not more than
years from the date of harvest.
21 days by surface or submerged culture. The culture medium
shall contain no substances known to cause toxic or allergic Only a final lot that complies with the following requirement
reactions in human beings or to cause the bacteria to become for count of viable units and with each of the requirements
virulent for guinea-pigs. The culture is harvested and given below under Identification, Tests and Assay may be
suspended in a sterile liquid medium that protects the viability released for use. Provided the test for virulent mycobacteria
of the vaccine as determined by a suitable method of viable has been carried out with satisfactory results on the final bulk
count. vaccine, it may be omitted on the final lot. Provided the test
for excessive dermal reactivity has been carried out with
Test for purity. Purity is checked by acid fast staining. satisfactory results on the working seed lot and on 5
FINAL BULK VACCINE consecutive final lots produced from it, the test may be omitted
on the final lot.
The final bulk vaccine is prepared from a single harvest or by
pooling a number of single harvests. A stabiliser may be Count of viable units
added; if the stabiliser interferes with the determination of
Determine the number of viable units per ml of the
bacterial concentration on the final bulk vaccine, the
reconstituted vaccine by viable count on solid medium using
determination is carried out before addition of the stabiliser.
a method suitable for the vaccine under examination or by
Only final bulk vaccine that complies with the following determination of adenosine triphosphate by a bioluminescence
requirements may be used in the preparation of the final lot. reaction. The ratio of the count of viable units after freeze-
Virulent mycobacteria. Examine as prescribed under Tests. drying to that before is not less than that approved for the
particular product.
Sterility (2.2.11). Complies with the test for sterility using 10
ml for each medium except for the presence of mycobacteria. Identification
BCG vaccine is identified by microscopic examination of the
Count of viable units bacilli in stained smears demonstrating their acid-fast property
Determine the number of viable units per ml by viable count and by the characteristic appearance of colonies grown on
on solid medium using a method suitable for the vaccine under solid medium. Alternatively, molecular biology techniques (like
examination or by determination of adenosine triphosphate nucleic acid amplification) may be used.
by a bioluminescence reaction. Carry out the test in parallel Tests
on a reference preparation of the same strain.
Virulent mycobacteria
Bacterial concentration
If this test is satisfactory at final bulk stage it can be omitted at
Determine the total bacterial concentration by a suitable the final lot.
method, either directly by determining the mass of the micro-
organisms, or indirectly by an opacity method that has been Inject subcutaneously or intramuscularly into each of 6 guinea-
calibrated in relation to the mass of the organisms; if the pigs, each weighing between 250 and 400 g and having received
bacterial concentration is determined before addition of a no treatment likely to interfere with the test, a quantity of
stabiliser, the concentration in the final bulk vaccine is vaccine equivalent to at least 50 human doses. Observe the
established by calculation. The total bacterial concentration animals for at least 42 days. At the end of this period, kill the
is within the limits approved for the particular product by guinea-pigs and examine by autopsy for signs of infection
National Regulatory Authority. with tuberculosis, ignoring any minor reactions at the site of
injection. Animals that die during the observation period are
The ratio of the count of viable units to the total bacterial also examined for signs of tuberculosis. The vaccine complies
concentration is not less than that approved for the particular with the test if none of the guinea-pigs shows signs of
product by National Regulatory Authority. tuberculosis and if not more than one animal dies during the
observation period. If 2 animals die during this period and
FINAL LOT
autopsy does not reveal signs of tuberculosis repeat the test
The final bulk vaccine is distributed into sterile containers on 6 other guinea-pigs. The vaccine complies with the test if
and freeze-dried to a moisture content favourable to the not more than one animal dies during the 42 days following
stability of the vaccine; the containers are closed either under the injection and autopsy does not reveal any sign of
vacuum or under a gas that is not deleterious to the vaccine. tuberculosis.

26
IP 2007 DIPHTHERIA AND TETANUS VACCINE (ADSORBED)

Sterility (2.2.11). The reconstituted vaccine complies with the Production


test for sterility except for the presence of mycobacteria.
General provisions
Excessive dermal reactivity
Bulk purified diphtheria and tetanus toxoids
Use 6 healthy white or pale-coloured guinea-pigs, each
weighing not less than 250 g and having received no treatment The bulk purified diphtheria and tetanus toxoids are prepared
likely to interfere with the test. Inject intradermally into each as described in the monographs on Diphtheria vaccine
guinea-pig, according to a randomised plan, 0.1 ml of the (adsorbed) and Tetanus vaccine (adsorbed) and comply with
reconstituted vaccine and of 2 tenfold serial dilutions of the the requirements prescribed therein.
vaccine and identical doses of the comparison vaccine. Sterility (2.2.11). Carry out test for sterility using 10 ml of
Observe the lesions formed at the site of the injection for 4 bulk for each sterility medium.
weeks. The vaccine complies with the test if the reaction it
produces is not markedly different from that produced by the Absence of toxin and irreversibility of toxoid
comparison vaccine. Inject subcutaneously into each of 5 guinea-pigs at least 500 Lf
of the non-incubated bulk purified toxoid in a volume of 1 ml,
Temperature stability
using the same buffer solution as for the final vaccine, without
Maintain samples of the freeze-dried vaccine at 37° for 4 weeks. adsorbent. Animals that die shall be autopsied and examined
Determine the number of viable units in the heated vaccine for symptoms of diphtheria intoxication (red adrenals). The
and in unheated vaccine as described below. The number of bulk purified toxoid shall pass the test if no guinea-pig shows
viable units in the heated vaccine is not less than 20.0 per cent symptoms of specific intoxication within six weeks of injection
of that in unheated vaccine. and if at least 80 per cent of the animals survive the test period.
The guinea-pigs shall not have been used previously for
Water (2.3.43). Not more than 3.0 per cent, determined by the
experimental purposes.
semi-micro determination of water.
Alternatively, a cell-culture test system may be used; in this
Assay case, the sensitivity of the test shall have been demonstrated
to be not less than that of the guinea-pig test, and the test
Determine the number of viable units in the reconstituted
procedures shall be approved by the National Regulatory
vaccine by viable count on solid medium or using a suitable
Authority.
validated biochemical method for the vaccine under
examination. The number is within the range stated on the Each bulk purified toxoid shall be tested to ensure that
label. Determine the number of viable units in the comparison reversion to toxicity cannot take place on storage. The bulk
vaccine in parallel. purified toxoid shall be diluted in order to obtain the same
concentration and chemical environment as that present in
Labelling. The label states (1) the minimum and maximum
the final bulk vaccine, except for the presence of adjuvant.
number of viable units per ml in the reconstituted vaccine; (2)
that the vaccine must be protected from direct sunlight; (3) To determine whether reversion has occurred, diluted toxoids
that the vaccine is to be used immediately after broaching the that have been stored at 37° for six weeks shall be tested. The
container; (4) the age group for which the vaccine is intended; test employed shall be approved by the National Regulatory
(5) the dose for each age group; (6) follow instructions as Authority and should be sufficiently sensitive to detect very
mentioned in the product insert/leaflet. small amounts of toxin. No toxicity shall be detected.
Intradermal tests in guinea-pigs and cell-culture tests both
are considered to be suitable.
Diphtheria and Tetanus Vaccine Antigenic purity
(Adsorbed)
Not less than 1500 Lf per mg of protein nitrogen for diphtheria
Diphtheria and Tetanus Vaccine (Adsorbed) is a preparation toxoid and not less than 1000 Lf/mg of protein nitrogen for
of diphtheria formol toxoid and tetanus formol toxoid adsorbed tetanus toxoid.
on mineral carrier. The formol toxoids are prepared from the
toxins produced by the growth of Corynebacterium FINAL BULK VACICNE
diphtheriae and Clostridium tetani, respectively. The final bulk vaccine is prepared by adsorption of suitable
The specification for individual component used in formulation quantities of bulk purified diphtheria toxoid and tetanus toxoid
is referred in the text of individual monograph. onto mineral carrier such as hydrated aluminium phosphate,

27
DIPHTHERIA AND TETANUS VACCINE (ADSORBED) IP 2007

aluminium hydroxide; the resulting mixture is approximately Only a final lot that is satisfactory with respect to each of the
isotonic with blood. Suitable antimicrobial preservatives may requirements given below under Identification, Tests and
be added. Antimicrobial preservatives of the phenolic type Assay may be released for use. Provided the tests for specific
must not be used. toxicity, free formaldehyde and antimicrobial preservative and
Only final bulk vaccine that complies with the following the assay have been carried out with satisfactory results on
requirements may be used in the preparation of the final lot. the final bulk vaccine, they may be omitted on the final lot.

Identification Identification
A. Dissolve sufficient sodium citrate in the vaccine under A. Diphtheria toxoid is identified by a suitable immuno-
examination to give a 10 per cent w/v concentration. Maintain chemical method (2.2.14).
at 37o for about 16 hours and centrifuge. The clear supernatant Dissolve in the vaccine under examination by adding sufficient
reacts with a suitable diphtheria antitoxin and yields a sodium citrate to give a 10 per cent w/v solution. Maintain at
precipitate. 37° for about 16 hours and centrifuge until a clear supernatant
liquid is obtained. The clear supernatant reacts with a suitable
B. The clear supernatant obtained in test A reacts with a suitable
diphtheria antitoxin, giving a precipitate or visible floccules.
tetanus antitoxin and yields a precipitate.
B. Tetanus toxoid is identified by a suitable immuno-chemical
pH (2.4.24). 6.0 to 7.0. method (2.2.14).
Specific toxicity. Use 5 normal, healthy guinea-pigs weighing
The clear supernatant liquid obtained during test A reacts
between 250 and 350 g which have been maintained for at
with a suitable tetanus antitoxin, giving a precipitate or visible
least 1 week on a uniform, unrestricted diet, and have not
floccules.
been previously treated with any material that will interfere
with the test. Weigh the animals separately and record their Tests
weights. Inject subcutaneously into each animal 5 times the
dose stated on the label. Weigh all the animals at weekly Sterility (2.2.11). Complies with the test for sterility.
intervals for 6 weeks. None of the animals shows any Abnormal toxicity (2.2.1). Complies with the test for abnormal
symptoms of diphtheria or tetanus toxaemia or dies from toxicity
diphtheria within 42 days or loses weight at the end of the
test. If more than one animal dies from non-specific causes or Aluminium (2.3.9.). Not more than 1.25 mg per single human
loses weight, repeat the test. If an animal dies or loses weight dose when hydrated aluminium phosphate or aluminium
in the second test, the vaccine fails the test. hydroxide is used as the adsorbent.
pH (2.4.24). The pH of the vaccine is within the range approved
Assay
for the product (6.0 to 7.0).
Diphtheria toxoid Free formaldehyde (2.3.20). Maximum 0.2 g/l.
Complies with the test as stated under Diphtheria Vaccine Antimicrobial preservative. Where applicable, determine the
(Adsorbed). amount of antimicrobial preservative by a suitable chemical
Tetanus toxoid method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the quantity stated on the label.
Complies with the test as stated under Tetanus Vaccine
(Adsorbed). Assay
Antimicrobial preservative. Where applicable, determine the
Diphtheria component
amount of antimicrobial preservative by a suitable chemical
method. The amount is not less than 85.0 per cent and not Carry out one of the described methods for the assay of
greater than 115.0 per cent of the intended amount. Diphtheria Vaccine (Adsorbed).
Free formaldehyde (2.3.20). Maximum 0.2 g/l Viz a) Intradermal challenge method, b) Lethal challenge
Sterility (2.2.11). Carry out test for sterility using 10 ml of method, c) Antibody induction method, d) Validated serological
bulk for each sterility medium. assay in guinea pigs or mice as approved by National
Regulatory Authority.
FINAL LOT
Tetanus component
The final bulk vaccine is filled and stored aseptically into
sterile containers. The containers are closed so as to prevent Carry out one of the described methods for the assay of
contamination. Tetanus Vaccine (Adsorbed) Viz a) Antibody induction

28
IP 2007 DIPHTHERIA AND TETANUS VACCINE (ADSORBED) FOR ADULTS AND ADOLESCENTS

method; b) Challenge method in guinea pigs/mice; c) Validated Specific toxicity


serological assay in guinea pigs or mice as approved by
Inject subcutaneously 5 times the single human dose stated
National Regulatory Authority.
on the label into each of 5 healthy guinea-pigs, each weighing
Labelling. The label states (1) the human dose; (2) the minimum between 250 and 350 g, that have not previously been treated
Lf units per single human dose or the minimum International with any material that will interfere with the test. If within 42
Units per single human dose if potency test done by challenge days of the injection any of the animals shows signs of or dies
method; (3) the name and the amount of the adsorbent and from diphtheria toxaemia or tetanus, the vaccine does not
preservative; (4) that the vaccine must be shaken before use; comply with the test. If more than 1 animal dies from non-
(5) that the vaccine is not to be frozen. specific causes, repeat the test once; if more than 1 animal
dies in the second test, the vaccine does not comply with the
test.
Diphtheria and Tetanus Vaccine Antimicrobial preservative. Where applicable, determine the
(Adsorbed) for Adults and Adolescents amount of antimicrobial preservative by a suitable
physicochemical method. The amount is not less than 85.0
Diphtheria and Tetanus Vaccine (Adsorbed) for Adults and per cent and not greater than 115.0 per cent of the intended
Adolescents is a preparation of diphtheria formol toxoid and amount.
tetanus formol toxoid adsorbed on a mineral carrier. The formol
toxoids are prepared from the toxins produced by the growth Free formaldehyde (2.3.20). Maximum 0.2 g/l.
of Corynebacterium diphtheriae and Clostridium tetani, Sterility (2.2.11). Carry out the test for sterility using 10 ml for
respectively. each medium.

Production FINAL LOT


The final bulk vaccine is distributed aseptically into sterile,
General provisions
tamper-proof containers. The containers are closed so as to
Bulk purified diphtheria and tetanus toxoids prevent contamination.

The bulk purified diphtheria and tetanus toxoids are prepared Only a final lot that is satisfactory with respect to each of the
as described in the monographs on Diphtheria vaccine requirements given below under Identification, Tests and
(adsorbed) and Tetanus vaccine (adsorbed) and comply with Assay may be released for use. Provided the tests for free
the requirements prescribed therein. formaldehyde and antimicrobial preservative and the assay
have been carried out with satisfactory results on the final
FINAL BULK VACCINE bulk vaccine, they may be omitted on the final lot.
The vaccine is prepared by adsorption of suitable quantities Identification
of bulk purified diphtheria toxoid and tetanus toxoid onto a
A. Diphtheria toxoid is identified by a suitable immunochemical
mineral carrier such as hydrated aluminium phosphate or
method (2.2.14). The following method, applicable to certain
aluminium hydroxide. Suitable antimicrobial preservatives may
vaccines, is given as an example. Dissolve in the vaccine under
be added. Certain antimicrobial preservatives, particularly
examination sufficient sodium citrate to give a 10 per cent
those of the phenolic type, adversely affect the antigenic
w/v solution. Maintain at 37° for about 16 hours and centrifuge
activity and must not be used.
until a clear supernatant is obtained. The clear supernatant
Only final bulk vaccine that complies with the following reacts with a suitable diphtheria antitoxin, giving a precipitate.
requirements may be used in the preparation of the final lot. If a satisfactory result is not obtained with a vaccine adsorbed
on aluminium hydroxide, carry out the test as follows.
Identification
Centrifuge 15 ml of the vaccine under examination and suspend
A. Dissolve sufficient sodium citrate in the vaccine under the residue in 5 ml of a freshly prepared mixture of 1 volume of
examination to give a 10 per cent w/v concentration. Maintain a 56 g/l solution of sodium edetate and 49 volumes of a
at 37o for about 16 hours and centrifuge. The clear supernatant 90 g/l solution of disodium hydrogen phosphate. Maintain at
reacts with a suitable diphtheria antitoxin and yields a 37° for not less than 6 hours and centrifuge. The clear
precipitate. supernatant reacts with a suitable diphtheria antitoxin, giving
B. The clear supernatant obtained in test A reacts with a suitable a precipitate.
tetanus antitoxin and yields a precipitate. B. Tetanus toxoid is identified by a suitable immunochemical
pH (2.4.24 ). 6.0 to 7.0. method (2.2.14). The following method, applicable to certain

29
DIPHTHERIA, TETANUS AND PERTUSSIS VACCINE (ADSORBED) IP 2007

vaccines, is given as an example. The clear supernatant respectively. The Bordetella pertussis suspension is prepared
obtained during identification test A reacts with a suitable by growth of suitable strains in an appropriate medium, under
tetanus antitoxin, giving a precipitate. controlled conditions.
Tests The specification for individual component used in formulation
is referred in the text of individual monograph.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose.
Free formaldehyde (2.3.20). Maximum 0.2 g/l. Production
Antimicrobial preservative. Where applicable, determine the
General provisions
amount of antimicrobial preservative by a suitable
physicochemical method. The amount is not less than 85.0 The production method must be validated to demonstrate
per cent and not greater than 115.0 per cent of the intended that the product if tested, would comply with the tests for
amount. safety as described under monographs of Diphtheria Vaccine,
Tetanus Vaccine (Adsorbed) and Pertussis Vaccine.
Sterility (2.2.11). Complies with the test for sterility.
pH (2.4.24). 6.0 to 7.0. Bulk purified diphtheria and tetanus toxoids, bulk inactivated
B. pertussis suspension
Abnormal toxicity (2.2.1). Complies with the test for abnormal
toxicity. The bulk purified diphtheria and tetanus toxoids and
inactivated B. pertussis suspension are prepared as described
Assay in the monograph on Diphtheria Vaccine (Adsorbed), Tetanus
Vaccine (Adsorbed) and Pertussis Vaccine respectively and
Diphtheria component comply with the respective requirements.
Carry out the prescribed method for assay of Diphtheria
Vaccine by lethal challenge method described in the assay of FINAL BULK VACCINE
Diphtheria Vaccine (Adsorbed). The final bulk vaccine is prepared by adsorption of suitable
The lower confidence limit (P = 0.95) of the estimated potency quantities of bulk purified diphtheria toxoid and tetanus toxoid
is not less than 2 IU per single human dose. onto hydrated aluminium phosphate or aluminium hydroxide
and admixture of an appropriate quantity of a suspension of
Tetanus component inactivated B. pertussis. The B. pertussis concentration of the
Carry out one of the prescribed methods for the assay as final bulk vaccine does not exceed that corresponding to an
stated under Tetanus Vaccine (Adsorbed). opacity of 20 I.U. per single human dose. If two or more strains
of B. pertussis are used, the composition of consecutive lots
The lower confidence limit (P = 0.95) of the estimated potency of the final bulk vaccine shall be consistent with respect to
is not less than 40 IU per single human dose. the proportion of each strain as measured in opacity units.
Labelling. The label states (1) the human dose; (2) the minimum Suitable antimicrobial preservatives may be added to the bulk
number of International Units of each component per single vaccine. Antimicrobial preservatives particularly those of
human dose, if potency determined by challenge method or phenolic type which affect the antigenic activity must not be
the minimum Lf units per single human dose if test done by used.
antibody induction method; (3) the name and the amount of Only final bulk vaccine that complies with the following
the adsorbent; (4) that the vaccine must be shaken before requirements may be used in the preparation of the final lot.
use; (5) that the vaccine is not to be frozen.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 percent and not
Diphtheria, Tetanus and Pertussis more than115.0 percent of the intended amount.
Vaccine (Adsorbed) Sterility (2.2.11). Carry out test for sterility using 10 ml of
bulk for each sterility medium.
Diphtheria, Tetanus and Pertussis Vaccine (Adsorbed) is a
preparation of diphtheria formol toxoid, tetanus formol toxoid FINAL LOT
adsorbed on mineral carrier and a suspension of killed
Bordetella pertussis organisms. The formal toxoids are The final bulk vaccine is filled and stored aseptically into
prepared from the toxins produced by the growth of sterile containers. The containers are closed so as to prevent
Corynebacterium diphtheriae and Clostridium tetani, contamination.

30
IP 2007 DIPHTHERIA, TETANUS AND PERTUSSIS VACCINE (ADSORBED)

Only a final lot that is satisfactory with respect to each of the diphtheria toxemia or tetanus, the vaccine does not comply
requirements given below under Identification, Tests and with the test. If more than one animal dies from non-specific
Assay may be released for use. Provided the tests for specific causes, repeat the test once; if more than one animal dies in
toxicity of diphtheria, tetanus and pertussis components, free the second test, the vaccine does not comply with the test.
formaldehyde, antimicrobial preservative and the Assay have Pertussis component. Use not less than 10 healthy mice each
been carried out with satisfactory results on the final bulk weighing between 14 and 16 g for the vaccine group and for
vaccine, they may be omitted on the final lot. the saline control. Use mice of the same sex or distribute males
Identification and females equally between the groups. Allow the animals
access to food and water for at least 2 hours before injection
A. Diphtheria toxoid is identified by a suitable immuno- and during the test. Inject each mouse of the vaccine group
chemical method (2.2.14). intraperitoneally with 0.5 ml, containing a quantity of the
vaccine equivalent to not less than half the single human
Dissolve in the vaccine under examination by adding sufficient
dose. Inject each mouse of the control group with 0.5 ml of a
sodium citrate to give a 10 per cent w/v solution. Maintain at
0.9 per cent sterile solution of sodium chloride, preferably
37° for about 16 hours and centrifuge until a clear supernatant
containing the same amount of antimicrobial preservative as
is obtained; reserve the precipitate for identification test C.
that injected with the vaccine. Weigh the mice groups
The clear supernatant reacts with a suitable diphtheria
immediately before the injection and 72 hours and 7 days after
antitoxin, giving a precipitate.
the injection. The vaccine complies with the test if: (a) at the
B. Tetanus toxoid is identified by a suitable immuno-chemical end of 72 hours the total mass of the group of vaccinated mice
method (2.2.14). is not less than that preceding the injection; (b) at the end of
The clear supernatant obtained during identification test A 7 days the average increase in mass per vaccinated mouse is
reacts with a suitable tetanus antitoxin, giving a precipitate. not less than 60 per cent of that per control mouse; and (c) not
more than 5 per cent of vaccinated mice should die during the
C. The pertussis component is identified by agglutination of test. The test may be repeated and the results of the tests
the bacteria from the resuspended centrifugation residue (see combined.
identification test A; other suitable methods for separating
the bacteria from the adsorbent may also be used) by antisera Aluminium (2.3.9). Not more than 1.25 mg per single human
specific to B. pertussis or by the assay of the pertussis dose, when hydrated aluminium phosphate or aluminium
component. hydroxide is used as the adsorbent.
pH (2.4.24). 6.0 to 7.0.
Tests
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
Sterility (2.2.11). Complies with the test for sterility.
Antimicrobial preservative (2.2.2 ) Where applicable,
Abnormal toxicity (2.2.1). Each final lot shall be tested for
determine the amount of antimicrobial preservative by a
abnormal toxicity by injecting intraperitoneally one human
suitable chemical method. The content is not less than 85.0
dose , but not more than 0.25 ml into each of the five mice
per cent and not more than115.0 per cent of the intended
weighing between 17 to 22 g and at least one human dose but
amount.
not more than 1.0 ml into each of the two guinea pigs weighing
between 250 and 350 g. The preparation passes the test if Assay
none of the animals dies or shows signs of ill health in 7 days Diphtheria component
following the injection. If one of the animal dies or shows the
signs of ill health, repeat the test. The preparation passes the Carry out one of the methods for the assay as stated under
test if none of the animals in the second group dies or shows Diphtheria Vaccine (Adsorbed).
signs of ill health in the time interval specified. Tetanus component
Specific toxicity Carry out one of the methods for the assay as stated under
Tetanus Vaccine (Adsorbed).
Diphtheria and tetanus components. Inject subcutaneously
five times the single human dose stated on the label into each Pertussis component
of five healthy guinea-pigs, each weighing between 250 and
Carry out the assay as stated under Pertussis Vaccine.
350 g, that have not previously been treated with any material
that will interfere with the test. The animals should be weighted Labelling. The label states (1) in case done by challenge
every week and observations be made. If within 42 days of the method the minimum number of International Units; if units
injection any of the animals shows signs of or dies from (as applicable for each component) per single human dose;

31
D., T., P., (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE (ADSORBED) IP 2007

(2) In case done by antibody induction method the minimum The final product contains a suitable antimicrobial
number of International Units per single human dose of preservative. The antigenic properties of the vaccine are
pertussis component and minimum number of Lf of diphtheria adversely affected by the presence of certain antimicrobial
toxoid and tetanus toxoid; (3) the name and the amount of the preservatives particularly those of the phenolic type and some
adsorbent and preservative; (4) that the vaccine must be of the quaternary ammonium type and must not be used.
shaken before use; (5) that the vaccine is not to be frozen; (6)
for vaccine contained in single-dose containers where the Production
space is too small to accommodate the full name of the vaccine,
the abbreviation ‘DTP’ may be used in the label and the General provisions
container provided that the same code is also stated in the The production method shall have been shown to yield
label on the package. consistently the vaccines comparable with the vaccine of
proven clinical efficacy and safety in man.
If the vaccine is presented with the haemophilus component
Diphtheria, Tetanus, Pertussis (Whole in a separate vial, as part of consistency studies the assays of
the diphtheria, tetanus, pertussis and hepatitis B are carried
Cell), Hepatitis B (rDNA) and out on a suitable number of batches of vaccine reconstituted
Haemophilus Type b Conjugate as for use. For subsequent routine control, the assays of these
Vaccine (Adsorbed) components may be carried out without mixing with the
haemophilus component.
Diphtheria, Tetanus, Pertussis (Whole cell), Hepatitis B (rDNA)
The production method is validated to demonstrate that the
and Haemophilus Type b Conjugate Vaccine (Adsorbed) is a
product, if tested, would comply with the following test for
combined vaccine composed of diphtheria formol toxoid
specific toxicity of the diphtheria and tetanus component:
containing not less than 1,500 Lf, (2.2.16) per mg of protein
inject subcutaneously 5 times the single human dose stated
nitrogen, purified tetanus formol toxoid containing not less
on the label into each of 5 healthy guinea pigs, each weighing
than 1,000 Lf, (2.2.16), per mg of protein nitrogen, hepatitis B
between 250 and 350 g, that have not previously been treated
surface antigen and haemophilus type b conjugated to suitable
with any material that will interfere with the test. If within 42
protein with a mineral adsorbent to which a suspension of
days of the injection any of the animals shows signs of or dies
killed Bordetella pertussis has been added. Mineral adsorbent
from diphtheria, toxemia or tetanus, the vaccine does not
is a suspension of hydrated aluminium hydroxide, aluminium
comply with the test. If more than 1 animal dies from non-
phosphate or calcium phosphate, in saline solution or other
specific causes, repeat the test once; if more than 1 animal
appropriate solution isotonic with blood.
shows signs of or dies in the second test, the vaccine does
The formol toxoids are prepared from the toxin produced by not comply with the test.
the growth of Corynebacterium diphtheriae and Clostridium The stability of the final lot and the relevant intermediates is
tetani, respectively, in suitable media. The toxins are converted evaluated using one or more indicator tests. For the
to toxoids by treatment with formaldehyde solution by haemophilus component, such tests may include determination
methods which avoid reversibility of the toxoids. of molecular size, determination of free PRP in the conjugate
Hepatitis B surface antigen is a component protein of hepatitis and kinetics of depolymerisation. Taking account of the results
B virus; the antigen is obtained by recombinant DNA of the stability testing, release requirements are set for these
technology. indicator tests to ensure that the vaccine will be satisfactory
at the end of the period of validity.
The polysaccharide, polyribosyl ribitol phosphate, PRP is a
linear copolymer composed of repeated units of 3-β-D- Reference vaccine(s)
ribofuranosyl-(1→1)-ribitol-5-phosphate [(C10H19O12P)n], with
Provided valid assays can be performed, monocomponent
a defined molecular size and derived from a suitable strain of
reference vaccines may be used for the assays on the combined
Haemophilus influenzae type b. The carrier protein, when
vaccine. If this is not possible because of interaction between
conjugated to PRP, is capable of inducing a T-cell dependent
the components of the combined vaccine or because of the
B-cell immune response to the polysaccharide.
difference in composition between monocomponent reference
The product may be presented with the haemophilus vaccine and the test vaccine, a batch of combined vaccine
component in a separate container, the contents of which are shown to be effective in clinical trials or a batch representative
mixed with the other components immediately before or during thereof is used as a reference vaccine. For the preparation of
use. a representative batch, strict adherence to the production

32
IP 2007 D., T., P., (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE (ADSORBED)

process used for the batch tested in clinical trials is necessary. Antimicrobial preservative. Where applicable, determine the
The reference vaccine may be stabilized by a method that has amount of antimicrobial preservative by a suitable chemical
been shown to have no effect on the assay procedure. method. The amount is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended content.
Production of the components
Sterility (2.2.11). Carry out test for sterility using 10 ml of
The production of the components complies with the bulk for each sterility medium.
requirements of the monographs on Diphtheria Vaccine
(Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine, FINAL LOT
Hepatitis B Vaccine (rDNA) and Haemophilus Type b
Only a final lot that is satisfactory with respect to the test for
Conjugate Vaccine.
osmolality and with respect to each of the requirements given
FINAL BULK VACCINE below under Identification, Tests and Assay may be released
for use. Provided the tests for specific toxicity of diphtheria
Vaccine with all components in the same container toxoid, tetanus toxoid and pertussis component and
antimicrobial preservative and the assays for the diphtheria,
The final bulk is prepared by adsorption, separately or tetanus and pertussis components have been carried out with
together, of suitable quantities of bulk purified diphtheria satisfactory results on the final bulk vaccine, they may be
toxoid, bulk purified tetanus toxoid, bulk purified hepatitis B omitted on the final lot. Provided the content of free
surface antigen onto a mineral carrier such as aluminium formaldehyde has been determined on the bulk purified
hydroxide or hydrated aluminium phosphate, admixture of an antigens or on the final bulk and it has been shown that the
appropriate quantity of a suspension of inactivated content in the final lot will not exceed 0.2 g/l, the test for free
B. pertussis component and admixture of a suitable quantity formaldehyde may be omitted on the final lot. If an in vivo
of PRP conjugate; the resulting mixture is approximately assay is used for the hepatitis B component, provided it has
isotonic with blood. The B. pertussis concentration of the been carried out with satisfactory results on the final bulk
final bulk vaccine does not exceed that corresponding to an vaccine, it may be omitted on the final lot.
opacity of 20 IU per single human dose. If 2 or more strains of
B. pertussis are used, the composition of consecutive lots of Free PRP
the final bulk vaccine shall be consistent with respect to the
proportion of each strain as measured in opacity units. Suitable Unbound PRP is determined after removal of the conjugate,
antimicrobial preservatives may be added. for example by anion exchange, size exclusion or hydrophobic
chromatography (2.4.16), ultrafiltration or other validated
Vaccine with the haemophilus component in a separate methods. The amount of free PRP is not greater than that
container approved for the particular product.
The final bulk is prepared by adsorption, separately or Osmolality (2.4.23). The osmolality of the vaccine is within
together, of suitable quantities of bulk purified diphtheria the limits approved for the particular preparation.
toxoid, bulk purified tetanus toxoid, bulk purified hepatitis B pH (2.4.24). 6.0 to 7.0.
surface antigen onto a mineral carrier such as aluminium
hydroxide or hydrated aluminium phosphate, admixture of an Description. Whitish turbid liquid in which the mineral carrier
appropriate quantity of a suspension of inactivated B. tends to settle down slowly on keeping.
pertussis component and admixture of a suitable quantity of
PRP conjugate; the resulting mixture is approximately isotonic
Identification
with blood. The B. pertussis concentration of the final bulk Tests A, B, C, D and E may be omitted if test F is carried out.
vaccine does not exceed that corresponding to an opacity of Test F may be omitted if tests A, B, C, D and E are carried out.
20 IU per single human dose. If 2 or more strains of B. pertussis
are used, the composition of consecutive lots of the final bulk A. Diphtheria toxoid. Dissolve sufficient sodium citrate in
vaccine shall be consistent with respect to the proportion of the vaccine under examination to give a 10 per cent w/v
each strain as measured in opacity units. The final bulk is concentration. Maintain at 37o for about 16 hours and
filled separately. Suitable antimicrobial preservatives may be centrifuge. Reserve the residue for test C. The clear
added. The final bulk of the haemophilus component is supernatant reacts with a suitable diphtheria antitoxin and
prepared by dilution of the bulk conjugate to the final yields a precipitate.
concentration with a suitable diluent. A stabilizer may be added. B. Tetanus toxoid. The clear supernatant obtained in test A
Only a final bulk vaccine that complies with the following reacts with a suitable tetanus antitoxin and yields a precipitate.
requirements may be used in the preparation of the final lot. C. Pertussis component. To a suspension of the residue

33
D., T., P., (WHOLE CELL), HEPATITIS B (rDNA) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE (ADSORBED) IP 2007

obtained in test A in saline solution add a suitable Bordetella shows signs of ill health, repeat the test. The preparation
pertussis antiserum; agglutination indicates presence of passes the test if none of the animals in the second group dies
pertussis component. or shows signs of ill health in the time interval specified.
D. Hepatitis B surface antigen. The suspension of the residue Pyrogens (2.2.8). This test is carried out for Haemophilus
obtained in test A gives a positive reactions when tested by influenzae type b vaccine only if Haemophilus influenzae type
suitable in-vitro assay. b vaccine is presented as separate lyophilized vial. The vaccine
complies with the test for pyrogens. Inject per kg of the rabbit’s
E. PRP. The suspension of the residue obtained in the test A
mass a quantity of the vaccine equivalent to: 1 mg of PRP for
gives a positive reaction when tested by a suitable
a vaccine with diphtheria toxoid or CRM 197 diphtheria toxoid
immunochemical method for PRP.
as carrier; 0.1 mg of PRP for a vaccine with tetanus toxoid as
F. The vaccine confers an active immunity in mice and guinea- carrier protein; 0.025 mg of PRP for vaccine with OMP as
pigs when administered as directed in the test for Assay. carrier.

Tests Specific toxicity

If the product is presented with the haemophilus component Diphtheria and tetanus components
in a separate container; the tests for specific toxicity of Complies with the test as stated under Diphtheria and Tetanus
diphtheria toxoid, tetanus toxoid and pertussis component, Vaccine (Adsorbed).
aluminium, free formaldehyde, antimicrobial preservative
and sterility are carried out on the container with the Pertussis component
diphtheria, tetanus, pertussis and hepatitis B components; Complies with the test as stated under Diphtheria, Tetanus
the tests for PRP content, water (where applicable), sterility and Pertussis Vaccine (Adsorbed).
and pyrogens are carried out on the container with the
haemophilus component. Assay
If the haemophilus component is freeze-dried, some tests may Diphtheria toxoid (adsorbed)
be carried out on the freeze-dried product rather than on the
bulk conjugate where the freeze-drying process may affect Complies with the test as stated under Diphtheria and Tetanus
the component under test. Vaccine (Adsorbed).
PRP. Not less than 80.0 per cent of the amount of PRP stated Tetanus toxoid (adsorbed)
on the label. PRP is determined either by assay of ribose (2.7.1), Complies with the test as stated under assay of Tetanus Vaccine
or phosphorus (2.7.1), by an immunochemical method (2.2.14) (Adsorbed).
or by anion exchange liquid chromatography with pulsed
amperometric detection. Pertussis vaccine
Aluminium (2.3.9). Not more than 1.25 mg per single human Complies with the test as stated under Diphtheria, Tetanus
dose, if aluminium hydroxide or hydrated aluminium phosphate and Pertussis Vaccine (Adsorbed).
is used as the adsorbent.
Hepatitis B surface antigen (adsorbed)
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
Complies with the test as stated under Hepatitis B Vaccine
Antimicrobial preservative. Where applicable, determine the (Adsorbed).
amount of antimicrobial preservative by a suitable chemical
Storage. When stored under the prescribed conditions the
method. The content is not less than 85.0 per cent and not
vaccine may be expected to retain potency for not less than 2
greater than 115.0 per cent of the intended amount.
years from the date on which the potency test for the pertussis
Sterility (2.2.11). Complies with the test for sterility. component was started.
Abnormal toxicity (2.2.1). Each final lot shall be tested for Labelling. The label states (1) the human dose; (2) Diphtheria
abnormal toxicity by injecting intraperitoneally one human and Tetanus components; (a) in case done by challenge
dose, but not more than 0.25 ml into each of five mice weighing method, the minimum number of International Units (as
between 17 and 22 g and at least one human dose but not applicable for each component) per single human dose; (b) in
more than 1.0 ml into each of two guinea pigs weighing case done by antibody induction, the minimum Lf units per
between 250 and 350 g. The preparation passes the test if single human dose; (3) pertussis component – IU or IOU per
none of the animals dies or shows signs of ill health in seven single human dose; (4) hepatitis B component - mg HBsAg
days following the injection. If one of the animals dies or per single human dose; (5) haemophilus conjugate component

34
IP 2007 DIPHTHERIA TETANUS PERTUSSIS (WHOLE CELL), HEPATITIS B (rDNA) VACCINE (ADSORBED)

- mg PRP per single human dose; (6) the type and nominal from diphtheria, toxaemia or tetanus, the vaccine does not
amount of carrier protein per single human dose; (7) the name comply with the test. If more than 1 animal dies from non-
and amount of adsorbent and added preservative; (8) that the specific causes, repeat the test once; if more than 1 animal
vaccine must be shaken before use; (9) that the vaccine is not shows signs of or dies in the second test, the vaccine does
to be frozen. not comply with the test.
The stability of the final lot and the relevant intermediates is
evaluated using one or more indicator tests. Taking account
Diphtheria, Tetanus, Pertussis (Whole of the results of the stability testing, release requirements are
set for these indicator tests to ensure that the vaccine will be
Cell) and Hepatitis B (rDNA) Vaccine satisfactory at the end of the period of validity.
(Adsorbed) Reference vaccine(s)
Diphtheria, Tetanus, Pertussis and Hepatitis B (rDNA) Vaccine Provided valid assays can be performed, monocomponent
(Adsorbed) is a combined vaccine composed of diphtheria reference vaccines may be used for the assays on the combined
formol toxoid containing not less than 1,500 Limes flocculationis vaccine. If this is not possible because of interaction between
(Lf) (2.2.16), per mg of protein nitrogen, purified tetanus formol the components of the combined vaccine or because of the
toxoid containing not less than 1,000 Lf ( 2.2.16), per mg of difference in composition between monocomponent reference
protein nitrogen and hepatitis B surface antigen with a mineral vaccine and the test vaccine, a batch of combined vaccine
adsorbent to which a suspension of killed Bordetella pertussis shown to be effective in clinical trials or a batch representative
has been added. Mineral adsorbent is a suspension of thereof is used as a reference vaccine.
hydrated aluminium hydroxide, aluminium phosphate or
calcium phosphate in saline solution or other appropriate Production of the components
solution isotonic with blood.
The production of the components complies with the
The formol toxoids are prepared from the toxin produced by requirements of the monographs on Diphtheria Vaccine
the growth of Corynebacterium diphtheriae and Clostridium (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
tetani, respectively, in suitable media. The toxins are converted and Hepatitis B Vaccine (rDNA).
to toxoids by treatment with formaldehyde solution by
methods, which avoid reversibility of the toxoids. FINAL BULK VACCINE
Hepatitis B surface antigen is a component protein of hepatitis The final bulk vaccine is prepared by adsorption, separately
B virus; the antigen is obtained by recombinant DNA or together, of suitable quantities of bulk purified diphtheria
technology. toxoid, tetanus toxoid, pertussis components and hepatitis B
surface antigen onto a mineral carrier such as aluminium
The final product contains a suitable antimicrobial
hydroxide or hydrated aluminium phosphate. Suitable
preservative. The antigenic properties of the vaccine are
antimicrobial preservatives may be added. Only a final bulk
adversely affected by the presence of certain antimicrobial
vaccine that complies with the following requirements may be
preservatives particularly those of the phenolic type and some
used in the preparation of the final lot.
of the quaternary ammonium type and must not be used.
Antimicrobial preservative. Where applicable, determine the
Production amount of antimicrobial preservative by a suitable chemical
method. The amount is not less than 85.0 per cent and not
General provisions greater than 115.0 per cent of the intended content.
The production method shall have been shown to yield pH (2.4.24). 6.0 to 7.0.
consistently the vaccines comparable with the vaccine of Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
proven clinical efficacy and safety in man. for each sterility medium.
The production method is validated to demonstrate that the
FINAL LOT
product, if tested, would comply with the following test for
specific toxicity of the diphtheria and tetanus component: Only a final lot that is satisfactory with respect to the test for
inject subcutaneously 5 times the single human dose stated osmolality and with respect to each of the requirements given
on the label into each of 5 healthy guinea pigs, each weighing below under Identification, Tests and Assay may be released
between 250 and 350 g, that have not previously been treated for use. Provided the tests for specific toxicity of diphtheria
with any material that will interfere with the test. If within 42 toxoid, tetanus toxoid and pertussis component and
days of the injection any of the animals shows signs of or dies antimicrobial preservative and the assays for the diphtheria,

35
DIPHTHERIA TETANUS PERTUSSIS (WHOLE CELL), HEPATITIS B (rDNA) VACCINE (ADSORBED) IP 2007

tetanus and pertussis components have been carried out with Abnormal toxicity (2.2.1). Each final lot shall be tested for
satisfactory results on the final bulk vaccine, they may be abnormal toxicity by injecting intraperitoneally one human
omitted on the final lot. Provided the content of free dose, but not more than 0.25 ml into each of five mice weighing
formaldehyde has been determined on the bulk purified between 17 and 22 g and at least one human dose but not
antigens or on the final bulk and it has been shown that the more than 1.0 ml into each of two guinea pigs weighing
content in the final lot will not exceed 0.2 g/l, the test for free between 250 and 350 g. The preparation passes the test if
formaldehyde may be omitted on the final lot. If an in vivo none of the animals dies or shows signs of ill health in seven
assay is used for the hepatitis B component, provided it has days following the injection. If one of the animals dies or
been carried out with satisfactory results on the final bulk shows signs of ill health, repeat the test. The preparation
vaccine, it may be omitted on the final lot. passes the test if none of the animals in the second group dies
Osmolality (2.4.23). The osmolality of the vaccine is within or shows signs of ill health in the time interval specified.
the limits approved for the particular preparation. Specific toxicity
Description. Whitish turbid liquid in which the mineral carrier
tends to settle down slowly on keeping. Diphtheria and tetanus components
Complies with the test as stated under Diphtheria and Tetanus
Identification Vaccine (Adsorbed).
Tests A, B, C and D may be omitted if test E is carried out. Test Pertussis component
E may be omitted if tests A, B C and D are carried out. Complies with the test as stated under Diphtheria, Tetanus
A. Diphtheria toxoid. Dissolve sufficient sodium citrate in and Pertussis Vaccine (Adsorbed).
the vaccine under examination to give a 10 per cent w/v
Assay
concentration. Maintain at 37o for about 16 hours and
centrifuge. Reserve the residue for test C. The clear Diphtheria toxoid (adsorbed)
supernatant reacts with a suitable diphtheria antitoxin and Complies with the test as stated under Diphtheria and Tetanus
yields a precipitate. Vaccine (Adsorbed).
B. Tetanus toxoid. The clear supernatant obtained in test A Tetanus toxoid (adsorbed)
reacts with a suitable tetanus antitoxin and yields a precipitate.
Complies with the test as stated under assay for Tetanus
C. Pertussis component. To a suspension of the residue Vaccine (Adsorbed).
obtained in test A in saline solution add a suitable B. pertussis
antiserum; agglutination indicates presence of pertussis Pertussis vaccine
component. Complies with the test as stated under Diphtheria, Tetanus
D. Hepatitis B surface antigen. The suspension of the residue and Pertussis Vaccine (Adsorbed).
obtained in test A gives a positive reactions when tested by
Hepatitis B surface antigen (adsorbed)
suitable in-vitro assay.
Complies with the test as stated under Hepatitis B Vaccine
E. The vaccine confers an active immunity in mice and guinea-
(Adsorbed).
pigs when administered as directed under Assay.
Storage. When stored under the prescribed conditions the
Tests vaccine may be expected to retain potency for not less than 2
years from the date on which the potency test for the pertussis
pH (2.4.24). 6.0 to 7.0.
component was started.
Aluminium (2.3.9). Not more than 1.25 mg per single human Labelling. The label states (1) the human dose (ml); (2)
dose, if aluminium hydroxide or hydrated aluminium phosphate diphtheria and tetanus components, (a) in case done by
is used as the adsorbent. challenge method, the minimum number of International Units
Free formaldehyde (2.3.20). Maximum 0.2 g/l. (as applicable for each component) per single human dose
and (b) in case done by antibody induction, the minimum Lf
Antimicrobial preservative. Where applicable, determine the
units per single human dose; (3) pertussis component - IU or
amount of antimicrobial preservative by a suitable chemical
IOU per single human dose; (4) hepatitis B component - mg
method. The content is not less than 85.0 per cent and not
HBsAg per single human dose; (5) the name and amount of
greater than 115.0 per cent of the intended amount.
adsorbent and added preservative; (6) that the vaccine must
Sterility (2.2.11). Complies with the test for sterility. be shaken before use; (7) that the vaccine is not to be frozen.

36
IP 2007 D., T., P., (WHOLE CELL) AND HAEMOPHILUS TYPE B CONJUGATE VACCINE (ADSORBED)

Diphtheria, Tetanus, Pertussis (Whole The production method is validated to demonstrate that the
product, if tested, would comply with the following test for
Cell) and Haemophilus Type b specific toxicity of the diphtheria and tetanus component:
Conjugate Vaccine (Adsorbed) inject subcutaneously 5 times the single human dose stated
on the label into each of 5 healthy guinea pigs, each weighing
Diphtheria, Tetanus, Pertussis and Haemophilus type b between 250 and 350 g, that have not previously been treated
Conjugate Vaccine (Adsorbed) is a combined vaccine with any material that will interfere with the test. If within 42
composed of diphtheria formol toxoid containing not less than days of the injection any of the animals shows signs of or dies
1,500 Limes flocculationis; (Lf), (2.2.16) per mg of protein from diphtheria, toxemia or tetanus, the vaccine does not
nitrogen, purified tetanus formol toxoid containing not less comply with the test. If more than 1 animal dies from non-
than 1,000 Lf, (2.2.16), per mg of protein nitrogen, and specific causes, repeat the test once; if more than 1 animal
Haemophilus type b conjugated to suitable protein with a shows signs of or dies in the second test, the vaccine does
mineral adsorbent to which a suspension of killed Bordetella not comply with the test.
pertussis has been added. The mineral adsorbent is a
suspension of hydrated aluminium hydroxide, aluminium The stability of the final lot and the relevant intermediates is
phosphate or calcium phosphate, in saline solution or other evaluated using one or more indicator tests. For the
appropriate solution isotonic with blood. haemophilus component, such tests may include determination
of molecular size, determination of free PRP in the conjugate
The formol toxoids are prepared from the toxin produced by
and kinetics of depolymerisation. Taking account of the results
the growth of Corynebacterium diphtheriae and Clostridium
of the stability testing, release requirements are set for these
tetani, respectively in suitable media. The toxins are converted
indicator tests to ensure that the vaccine will be satisfactory
to toxoids by treatment with formaldehyde solution by
at the end of the period of validity.
methods which avoid reversibility of the toxoids.
The polysaccharide, polyribosyl ribitol phosphate, PRP is a Reference vaccine(s)
linear copolymer composed of repeated units of Provided valid assays can be performed, monocomponent
3-β-D-ribofuranosyl-(1→1)-ribitol-5-phosphate reference vaccines may be used for the assays on the combined
[(C10H19O12P)n], with a defined molecular size and derived from vaccine. If this is not possible because of interaction between
a suitable strain of Haemophilus influenzae type b. The carrier the components of the combined vaccine or because of the
protein, when conjugated to PRP, is capable of inducing a T- difference in composition between monocomponent reference
cell dependent B-cell immune response to the polysaccharide. vaccine and the test vaccine, a batch of combined vaccine
The product may be presented with the haemophilus shown to be effective in clinical trials or a batch representative
component in a separate container, the contents of which are thereof is used as a reference vaccine. For the preparation of
mixed with the other components immediately before or during a representative batch, strict adherence to the production
use. process used for the batch tested in clinical trials is necessary.
The reference vaccine may be stabilized by a method that has
The final product contains a suitable antimicrobial
been shown to have no effect on the assay procedure.
preservative. The antigenic properties of the vaccine are
adversely affected by the presence of certain antimicrobial Production of the components
preservatives particularly those of the phenolic type and some
of the quaternary ammonium type must not be used. The production of the components complies with the
requirements of the monographs on Diphtheria Vaccine
Production (Adsorbed), Tetanus Vaccine (Adsorbed), Pertussis Vaccine
(Whole Cell) and Haemophilus influenzae Type b Conjugate
General provisions
Vaccine.
The production method shall have been shown to yield
consistently the vaccines comparable with the vaccine of FINAL BULK VACCINE
proven clinical efficacy and safety in man.
Vaccine with all components in the same container
If the vaccine is presented with the haemophilus component
in a separate vial, as part of consistency studies, the assays The final bulk is prepared by adsorption, separately or
of the diphtheria, tetanus and pertussis are carried out on a together, of suitable quantities of bulk purified diphtheria
suitable number of batches of vaccine reconstituted for use. toxoid, bulk purified tetanus toxoid, onto a mineral carrier such
For subsequent routine control, the assays of these as aluminium hydroxide or hydrated aluminium phosphate,
components may be carried out without mixing with the admixture of an appropriate quantity of a suspension of
haemophilus component. inactivated B. pertussis component and admixture of a suitable

37
D., T., P., (WHOLE CELL) AND HAEMOPHILUS TYPE CONJUGATE VACCINE (ADSORBED) IP 2007

quantity of PRP conjugate; the resulting mixture is final lot. Provided the content of free formaldehyde has been
approximately isotonic with blood. The B. pertussis determined on the bulk purified antigens or on the final bulk
concentration of the final bulk vaccine does not exceed that and it has been shown that the content in the final lot will not
corresponding to an opacity of 20 IU per single human dose. exceed 0.2 g/l, the test for free formaldehyde may be omitted
If 2 or more strains of B. pertussis are used, the composition of on the final lot.
consecutive lots of the final bulk vaccine shall be consistent
Free PRP. Unbound PRP is determined after removal of the
with respect to the proportion of each strain as measured in
conjugate, for example by anion exchange, size exclusion or
opacity units. Suitable antimicrobial preservatives may be
hydrophobic chromatography (2.4.16), ultrafiltration or other
added.
validated methods. The amount of free PRP is not greater than
Vaccine with the haemophilus component in a separate approved for the particular product.
container Osmolality (2.4.23). The osmolality of the vaccine is within
the limits approved for the particular preparation.
The final bulk is prepared by adsorption, separately or
together, of suitable quantities of bulk purified diphtheria pH (2.4.24). 6.0 to 7.0.
toxoid, bulk purified tetanus toxoid onto a mineral carrier such
Description. Whitish turbid liquid in which the mineral carrier
as aluminium hydroxide or hydrated aluminium phosphate,
tends to settle down slowly on keeping.
admixture of an appropriate quantity of a suspension of
inactivated B. pertussis component and admixture of a suitable Production
quantity of PRP conjugate; the resulting mixture is
approximately isotonic with blood. The B. pertussis Identification
concentration of the final bulk vaccine does not exceed that
Tests A, B, C and D may be omitted if test E is carried out. Test
corresponding to opacity of 20 IU per single human dose. If 2
E may be omitted if tests A, B, C and D are carried out.
or more strains of B. pertussis are used, the composition of
consecutive lots of the final bulk vaccine shall be consistent A. Diphtheria toxoid. Dissolve sufficient sodium citrate in
with respect to the proportion of each strain as measured in the vaccine under examination to give a 10 per cent w/v
opacity units. The final bulk is filled separately. Suitable concentration. Maintain at 37o for about 16 hours and
antimicrobial preservatives may be added. The final bulk of centrifuge. Reserve the residue for test C. The clear
the haemophilus component is prepared by dilution of the supernatant reacts with a suitable diphtheria antitoxin and
bulk conjugate to the final concentration with a suitable diluent. yields a precipitate.
A stabilizer may be added. B. Tetanus toxoid. The clear supernatant obtained in test A
Only a final bulk vaccine that complies with the following reacts with a suitable tetanus antitoxin and yields a precipitate.
requirements may be used in the preparation of the final lot. C. Pertussis component. To a suspension of the residue
Antimicrobial preservative. Where applicable, determine the obtained in test A in saline solution add a suitable Bordetella
amount of antimicrobial preservative by a suitable chemical pertussis antiserum; agglutination indicates presence of
method. The amount is not less than 85.0 per cent and not pertussis component.
greater than 115.0 percent of the intended content. D. PRP. The suspension of the residue obtained in test A
Sterility (2.2.11). Carry out test for sterility using 10 ml of gives a positive reaction when tested by a suitable
bulk for each sterility medium. immunochemical method for PRP.
E. The vaccine confers an active immunity in mice and guinea
FINAL LOT pigs when administered as directed in the test for Potency.
Where the haemophilus component is in a separate container, Tests
the final bulk of the haemophilus component is freeze-dried.
If the product is presented with the haemophilus component
Only a final lot that is satisfactory with respect to the test for in a separate container; the tests for specific toxicity of
osmolality and with respect to each of the requirements given diphtheria toxoid, tetanus toxoid and pertussis component,
below under Identification, Tests and Assay may be released aluminium, free formaldehyde, antimicrobial preservative
for use. Provided the tests specific toxicity of diphtheria toxoid, and sterility are carried out on the container with the
tetanus toxoid and pertussis component and antimicrobial diphtheria, tetanus and pertussis components; the tests for
preservative and the assays for the diphtheria, tetanus and PRP content, water (where applicable), sterility and
pertussis components have been carried out with satisfactory pyrogens are carried out on the container with the
results on the final bulk vaccine, they may be omitted on the haemophilus component.

38
IP 2007 DIPHTHERIA VACCINE (ADSORBED)

If the haemophilus component is freeze-dried, some tests may Complies with the test as stated under Diphtheria and Tetanus
be carried out on the freeze-dried product rather than on the Vaccine (Adsorbed).
bulk conjugate where the freeze-drying process may affect
the component under test. Tetanus toxoid (adsorbed)

PRP. Not less than 80.0 per cent of the amount of PRP stated Complies with the test as stated under assay of Tetanus
on the label. PRP is determined either by assay of ribose (2.7.1) Vaccine (Adsorbed).
or phosphorus (2.7.1), by an immunochemical method (2.2.14) Pertussis vaccine
or by anion exchange liquid chromatography with pulsed
amperometric detection. Complies with the test as stated under Diphtheria, Tetanus
and Pertussis Vaccine (Adsorbed).
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
if aluminium hydroxide or hydrated aluminium phosphate is Storage. When stored under the prescribed conditions the
used as the adsorbent. vaccine may be expected to retain potency for not less than 2
years from the date on which the potency test for the pertussis
Free formaldehyde (2.3.20). Maximum 0.2 g/l. component was started.
Antimicrobial preservative. Where applicable, determine the Labelling. The label states (1) the human dose (ml); (2)
amount of antimicrobial preservative by a suitable chemical Diphtheria and Tetanus components (a) in case done by
method. The content is not less than 85.0 per cent and not challenge method the minimum number of international units
greater than 115.0 per cent of the intended amount. (as applicable for each component) per single human dose;
Sterility (2.2.11). Complies with the test for sterility. (b) in case done by antibody induction method, the minimum
Lf units per single human dose; (3) pertussis component – IU
Abnormal toxicity (2.2.1). Each final lot shall be tested for
or IOU per single human dose; (4) haemophilus conjugate
abnormal toxicity by injecting intraperitoneally one human
component - g PRP per single human dose; (5) the type and
dose , but not more than 0.25 ml into each of the five mice
nominal amount of carrier protein per single human dose; (6)
weighing between 17 to 22 g and at least one human dose but
the name and amount of adsorbent and added preservative;
not more than 1.0 ml into each of the two guinea pigs weighing
(7) that the vaccine must be shaken before use; (9) that the
between 250 and 350 g. The preparation passes the test if
vaccine is not to be frozen.
none of the animals dies or shows signs of ill health in 7 days
following the injection . If one of the animal dies or shows the
signs of ill health, repeat the test . The preparation passes the
test if none of the animals in the second group dies or shows Diphtheria Vaccine (Adsorbed)
signs of ill health in the time interval specified.
Diphtheria Vaccine (Adsorbed) is a preparation of diphtheria
Pyrogens (2.2.8). This test is carried out for Haemophilus formol toxoid with a mineral adsorbent. The formol toxoid is
influenzae type b vaccine only if Haemophilus influenzae prepared from the toxin produced by the growth of
type b vaccine is presented as separate lyophilized vial. The Corynebacterium diphtheriae.
vaccine complies with the test for pyrogens. Inject per kg of
the rabbit’s mass a quantity of the vaccine equivalent to: 1 mg Production
of PRP for a vaccine with diphtheria toxoid or CRM 197
diphtheria toxoid as carrier; 0.1 mg of PRP for a vaccine with General provisions
tetanus toxoid as carrier protein; 0.025 mg of PRP for vaccine
with OMP as carrier. The maximum number of Lf units per single human dose
of diphtheria vaccine (adsorbed) is 30.
Specific toxicity
Diphtheria and tetanus components Bulk purified toxoid
Complies with the test as stated under Diphtheria and Tetanus For the production of diphtheria toxin, from which toxoid is
Vaccine (Adsorbed). prepared, seed cultures are managed in a defined seed-lot
system in which toxinogenicity is conserved and, where
Pertussis component
necessary, restored by deliberate reselection. A highly
Complies with the test as stated under Diphtheria, Tetanus toxinogenic strain of Corynebacterium diphtheriae with
and Pertussis Vaccine (Adsorbed). known origin and history is grown in a suitable liquid medium.
Assay At the end of cultivation, the purity of each culture is tested
and contaminated cultures are discarded. Toxin-containing
Diphtheria toxoid (adsorbed) culture medium is separated aseptically from the bacterial mass

39
DIPHTHERIA VACCINE (ADSORBED) IP 2007

as soon as possible. The toxin content (Lf per ml) is checked preservatives and detoxifying agents should be determined
to monitor consistency of production. Single harvests may be to be below the concentration toxic to Vero cells. Non-specific
pooled to prepare the bulk purified toxoid. The toxin is purified toxicity may be eliminated by dialysis.
to remove components likely to cause adverse reactions in Use freshly trypsinised Vero cells at a suitable concentration,
humans. The purified toxin is detoxified with formaldehyde by for example 2.5 × 105 per ml and a reference diphtheria toxin
a method that avoids destruction of the immunogenic potency diluted in 100 Lf per ml diphtheria toxoid. A suitable reference
of the toxoid and reversion of the toxoid to toxin, particularly diphtheria toxin will contain either not less than 100 LD50/ml
on exposure to heat. Alternatively, purification may be carried or 67 to 133 lr/100 in 1 Lf and 25,000 to 50,000 minimal reacting
out after detoxification. doses for guinea-pig skin in 1 Lf (diphtheria toxin RP is
Only bulk purified toxoid that complies with the following suitable for use as the reference toxin). Dilute the toxin in 100
requirements may be used in the preparation of the final bulk Lf/ml diphtheria toxoid to a suitable concentration, for example
vaccine. 2 × 10-4 Lf per ml. Prepare serial twofold dilutions of the diluted
diphtheria toxin and use undiluted test samples (50 µl per
Sterility (2.2.11). Carry out test for sterility using 10 ml of
well). Distribute them in the wells of a sterile tissue culture
bulk for each sterility medium.
plate containing a medium suitable for Vero cells. To ascertain
Absence of toxin and irreversibility of toxoid that any cytotoxic effect noted is specific to diphtheria toxin,
prepare in parallel dilutions where the toxin is neutralised by a
Inject subcutaneously into each of 5 guinea-pigs at least 500 Lf
suitable concentration of diphtheria antitoxin, for example
of the non-incubated bulk purified toxoid in a volume of
100 IU/ml. Include control wells without toxoid or toxin and
1 ml, using the same buffer solution as for the final vaccine,
with non-toxic toxoid at 100 Lf per ml on each plate to verify
without adsorbent. Animals that die shall be autopsied and
normal cell growth. Add cell suspension to each well, seal the
examined for symptoms of diphtheria intoxication (red
plates and incubate at 37° for 5 to 6 days. Cytotoxic effect is
adrenals). The bulk purified toxoid shall pass the test if no
judged to be present where there is complete metabolic
guinea-pig shows symptoms of specific intoxication within
inhibition of the Vero cells, indicated by the pH indicator of
six weeks of injection and if at least 80 per cent of the animals
the medium. Confirm cytopathic effect by microscopic
survive the test period. The guinea-pigs shall not have been
examination or suitable staining such as MTT dye. The test is
used previously for experimental purposes.
invalid if 5 × 10-5 Lf per ml of reference diphtheria toxin in 100
Alternatively, a cell-culture test system may be used; in this Lf per ml toxoid has no cytotoxic effect on Vero cells or if the
case, the sensitivity of the test shall have been demonstrated cytotoxic effect of this amount of toxin is not neutralised in
to be not less than that of the guinea-pig test, and the test the wells containing diphtheria antitoxin. The bulk purified
procedures shall be approved by the National Regulatory toxoid complies with the test if no toxicity neutralisable by
Authority. antitoxin is found in either sample.
Each bulk purified toxoid shall be tested to ensure that Antigenic purity. Not less than 1,500 Lf per mg of protein
reversion to toxicity cannot take place on storage. The bulk nitrogen.
purified toxoid shall be diluted in order to obtain the same
concentration and chemical environment as those present in FINAL BULK VACCINE
the final bulk vaccine, except for the presence of adjuvant.
The final bulk vaccine is prepared by adsorption of a suitable
To determine whether reversion has occurred, diluted toxoids quantity of bulk purified toxoid onto a mineral carrier such as
that have been stored at 37° for six weeks shall be tested. The hydrated aluminium phosphate or aluminium hydroxide; the
test employed shall be approved by the National Regulatory resulting mixture is approximately isotonic with blood. Suitable
Authority and should be sufficiently sensitive to detect very antimicrobial preservatives may be added. Certain
small amounts of toxin. No toxicity shall be detected. antimicrobial preservatives, particularly those of the phenolic
Intradermal tests in guinea-pigs and cell-culture tests both type, adversely affect the antigenic activity and must not be
are considered to be suitable. used.

Cell culture method Only a final bulk vaccine that complies with the following
requirements may be used in the preparation of the final lot.
Using the same buffer solution as for the final vaccine, without
adsorbent, prepare a solution of bulk purified toxoid at 100 Lf Identification
per ml. Divide the solution into 2 equal parts. Maintain 1 part
at 5° ± 3° and the other at 37° for 6 weeks. Carry out a test in Dissolve sufficient sodium citrate in the vaccine under
Vero cells for active diphtheria toxin using 50 µl per well of examination to give a 10 per cent w/v concentration. Maintain
both samples. The sample should not contain antimicrobial at 37o for about 16 hours and centrifuge. The clear supernatant

40
IP 2007

reacts with a suitable diphtheria antitoxin and yields a toxin preparation has been shown to be stable, it is not
precipitate. necessary to verify the activity for every assay.
pH (2.4.24). 6.0 to 7.0. Preparation of the challenge toxin solutions. Immediately prior
to use, dilute the challenge toxin with a suitable diluent to
Specific toxicity. Use 5 normal, healthy guinea-pigs weighing
obtain a challenge toxin solution containing about 512x10-4 Lf
between 250 and 350 g, which have been maintained for at
in 0.2 ml. Dilute a portion of this challenge toxin solution to
least 1 week on a uniform, unrestricted diet, and have not
give a series of five 4-fold dilutions.
been previously treated with any material that will interfere
with the test. Weigh the animals separately and record their Determination of potency of the vaccine. Prepare in saline
weights. Inject subcutaneously into each animal 5 times the solution dilutions of the vaccine under examination and of
dose stated on the label. Weigh all the animals at weekly the Standard preparation such that, for each, the dilutions
intervals for 6 weeks. None of the animals shows any form a series differing by not more than 2.5 fold steps and in
symptoms of diphtheria or tetanus toxaemia or dies from which the dilutions of intermediate concentration, when
diphtheria within 42 days or loses weight at the end of the injected subcutaneously in 1.0 ml volumes into guinea-pigs,
test. If more than one animal dies from non-specific causes or result in an intradermal score of approximately three when the
loses weight, repeat the test. If an animal dies or loses weight animals are challenged. Allocate the dilutions, one to each of
in the second test, the vaccine fails the test. the groups of guinea-pigs, and inject subcutaneously 1.0 ml
of each dilution into each guinea-pig in the group to which
Potency. Determine by any of the methods of biological assay
that dilution is allocated. After 28 days shave both flanks of
of Adsorbed Diphtheria Vaccine described.
each guinea-pig and inject each animal intradermally with 0.2
Biological assay of adsorbed diphtheria vaccine ml of the challenge toxin solution and with 0.2 ml of each of
(a) Intradermal challenge method the five dilutions thereof in such a way as to minimize
interference between adjacent sites. If necessary, inject the
The potency of adsorbed diphtheria vaccine is determined by unvaccinated control guinea-pigs with dilutions containing
comparing the dose necessary to protect guinea-pigs against 8x10-5, 4x10-5, 2x10-5, 1x10-5 and 5x10-6 Lf of the challenge toxin.
the erythrogenic effects of a range of intradermal injections of Examine all the injection sites 48 hours after injection of the
diphtheria toxin with the dose of the Standard preparation of challenge toxin and record the incidence of specific diphtheria
adsorbed diphtheria toxoid necessary to give the same erythema. Record also the number of sites free from such
protection. For this comparison, the Standard preparation of reactions as the intradermal challenge score. Tabulate the
adsorbed diphtheria toxoid and a suitable preparation of intradermal challenge scores for all the animals receiving the
diphtheria toxin, for use as a challenge toxin, are required. same dilution of vaccine and use those data with a suitable
transformation, such as (score)2 or arcsin [(score/6)2], to obtain
Standard preparation
an estimate of the relative potency for each of the test
The Standard preparation is International standard of preparations by parallel-line quantitative analysis.
Diphtheria toxoid, adsorbed, or another suitable preparation
The test is not valid unless (a) for both the preparation under
the potency of which has been determined in relation to the
examination and the Standard preparation, the mean score
International Standard.
obtained at the lowest dose level is more than three; (b) if
Suggested method applicable, the toxin dilution that contains 4x10-5 Lf gives a
Test animals. Use white guinea-pigs, weighing between 250 positive erythema in at least 80.0 per cent of the control guinea-
and 350 g, from the same stock. Distribute the guinea-pigs pigs and the dilution that contains 2x10-5 Lf gives no reaction
into no fewer than six equal groups; use groups containing a in at least 80 per cent of the guinea-pigs (if these criteria are
number of animals sufficient to obtain results that fulfill the not met a different toxin has to be selected); (c) the fiducial
requirements for a valid Assay prescribed below. The guinea- limits of the assay fall between 50.0 and 200.0 per cent of the
pigs are all of the same sex or the males and females are estimated potency; (d) the statistical analysis shows no
distributed equally among the groups. If the challenge toxin deviation from linearity and parallelism. The test may be
to be used has not been shown to be stable or has not been repeated but when more than one test is performed the results
adequately standardized, include five guinea-pigs as of all valid tests must be combined in the estimate of potency.
unvaccinated controls. The lower fiducial limit of error of the estimated potency is not
Selection of the challenge toxin. Select a preparation of less than 30 Units per dose.
diphtheria toxin containing 67 to 133 Limes reactionis/100 (Lr/
(b) Lethal challenge method
100) in Limes flocculationis (Lf) and 25,000 to 50,000 minimal
reacting doses for guinea-pig skin in 1 Lf. If the challenge Test animals. Use healthy, white or light-coloured guinea-

41
DIPHTHERIA VACCINE (ADSORBED) IP 2007

pigs from the same stock, weighing between 250 and 350 g. limits of the 95.0 per cent confidence interval should be within
Distribute them into six groups of sixteen; and four groups of 50 to 200 of the estimated potency.
four. The guinea-pigs should all be of the same sex or the
males and females should be distributed equally between the (c) Antibody induction method
six groups of sixteen. Inject subcutaneously on each of two occasions separated
Challenge toxin. Select a preparation of diphtheria toxin by an interval of not more than 4 weeks, one-fiftieth of the
containing not less than 100 LD50 in 1.0 ml. stated human dose diluted to 1 ml with saline solution, into
each of 10 normal, healthy guinea-pigs weighing between 250
Preparation of the challenge toxin solutions. Immediately prior and 350 g. Not earlier than 2 weeks and not later than 3 weeks
to use, prepare from the challenge toxin by dilution in after the second injection, collect the serum from each animal
phosphate buffered saline pH 7.4, or normal saline a challenge and determine the antitoxin content of the serum of each animal,
toxin containing approximately 100 LD50 in 1.0 ml. Dilute as described under Diphtheria Antitoxin or any other method
portions of this challenge toxin solution to 2LD50, 1 LD50 and approved by National Regulatory Authority. The geometric
½ LD50 in the same solution mean of the antitoxin contents shall be not less than 2.0 Units
Determination of potency of the vaccine. Prepare in saline per ml with reference to the Diphtheria antitoxin standard.
solution three dilutions of the vaccine under examination and
three dilutions of the Standard preparation such that for each, (d) Any other validated serological assay in guinea pigs or
the dilutions form a series differing by not more than 2.5 fold mice as approved by National Regulatory Authority
steps and in which the dilutions of intermediate concentration, Antimicrobial preservative. Where applicable,
when injected subcutaneously in 1.0 ml volumes into guinea- determine the amount of antimicrobial preservative by
pigs, protect approximately 50 per cent of the animals from the a suitable chemical method. The amount is not less than
lethal effects of the subcutaneous injection of the quantity of 85.0 per cent and not greater than 115.0 per cent of the
diphtheria toxin prescribed for this test. Allocate the six intended amount.
dilutions, one to each of the six groups of sixteen guinea-pigs,
and inject subcutaneously 1.0 ml of each dilution into each Sterility (2.2.11). Carry out test for sterility using 10 ml of
guinea-pig in the groups to which that dilution is allocated. bulk for each sterility medium.
After 28 days inject subcutaneously into each animal in the
six groups of sixteen, 1.0 ml of the challenge toxin solution. FINAL LOT
Allocate the challenge toxin solution and the three dilutions The final bulk vaccine is distributed aseptically into sterile,
made from it, one to each of the four groups of four guinea- tamper-proof containers. The containers are closed so as to
pigs and inject subcutaneously 1.0 ml of each toxin solution prevent contamination.
into each guinea-pig in the group to which that solution is
Only a final lot that is satisfactory with respect to each of the
allocated. Examine the guinea pigs twice in a day, remove
requirements given below under, Identification, Tests and
dead animals and kill the animals showing definite signs of
Assay may be released for use. Provided the tests for specific
diphtheria. Count the number of surviving animals 5 days
toxicity, free formaldehyde and antimicrobial preservative and
later and calculate the potency of the vaccine under
the assay have been carried out with satisfactory results on
examination relative to the potency of the Standard preparation
the final bulk vaccine, they may be omitted on the final lot.
on the basis of the number of animals that survive in each of
the six groups of sixteen, using appropriate statistical methods. Identification
The test is not valid unless (a) for the vaccine under Diphtheria toxoid is identified by a suitable immunochemical
examination and the Standard preparation the 50.0 per cent method. The following method, applicable to certain vaccines,
protective doses lie between the largest and smallest doses of is given as an example. Dissolve in the vaccine under
the preparations given to the guinea-pigs; (b) the survivors examination sufficient sodium citrate to give a 10 per cent w/
among the four groups of guinea-pigs injected with the v solution. Maintain at 37° for about 16 hours and centrifuge
challenge toxin and its dilutions indicate that the challenge until a clear supernatant liquid is obtained. The clear
was approximately 100 LD50 and; (c) statistical analysis shows supernatant liquid reacts with a suitable diphtheria antitoxin,
parallelism, linearity and a significant slope of the dose- giving a precipitate.
response lines. The test may be repeated any number of times
but when more than one test is performed the results of all Tests
valid tests must be combined in the estimate of potency.
Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
If the lower limit of 95.0 per cent confidence interval of estimated if aluminium hydroxide or hydrated aluminium phosphate is
potency is less than 30 IU per single human dose then the used as the absorbent.

42
IP 2007 HAEMOPHILUS TYPE B CONJUGATE

Free formaldehyde (2.3.20). Maximum 0.2 g/l. immunogenicity test on mice. Taking account of the results of
Antimicrobial preservative. Where applicable, determine the the stability testing, release requirements are set for these
amount of antimicrobial preservative by a suitable chemical indicator tests to ensure that the vaccine will be satisfactory
method. The content is not less than 85.0 per cent and not at the end of the period of validity.
greater than 115.0 per cent of the intended amount. SEED LOT
Sterility (2.2.11). Complies with the test for sterility.
The strain of H. influenzae type b used in preparing
Abnormal toxicity (2.2.1). Complies with the test for abnormal Haemophilus type b conjugate vaccine shall be identified by
toxicity. a record of its history, including the source from which it was
pH (2.4.24). 6.0 to 7.0. obtained and the tests made to determine the characteristics
of the strain. The strain shall have been shown to be capable
Assay of producing Type b polysaccharide.

Carry out one of the prescribed methods for the assay of The production of PRP and of the carrier protein is based on
Diphtheria Vaccine (Adsorbed). defined seed lot systems. Master seed lot and working seed
lot shall be properly characterized and defined. Cultures
The lower confidence limit (P = 0.95) of the estimated potency derived from the working seed shall have the same
is not less than 30 IU per single human dose. characteristics as of the master seed lot. The sample of culture
Labelling. The label states (1) the human dose; (2) the minimum of single harvests taken before killing shall be tested for
Lf units per single human dose or the minimum International contamination by examination of Gram-stained smears and by
Units per single human dose if potency test done by challenge inoculation on suitable media.
method; (3) the name and the amount of the adsorbent and H. Influenzae Type b Polysaccharide (PRP)
preservative; (4) that the vaccine must be shaken before use;
(5) that the vaccine is not to be frozen. H. influenzae Type b is grown in a liquid medium that does
not contain high-molecular-weight polysaccharides; if any
ingredient of the medium contains blood-group substances,
the process shall be validated to demonstrate that after the
Haemophilus Type b Conjugate purification step they are no longer detectable. The culture
Vaccine may be inactivated. PRP is separated from the culture liquid
and purified by a suitable method. Volatile matter, including
Haemophilus Type b Conjugate Vaccine is a liquid or freeze- water, In the purified polysaccharide is determined by methods
dried preparation of a polysaccharide, derived from a suitable such as thermogravimetry, Karl Fischer or any other suitable
strain of Haemophilus influenzae type b, covalently bound method. All chemical analysis shall be based on the dry weight
to a carrier protein. The polysaccharide, polyribosylribitol of the polysaccharide, in its salt form.
phosphate, referred to as PRP, is a linear copolymer composed
Only those pools of PRP that comply with the following
of repeated units of 3-β-D-ribofuronosyl-(11)-ribitol-5-
requirements may be used in the preparation of the conjugate.
phosphate [(C10H19O12P)n], with a defined molecular size. The
The partially purified PRP shall be stored frozen at or below
carrier protein, when conjugated to PRP, is capable of inducing
-20°.
a T-cell-dependent B-cell immune response to the
polysaccharide. Identification
Production The PRP is identified by an immunochemical method (2.2.14)
or other suitable method (e.g. 1H or 13C NMR spectroscopy).
General provisions
Molecular size. The percentage of PRP eluted before a given
The production method shall have been shown to yield K0 value or within a range of K0 values, is determined by gel
consistently H. influenzae type b conjugate vaccines of filtration or high performance size-exclusion chromatography
adequate safety and immunogenicity in humans. The (HPSEC) (2.4.16), either alone or in combination with light
production method is validated to demonstrate that the product, scattering and refractive index detectors (e.g. multiple angle
if tested, would comply with the test for safety and efficacy of laser light scattering i.e. MALLS) or any other suitable method.
Vaccines. The stability of the final lot and relevant An acceptable value is established for the particular product
intermediates is evaluated using one or more indicator tests. and each batch of PRP must be shown to comply with this
Such tests may include determination of molecular size, limit. Limits for currently approved products, using the
determination of free PRP in the conjugate and the indicated stationary phases, are shown for information in

43
HAEMOPHILUS TYPE B CONJUGATE IP 2007

Table 1 - Specifications for different components of Haemophilus Type b Conjugate Vaccine.

Polysaccharide Carrier Protein Conjugation


Type of PRP Nominal Type Purity Nominal Coupling Procedure
amount amount method
per dose per dose
Polysaccharide 25 µg Diphtheria >1500 Lf per mg 18 µg Cyanogen Activated diphtheria
(size reduced) toxoid of protein nitrogen bromide toxoid (D-AH+),
K0 60.0 per cent: activation cyanogen bromide
0.6-0.7 of PRP activated PRP
Polysaccharide: 10 µg Tetanus >1500 Lf per mg 20 µg Carbodiimide- ADH-activated PRP
PRP ≥ 50.0 per toxoid of protein nitrogen mediated (PRP-cov.-AH) +
cent ≤ K0: 0.30 coupling tetanus toxoid +
EDAC
Polysaccharide 10 µg CRM 197 >90.0 per cent 25µg Reductive Direct coupling of
(size reduced) diphtheria diphtheria amination one- PRP to CRM 197
Dp=15-35 or 10-35 protein protein (step method) or (cyanoboro-hydride
N-hydroxy- activated)
succinimide
activation
Polysaccharide 15 µg Meningococcal Outer membrane 125 or Thioether PRP activation by
(size-reduced) group B outer protein vesicles 250 µg bond CDI PRP-IM +
K0 0.3 – 0.6 membrane <8.0 per cent of BuA2 + BrAc =
protein (OMP) lipopolysaccharide PRP-BuA2-BrAc +
thioactivated OMP
Abbreviations:
ADH = adipic acid dihydrazide Dp = degree of polymerization
BrAc = bromoacetyl chloride EDAC = l-ethyl-3-(3-dimethylaminopropyl) carbodimide
BuA2 = butane-1, 4-diamide IM = imidazolium
CDl = carbonyldi-imidazole Mw = weight-average molecular weight.

Table 2 - Requirements on bulk conjugate

Test Protein Carrier


Specifications Diphtheria toxoid Tetanus toxoid CRM 197 OMP
Free Polysaccharide (PRP) <37.0 per cent <20.0 per cent <25.0 per cent <15.0 per cent
Unbound protein <5.0 per cent, <1.0 per cent, <1.0 per cent or Not applicable
where applicable where applicable <2.0 per cent,
depending on the
coupling method
PRP to protein ratio 1.25-1.75 0.30-0.55 0.3-0.7 0.05-0.1
Molecular size (K0): Cross- 95.0 per cent <0.75 60.0 per cent <0.2 50.0 per cent 85.0 per cent < 0.25
linked agarose for 0.3-0.6
chromatography R
Cross-linked agarose for 0.6-0.7 85.0 per cent <0.5
chromatography R1

44
IP 2007 HAEMOPHILUS TYPE B CONJUGATE

Tables 1 and 2. Where applicable, the molecular-size Diphtheria toxoid. Diphtheria toxoid is produced as stated
distribution is also determined after chemical modification of under Diphtheria Vaccine (Adsorbed) and complies with the
the polysaccharide. requirements prescribed there for bulk purified toxoid.
A validated determination of the degree of polymerization or Tetanus toxoid. Tetanus toxoid is produced as stated under
of the weight-average molecular weight and the dispersion of Tetanus Vaccine (Adsorbed) and complies with the
molecular masses may be used instead of the determination of requirements prescribed there for bulk purified toxoid except
molecular size distribution. that the antigenic purity is not less than 1500 Lf per mg of
Ribose (2.7.1). Not less than 32.0 per cent, calculated with protein nitrogen.
reference to the dried substance, as estimated by Bial reaction Diphtheria protein CRM 197. Suitable tests are carried out,
for pentose, using D-ribose as a standard or any other suitable for validation or routinely, to demonstrate that the product is
assay. non-toxic. The protein obtained contains not less than 90.0
per cent of diphtheria CRM 197 protein, when prepared by
Phosphorus (2.7.1). 6.8 per cent to 9.0 per cent, calculated
liquid chromatography (2.4.14) or any other suitable method.
with reference to the dried substance.
The carrier protein shall be characterized by a suitable chemical
Protein (2.3.49). Not more than 1.0 per cent, calculated with or physicochemical method like SDS-PAGE, HPLC, isoelectric
reference to the dried substance. Use sufficient PRP to allow focusing, amino acid sequencing, circular dichroism,
detection of 1 per cent of protein (e.g. a minimum of 1 mg of fluorescence spectroscopy, peptide mapping or mass
PRP). spectroscopy, as appropriate.
Nucleic acid (2.7.1). Not more than 1.0 per cent, calculated OMP (Meningococcal group B outer membrane protein
with reference to the dried substance by spectroscopy or any complex)
other suitable method.
OMP complex of Neisseria meningitidis complies with the
Bacterial endotoxins (2.2.3 ). Not more than 25 IU of endotoxin following requirements for lipopolysaccharide and pyrogens.
per microgram of PRP.
Lipopolysaccharide. Not more than 8.0 per cent of
Residual reagents. Where applicable, tests are carried out to lipopolysaccharide, determined by a suitable method.
determine residues of reagents used during inactivation and
purification. An acceptable value for each reagent is Pyrogens (2.2.8). Inject into each rabbit 0.25 µg of OMP per kg
established for the particular product and each batch of PRP body weight, for determining the pyrogenic effect.
must be shown to comply with this limit. Where validation Bulk conjugate
studies have demonstrated removal of a residual reagent, the
test on PRP may be omitted. PRP is chemically modified to enable conjugation; it is usually
partly depolymerised either before or during this procedure.
Carrier protein Reactive functional groups or spacers may be introduced into
the carrier protein or PRP prior to conjugation. The conjugate
The carrier protein is chosen in a way so that when the PRP is
is obtained by the covalent binding of PRP and carrier protein.
conjugated it is able to induce a T-cell-dependent immune
Where applicable, unreacted but potentially reactogenic
response. Currently approved carrier proteins and coupling
functional groups are made unreactive by means of capping
methods are listed for information in Table 1. The carrier
agents; the conjugate is purified to remove reagents. Where
proteins are produced by culture of suitable microorganisms;
validation studies have demonstrated removal of a residual
the bacterial purity of the culture is verified; the culture may
reagent (eg. CN, Br etc.), the test on bulk conjugate may be
be inactivated; the carrier protein is purified by a suitable
omitted.
method.
Only a bulk conjugate that complies with the following
Only a carrier protein that complies with the following
requirements may be used in preparation of the final bulk
requirements may be used in preparation of the conjugate.
vaccine. For each test and for each particular product, limits
Identification of acceptance are established and each batch of conjugate
must be shown to comply with these limits. Limits applied to
The carrier protein is identified by a suitable immunochemical currently approved products for some of these tests are listed
method (2.2.14). for information in Table 2.
Sterility (2.2.11). Carry out the test for sterility using for each PRP. The PRP content is determined by assay of phosphorus
medium 10 ml or the equivalent of one hundred doses, (2.7.1) or by assay of ribose (2.7.1) or by an immunochemical
whichever is less. method (2.2.14) or by any suitable method.

45
HEPATITIS A (INACTIVATED) AND HEPATITIS B (rDNA) VACCINE (ADSORBED) IP 2007

Protein (2.7.1). The protein content is determined by a suitable Identification


chemical method.
The vaccine is identified by a suitable immunochemical method
PRP to protein ratio. Determine the ratio by calculation. (2.2.14) for PRP or the assay serves also to identify the vaccine.
Molecular size. Molecular-size distribution is determined by
gel filtration or size-exclusion chromatography (2.4.16), using Tests
a gel matrix, appropriate to the expected size of the conjugate. Sterility (2.2.11). Complies with the test for sterility.
Free PRP. Unbound PRP is determined after removal of the Abnormal toxicity (2.2.1). Complies with the test for abnormal
conjugate, for example by size-exclusion or hydrophobic toxicity.
chromatography (2.4.16), ultra-filtration or other validated
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
methods.
per kg of the rabbit’s mass a quantity of the vaccine equivalent
Free carrier protein. Free carrier protein is determined by a to 1 µg of PRP for a vaccine with diphtheria toxoid or CRM 197
suitable method (which may include deriving the content by diphtheria protein as carrier; 0.1 µg of PRP for a vaccine with
calculation from the results of other tests). The amount is tetanus toxoid as carrier; 0.025 µg of PRP for a vaccine with
within the limits approved for the particular product. OMP as carrier.
Unreacted functional groups. No unreacted functional groups pH (2.4.24). The pH of the vaccine, reconstituted if necessary,
are detectable in the bulk conjugate unless process validation is within the range approved for the product (6.5 to 7.5).
has shown that unreacted functional groups detectable at
Aluminium (2.3.9). When aluminium hydroxide or hydrated
this stage are removed during the subsequent manufacturing
aluminium phosphate is used as the adsorbent,
process (for example, owing to short half-life).
Antimicrobial preservative. Where applicable, determine the
Residual reagents. Removal of residual reagents such as
amount of antimicrobial preservative by a suitable chemical
cyanide, EDAC (ethyldimethylaminopropylcarbodimide) and
method. The content is not less than 85.0 per cent and not
phenol is confirmed by suitable tests or by validation of the
greater than 115.0 per cent of the intended amount.
purification process.
Water (2.3.43). Not more than 3.0 per cent.
Sterility (2.2.11). Carry out the tests for sterility using for
each medium 10 ml or the equivalent of one hundred doses, PRP. Not less than 80.0 per cent of the amount of PRP stated
whichever is less. on the label as determined by ribose assay (2.7.1) or by
phosphorus assay (2.7.1) or by an immunochemical method
FINAL BULK VACCINE (2.2.14) or by any other suitable method like colorimetery or
by anion exchange liquid chromatography (2.4.14) with pulsed
An adjuvant, an antimicrobial preservative and a stabilizer amperometric detection.
may be added to the bulk conjugate before dilution to the final
concentration with a suitable diluent. Free PRP. Unbound PRP is determined after removal of the
conjugate for example by size-exclusion or hydrophobic
Only a final bulk vaccine that complies with the following chromatography (2.4.16), ultra filtration or other validated
requirements may be used in preparation of the final lot. methods.
Antimicrobial preservative. Where applicable, determine the Labelling. The label states (1) the number of micrograms of
amount of antimicrobial preservative by a suitable chemical PRP per human dose; (2) the type and nominal amount of
method. The content is not less than 85.0 per cent and not carrier protein per single human dose; (3) for vaccine contained
greater than 115.0 per cent of the intended amount. in single-dose containers where the space is too small to
Sterility (2.2.11). Carry out test for sterility using 10 ml of accommodate the full name of the vaccine the abbreviation
bulk for each sterility medium. ‘Hib’ may be used in the label on the container provided that
the same code is also stated in the label on the package.
FINAL LOT
The final lots are filled in suitable containers, under stringent
aseptic conditions. Hepatitis A (Inactivated) and Hepatitis
Only a final lot that is satisfactory with respect to each of the B (rDNA) Vaccine (Adsorbed)
requirements given under Identification, Tests and Assay may
be released for use. Provided the test for antimicrobial Hepatitis A (Inactivated) and Hepatitis B (rDNA) Vaccine
preservative has been carried out on the final bulk vaccine, it (Adsorbed) is a suspension consisting of a suitable strain of
may be omitted on the final lot. hepatitis A virus, grown in cell cultures and inactivated by a

46
IP 2007 HEPATITIS B (rDNA) VACCINE

validated method, and of hepatitis B surface antigen (HBsAg), carried out with satisfactory results on the final bulk vaccine,
a component protein of hepatitis B virus obtained by it may be omitted on the final lot.
recombinant DNA technology; the antigens are adsorbed on
Identification
a mineral carrier such as aluminium hydroxide or hydrated
aluminium phosphate. The vaccine is shown to contain hepatitis A virus antigen and
hepatitis B surface antigen by suitable immunochemical
Production methods (2.2.14), using specific antibodies or by the mouse
General provisions immunogenicity tests described under assay.

The two components are prepared as described in the Tests


monographs on Hepatitis A Vaccine (Inactivated, Adsorbed)
Aluminium (2.3.9). Maximum 1.25 mg per single human dose
and Hepatitis B Vaccine (rDNA) and comply with the if aluminium hydroxide or hydrated aluminium phosphate is
requirements prescribed therein. The production method is used as the adsorbent.
validated to demonstrate that the product, if tested, would
comply with the test for abnormal toxicity for antisera and Free formaldehyde (2.3.20). Maximum 0.2 g/l.
vaccines. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Reference preparation
method. The content is not less than 85.0 per cent and not
The reference preparation is part of a representative batch greater than 115.0 per cent of the intended amount.
shown to be at least as immunogenic in animals as a batch
Sterility (2.2.11). Complies with the test for sterility.
that, in clinical studies in young, healthy adults, produces not
less than 95.0 per cent seroconversion, corresponding to a Bacterial endotoxins (2.2.3). Less than 2 IU per human dose.
level of neutralizing antibody recognized to be protective, Assay
after a full-course primary immunization. For hepatitis A, an
antibody level not less than 20 mIU/ml determined by enzyme- Hepatitis A component
linked immunosorbent assay is recognized as being protective. Complies with the assay as stated under Inactivated Hepatitis
For hepatitis B, antibody level not less than 10 mIU/ml against A Vaccine (Adsorbed).
HBsAg is recognized as being protective.
Hepatitis B component
FINAL BULK VACCINE
Complies with the assay as stated under Hepatitis B Vaccine
The final bulk vaccine is prepared from one or more inactivated (rDNA).
harvests of hepatitis A virus and one or more batches of Labelling. The label states (1) the amount of hepatitis A virus
purified antigen of Hepatitis B (rDNA). antigen and hepatitis B surface antigen per container; (2) the
Only a final bulk vaccine that complies with the following type of cells used for production of the vaccine; (3) the name
requirements may be used in the preparation of the final lot. and amount of the adsorbent used; (4) that the vaccine must
be shaken before use; (5) that the vaccine must not be frozen.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount. Hepatitis B Vaccine (rDNA)
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk Hepatitis B Vaccine (rDNA) is a non-infectious preparation
for each sterility medium. containing the purified major surface antigen of Hepatitis B
FINAL LOT virus (HBsAg). This preparation is white or almost white
translucent liquid in which the mineral carrier tends to settle
Only a final lot that complies with each of the requirements down slowly on keeping but is free from foreign particles/
given below under Identification, Tests and Assay may be floccules.
released for use. Provided that the tests for free formaldehyde
(where applicable) and antimicrobial preservative content Production
(where applicable) have been carried out on the final bulk
vaccine with satisfactory results, they may be omitted on the General provisions
final lot. If the assay of the hepatitis A and/or the hepatitis B The antigen is manufactured by recombinant DNA technology
component is carried out in vivo, then provided it has been by culturing genetically engineered yeast cells or other suitable

47
HEPATITIS B (rDNA) VACCINE IP 2007

cell lines which carry the gene that codes for major surface Total protein (2.3.49). The total protein is determined by a
antigen of the Hepatitis-B virus as approved by the competent validated method. The content is within the limits approved
authority. Several physico-chemical steps are employed to for the specific product.
purify the Hepatitis-B surface antigen (HBsAg). The vaccine Antigen content and identification. The quantity and
may contain the product of the S gene (major protein), a specificity of HBsAg is determined in comparison with the
combination of the S gene and pre-S2 gene products (middle International standard for HBsAg subtype ad or an in-house
protein) or a combination of S gene, the pre-S2 gene, and pre- reference, by a suitable immunochemical method such as
S1 gene products (large protein). The purity of the antigen is radioimmunoassay (RIA), enzyme-linked immunosorbent
determined by comparison with a reference preparation using assay (ELISA), immunoblot (preferably using a monoclonal
liquid chromatography or other suitable methods such as SDS- antibody directed against a protective epitope) or single radial
PAGE with any suitable staining method. The purified antigen diffusion. The antigen/protein ratio is within the limits approved
is finally adsorbed on aluminium hydroxide or aluminium for the specific product.
phosphate.
The molecular weight of the major band in a sodium dodecyl
The method used for production of the vaccine must have
sulphate polyacrylamide gel electrophoresis (SDS-PAGE)
been shown to yield a product consistently complying with
under reduced conditions corresponds to the value expected
the requirements for immunogenicity and safety. It must also
from the gene sequence and possible glycosylation.
have been shown to induce specific, protective antibodies in
human beings. Antigenic purity. The purity of the antigen is determined by
comparison with a reference preparation using liquid
The production method must be validated to demonstrate
chromatography or other suitable methods such as SDS-PAGE
that the product if tested, would comply with the tests for
with staining. A suitable method is sensitive enough to detect
safety and efficacy.
a potential contaminant at a concentration of 1.0 per cent of
Reference preparation. A part of a batch shown to be at least total protein. Not less than 95.0 per cent of the total protein
as immunogenic as a batch that was used in clinical studies consists of hepatitis B surface antigen.
and approved by National Regulatory Authority and Composition. The content of proteins, lipids, nucleic acids
determined by any suitable method. For hepatitis B, antibody and carbohydrates is determined.
level not less than 10 mIU/ml against HBsAg is recognized as
being protective.. Host-cell and vector-derived DNA (2.2.15). If mammalian cells
are used for production, not more than 10 pg of DNA in the
Characterisation of the substance quantity of purified antigen equivalent to a single human dose
of vaccine.
Development studies are carried out to characterize the
antigen. The complete protein, lipid and carbohydrate structure Caesium. If a caesium salt is used during production, a test
of the antigen is established. The morphological characteristics for residual caesium is carried out on the purified antigen. The
of the antigen particles are established by electron microscopy. content is within the limits approved for the specific product.
The buoyant density of the antigen particles is determined by Sterility (2.2.11). The purified antigen complies with the test
a physico-chemical method, for example gradient for sterility, carried out using 10 ml for each medium.
centrifugation. The antigenic epitopes are characterized. The
protein fraction of the antigen is characterized in terms of the Additional tests on the purified antigen may be required
primary structure (for example, by determination of the amino- depending on the production method used: for example, a test
acid composition, by partial amino-acid sequence analysis for residual animal serum where mammalian cells are used for
and by peptide mapping). production or tests for residual chemicals used during
extraction and purification.
PROPAGATION AND HARVEST
FINAL BULK VACCINE
Identity, microbial purity, plasmid retention and consistency An antimicrobial preservative and an adjuvant may be
of yield are determined at suitable production stages. If included in the vaccine.
mammalian cells are used, tests for extraneous agents and
mycoplasmas are performed in accordance with tests for Only a final bulk vaccine that complies with the following
extraneous agents in viral vaccines for human use. requirements may be used in the preparation of the final lot.
Antimicrobial preservative. Where applicable, determine the
PURIFIED ANTIGEN
amount of antimicrobial preservative by a suitable chemical
Only a purified antigen that complies with the following method. The content is not less than 85.0 per cent and not
requirements may be used in the preparation of the final bulk. greater than 115.0 per cent of the intended amount.

48
IP 2007 HEPATITIS B (rDNA) VACCINE

Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk the test. Use animals of the same sex in the test. Inject similar
for each sterility medium. groups of animals with the reference preparation of Hepatitis-
FINAL LOT B vaccine (r DNA). One group of control animals remains
unvaccinated but is injected intraperitoneally with the same
Only a final lot that complies with each of the requirements volume of the diluent alone. Anaesthetize and bleed the
given below under Identification, Tests and Assay may be animals 28 to 42 days later, keeping the individual sera separate.
released for use. Provided that the tests for free formaldehyde, Assay the individual sera for specific HBsAg antibody
antimicrobial preservative content and the assay in animals, concentration by a suitable immunochemical method such as
where applicable, have been carried out on the final bulk ELISA or RIA.
vaccine with satisfactory results, they may be omitted on the
Calculate the result of the assay by standard statistical
final lot.
methods (5.7). From the distribution of reaction levels
Identification measured on all the sera in the unvaccinated (control group),
the maximum reaction level that can be expected to occur in an
The assay or, where applicable, the electrophoretic profile, unvaccinated animal for that particular assay is determined.
also serves to identify the vaccine. Any response in the vaccinated animals that exceeds this
level is by definition seroconversion. The percentage of
Tests animals showing seroconversion in each group is transformed
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, (for example, probit transformation) and a parallel line model,
if aluminium hydroxide or hydrated aluminium phosphate is using the log dose response curve, is applied to the data. The
used as the adsorbant. potency of the preparation under examination relative to the
reference preparation is thus established. The test is not valid
Antimicrobial preservative. Where applicable, determine the unless (a) for both the test and reference vaccine, the ED50 lies
amount of antimicrobial preservative by a suitable chemical between the smallest and the largest doses given to animals ;
method. The content is not less than 85.0 per cent and not (b) the statistical analysis shows no deviation from linearity
greater than 115.0 per cent of the intended amount. or parallelism; (c) the fiducial limits of the estimated relative
Sterility (2.2.11). Complies with the test for sterility. potency fall between 33.0 and 300.0 per cent of the estimated
potency.
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
the equivalent of one human dose into each rabbit. Method B (In vitro)
A validated test for bacterial endotoxins may be used instead The potency of the vaccine under examination is determined
of the test for pyrogens. by an in vitro method that has been validated against the
Assay. The vaccine complies with the assay of Hepatitis-B biological test.
Vaccine (rDNA) described below. Enzyme Linked Immunosorbent Assay (ELISA) using
Potency. The upper fiducial limit (P = 0.95) of the estimated monoclonal antibodies specific for protection inducing
relative potency is not less than 1.0. epitopes of HBsAg have been shown to be suitable. Adequate
number of dilutions and replicates of the vaccine under
Determine the potency either in animals (Method A) or by a examination and the reference standard are employed in the
validated in vitro procedure (Method B) described below: assay. The data obtained is analyzed by a parallel-line model
Method A (Biological) and may be suitably transformed for statistical evaluation.
Commercially available kits for measuring HBsAg in vitro may
The potency of the vaccine under examination is determined be used provided they are validated to produce equally precise
in animals by comparing in given conditions its capacity to and accurate results.
induce specific anti-HBsAg antibodies in mice or guinea-pigs
The test is not valid unless (a) the statistical analysis shows
with the same capacity as with the reference standard.
no deviation from linearity or parallelism; (b) the fiducial limits
Inject intraperitoneally not less than three doses of suitable of the estimated relative potency fall between 80.0 and 125.0
dilutions of the vaccine under examination diluted with per cent of the estimated potency.
adjuvant used in the vaccine into groups of a suitable strain
The acceptance criteria are approved for a given reference
of mice, weighing between 15 and 20 g (about 5 weeks old), of
preparation by the National Regulatory Authority in the light
either sex distributed randomly into several groups of mice.
of the validation data.
Healthy guinea pigs weighing between 300 and 350g (about 7
weeks old) that have not been previously treated with any Labelling. The label states (a) the amount of HBsAg per dose;
material that will interfere with the test will also be suitable for (b) the type of cells used for production of the vaccine; (c) the

49
INACTIVATED HEPATITIS A VACCINE (ADSORBED) IP 2007

name and amount of the adjuvant; (d) that the vaccine must Virus concentration. The virus concentration of each master
be shaken before use; (e) that the vaccine must not be frozen. and working seed lot is determined to monitor consistency of
production.
Extraneous agents (2.7.3). The master and working seed lots
Inactivated Hepatitis A Vaccine comply with the requirements for seed lots for virus vaccines.
In addition, if primary monkey cells have been used for
(Adsorbed) isolation of the strain, measures are taken to ensure that the
Hepatitis A Vaccine (Inactivated, Adsorbed) is a liquid strain is not contaminated with simian viruses such as simian
preparation of a suitable strain of hepatitis A virus grown in immunodeficiency virus and filoviruses.
cell cultures, inactivated by a validated method and adsorbed
on a mineral carrier. The vaccine is an opalescent suspension. PROPAGATION AND HARVEST

The vaccine complies with the monograph on Vaccines. All processing of the cell bank and subsequent cell cultures is
done under aseptic conditions in an area where no other cells
Production are being handled. Animal serum (but not human serum) may
Production of the vaccine is based on a virus seed-lot system be used in the cell culture media. Serum and trypsin used in
and a cell-bank system. The production method shall have the preparation of cell suspensions and media are shown to
been shown to yield consistently vaccines that comply with be free from extraneous agents. The cell culture media may
the requirements for immunogenicity, safety and stability. contain a pH indicator, such as phenol red and approved
antibiotics at the lowest effective concentration. Not less than
The production method is validated to demonstrate that the
500 ml of the cell cultures employed for vaccine production is
product, if tested, would comply with the test for safety and
set aside as uninfected cell cultures (control cells). Multiple
efficacy.
harvests from the same production cell culture may be pooled
Unless otherwise justified and authorised, the virus in the and considered as a single harvest.
final vaccine shall not have undergone more passages from
the master seed lot than were used to prepare the vaccine Only a single harvest that complies with the following
shown in clinical studies to be satisfactory with respect to requirements may be used in the preparation of the vaccine.
safety and efficacy. When the determination of the ratio of virus concentration to
antigen content has been carried out on a suitable number of
Reference preparation. A part of a batch shown to be at least single harvests to demonstrate consistency, it may
as immunogenic as a batch that, in clinical studies in young subsequently be omitted as a routine test.
healthy adults, produced not less than 95.0 per cent
seroconversion, corresponding to a level of neutralising Identification
antibody accepted to be protective, after a full-course of
primary immunisation is used as a reference preparation. An The test for antigen content also serves to identify the single
antibody level of 20 mIU /ml determined by enzyme-linked harvest.
immunosorbent assay is recognised as being protective. Sterility (2.2.11). The single harvest complies with the test for
Substrate for virus propagation sterility, carried out using 10 ml for each medium.
The virus is propagated in a human diploid cell line or in a Mycoplasmas (2.7.4). The single harvest complies with the
continuous cell line approved by the competent authority. test for mycoplasmas carried out using 1ml for each medium.
SEED LOT Control cells. The control cells of the production cell culture
The strain of hepatitis A virus used to prepare the master seed comply with a test for identity and the requirements for
lot shall be identified by historical records that include extraneous agents.
information on the origin of the strain and its subsequent Antigen content. Determine the hepatitis A antigen content
manipulation. by a suitable immunochemical method (2.2.14) to monitor
Only a seed lot that complies with the following requirements production consistency; the content is within the limits
may be used for virus propagation. approved for the particular product.
Description. A clear, colourless or light coloured liquid. Ratio of virus concentration to antigen content. The
consistency of the ratio of the concentration of infectious
Identification virus, as determined by a suitable cell culture method, to
Each master and working seed lot is identified as hepatitis A antigen content is established by validation on a suitable
virus using specific antibodies. number of single harvests.

50
IP 2007 INACTIVATED HEPATITIS A VACCINE (ADSORBED)

PURIFICATION AND PURIFIED HARVEST Inactivation. Carry out an amplification test for residual
infectious hepatitis A virus by inoculating a quantity of the
The harvest, which may be a pool of several single harvests,
inactivated harvest equivalent to 5 per cent of the batch or if
is purified by validated methods. If continuous cell lines are
the harvest contains the equivalent of 30,000 doses or more,
used for production, the purification process shall have been
not less than 1,500 doses of vaccine into cell cultures of the
shown to reduce consistently the level of host-cell DNA. Only
same type as those used for production of the vaccine and
a purified harvest that complies with the following requirements
incubating the cells for at least 28 days. Make two passages
may be used in the preparation of the inactivated harvest.
and at the end of incubation carry out a test of suitable
Virus concentration. The concentration of infective virus in sensitivity for residual infectious virus. No evidence of
the purified harvest is determined by a suitable cell culture hepatitis A virus multiplication is found in the samples taken
method to monitor production consistency and as a starting at the end of the inactivation process. Use infective virus
point for monitoring the inactivation curve. inocula concurrently as positive controls to demonstrate
Antigen:total protein ratio. Determine the hepatitis A virus cellular susceptibility and absence of interference.
antigen content by a suitable immunochemical method (2.2.14). Sterility (2.2.11). The inactivated viral harvest complies with
Determine the total protein by a validated method. The ratio the test for sterility, carried out using 10 ml for each medium.
of hepatitis A virus antigen content to total protein content is
Bacterial endotoxins (2.2.3). Not more than 2 IU of endotoxin
within the limits approved for the particular product.
in the equivalent of a single human dose.
Bovine serum albumin. Not more than 50 ng in the equivalent
Antigen content. Determine the hepatitis A virus antigen
of a single human dose, determined by a suitable
content by a suitable immunochemical method (2.2.14).
immunochemical method (2.2.14). Where appropriate in view
of the manufacturing process, other suitable protein markers Residual chemicals. As stated under Purification and Purified
may be used to demonstrate effective purification. Harvest.
Residual host-cell DNA (2.2.15). If a continuous cell line is FINAL BULK VACCINE
used for virus propagation, the content of residual host-cell
DNA, determined using a suitable method is not greater than The final bulk vaccine is prepared from one or more inactivated
100 pg in the equivalent of a single human dose. harvests. Approved adjuvants, stabilisers and antimicrobial
preservatives may be added.
Residual chemicals. If chemical substances are used during
the purification process, tests for these substances are carried Only a final bulk vaccine that complies with the following
out on the purified harvest (or on the inactivated harvest), requirements may be used in the preparation of the final lot.
unless validation of the process has demonstrated total Antimicrobial preservative. Where applicable, determine the
clearance. The concentration must not exceed the limits amount of antimicrobial preservative by a suitable chemical
approved for the particular product. method. The content is not less than 85.0 per cent and not
INACTIVATION AND INACTIVATED HARVEST greater than 115.0 per cent of the intended amount.
Sterility (2.2.11). Carry out test for sterility using 10 ml of
Several purified harvests may be pooled before inactivation.
bulk for each sterility medium.
In order to avoid interference with the inactivation process,
virus aggregation must be prevented or aggregates must be FINAL LOT
removed immediately before and/or during the inactivation
process. The virus suspension is inactivated by a validated The final bulk vaccine is distributed aseptically into sterile
method; the method shall have been shown to be consistently containers. The containers are then closed so as to avoid
capable of inactivating hepatitis A virus without destroying contamination.
the antigenic and immunogenic activity; as part of the Only a final lot that complies with each of the requirements
validation studies, an inactivation curve is plotted representing given below under Identification, Tests and Assay may be
residual live virus concentration measured on at least three released for use. Provided that the tests for free formaldehyde
occasions (for example, on days 0, 1 and 2 of the inactivation (where applicable) and antimicrobial preservative content
process). If formaldehyde is used for inactivation, the presence (where applicable) and the assay have been carried out on the
of excess free formaldehyde is verified at the end of the final bulk vaccine with satisfactory results, these tests may be
inactivation process. omitted on the final lot. If the assay is carried out using mice
Only an inactivated harvest that complies with the following or other animals, then provided it has been carried with
requirements may be used in the preparation of the final bulk satisfactory results on the final bulk vaccine, it may be omitted
vaccine. on the final lot.

51
INACTIVATED HEPATITIS B VACCINE IP 2007

Identification antibodies against hepatitis A virus by a suitable


immunochemical method (2.2.14).
The vaccine is shown to contain hepatitis A virus antigen by
a suitable immunochemical method using specific antibodies Calculations. Carry out the calculations by the usual statistical
or by the mouse immunogenicity test described under Assay. methods (5.7) for an assay with a quantal response.
From the distribution of reaction levels measured on all the
Tests
sera in the unvaccinated group, determine the maximum
Aluminium (2.3.9). Maximum 1.25 mg per single human dose, reaction level that can be expected to occur in an unvaccinated
if hydrated aluminium phosphate or aluminium hydroxide is animal for that particular assay. Any response in vaccinated
used as the adsorbent. animals that exceeds this level is by definition a seroconversion.
Free formaldehyde (2.3.20). When formaldehyde has been Make a suitable transformation of the percentage of animals
used for inactivation, the vaccine complies with the test showing seroconversion in each group (for example, a probit
prescribed in Vaccines. transformation) and analyse the data according to a parallel-
line log dose-response model. Determine the potency of the
Antimicrobial preservative. Where applicable, determine the
test preparation relative to the reference preparation.
amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not Validity conditions. The test is not valid unless (a) for both
greater than 115.0 per cent of the intended amount. the test and the reference vaccine, the ED50 lies between the
smallest and the largest doses given to the animals; (b) the
Sterility (2.2.11). Complies with the test for sterility.
statistical analysis shows no significant deviation from
Abnormal toxicity (2.2.1). Complies with the test for abnormal linearity or parallelism; (c) the fiducial limits of the estimated
toxicity. relative potency fall between 33.0 and 300.0 per cent of the
estimated potency.
Assay
Potency. The upper fiducial limit (P = 0.95) of the estimated
The vaccine complies with the assay of Hepatitis A vaccine. relative potency is not less than 1.0.
The assay is carried out either in vivo, by comparing in given In vitro assay
condition the capacity to induce specific antibodies in mice
with the same capacity of a reference preparation or in vitro Carry out an immunochemical determination of antigen content
by an immunochemical determination of antigen content (2.2.14) with acceptance criteria validated against the in vivo
(2.2.14). test. The acceptance criteria are approved for a given reference
preparation by the National Regulatory Authority in the light
In vivo assay of the validation data.
The test on mice shown below is given as an example of a Labelling. The label states (1) the biological origin of the cells
method that has been found suitable for a given vaccine; and; (2) the adjuvant used for the preparation of the vaccine.
other validated methods may also be used.
Selection and distribution of the test animals. Healthy mice
from the same stock, about 5 weeks old and from a strain
shown to be suitable should be used in the test. Use animals
Inactivated Hepatitis B Vaccine
of the same sex. Distribute the animals in at least seven equal Inactivated Hepatitis B Vaccine is a non-infectious inactivated
groups of a number suitable for the requirements of the assay. liquid preparation derived from the surface antigen of Hepatitis
Determination of potency of the vaccine under examination. B virus (HbsAg). This preparation is white or almost white
Using a 0.9 per cent w/v solution of sodium chloride containing translucent liquid in which the mineral carrier tends to settle
the aluminium adjuvant used for the vaccine, prepare at least down slowly on keeping but is free from foreign particles /
three dilutions of the vaccine under examination and matching floccules.
dilutions of the reference preparation. Allocate the dilutions
Production
one to each of the groups of animals and inject
subcutaneously not more than 0.5 ml of each dilution into The antigen is harvested and purified from the plasma of human
each animal in the group to which that dilution is allocated. carriers of Hepatitis B virus. The surface antigen contains all
Maintain a group of unvaccinated controls, injected the three antigen species (S, Pre-S1, Pre-S2). The individual
subcutaneously with the same volume of diluent. After 28 to donor plasma is shown by sensitive tests to be seronegative
32 days, anaesthetise and bleed all animals, keeping the for HIV-I and HIV-2 and for HCV. The plasma pool is tested for
individual sera separate. Assay the individual sera for specific freedom from adventitious viruses and blood borne

52
IP 2007 INACTIVATED HEPATITIS B VACCINE

transmissible pathogens by appropriate methods. The purified The antigen/protein ratio is within the limits approved for the
antigen is further inactivated by a validated method, usually specific product.
with formalin or any other inactivating agent, to render the The molecular weight of the major band in a sodium dodecyl
hepatitis B virus harmless. The preparation is also tested for sulphate polyacrylamide gel electrophoresis (SDS-PAGE)
the residual HBV DNA using a sensitive test approved by the under reduced conditions corresponds to the value expected
competent authority and the level is shown to be less than 1 from the gene sequence and possible glycosylation.
pg HBV DNA per 50 doses.
Antigenic purity. The purity of the antigen is determined by
The method used for production of the vaccine must have comparison with a reference preparation using liquid
been shown to yield a product consistently complying with chromatography or other suitable methods such as SDS-PAGE
the requirements of immunogenicity, safety and stability. The with staining. A suitable method is sensitive enough to detect
production method must also be validated to demonstrate a potential contaminant at a concentration of 1.0 per cent of
that the product, if tested, would comply with the tests for total protein. Not less than 95.0 per cent of the total protein
safety and efficacy. consists of hepatitis B surface antigen.
Reference preparation. A part of a batch shown to be at least Composition. The content of proteins, lipids, nucleic acids
as immunogenic as a batch that produced in clinical studies in and carbohydrates is determined.
young healthy adults not less than 95.0 per cent
seroconversion, corresponding to a level of neutralizing Sterility (2.2.11). The purified antigen complies with the test
antibody accepted to be protective (HbsAg antibody titre not for sterility carried out using 10 ml for each medium.
less than 10 mIU/ml after a full course of primary immunization Additional tests on the purified antigen may be required
determined by enzyme-linked immunosorbent assay (ELISA) depending on the production method used.
is used as a reference preparation.
FINAL BULK VACCINE
Characterisation of the substance
An antimicrobial preservative and an adjuvant may be
Development studies are carried out to characterize the included in the vaccine.
antigen. The complete protein, lipid and carbohydrate structure
of the antigen is established. The morphological characteristics Only a final bulk vaccine that complies with the following
of the antigen particles are established by electron microscopy. requirements may be used in the preparation of the final lot.
The buoyant density of the antigen particles is determined by Antimicrobial preservative. Where applicable, determine the
a physico-chemical method (2.4.29), for example gradient amount of antimicrobial preservative by a suitable chemical or
centrifugation. The antigenic epitopes are characterized. The physico-chemical method. The amount is not less than the
protein fraction of the antigen is characterized in terms of the 85.0 per cent and not greater than 115.0 per cent of that stated
primary structure (for example, by determination of the amino- on the label.
acid composition, by partial amino-acid sequence analysis
Sterility (2.2.11). Carry out test for sterility using 10 ml of
and by peptide mapping).
bulk for each sterility medium.
PROPAGATION AND HARVEST
FINAL LOT
Identity, microbial purity, plasmid retention and consistency
of yield are determined at suitable production stages. Only a final lot that complies with each of the requirements
given below under Identification, Tests and Assay may be
PURIFIED ANTIGEN
released for use. Provided that the tests for free formaldehyde,
Only a purified antigen that complies with the following antimicrobial preservative content and the assay in animals,
requirements may be used in the preparation of the final bulk. where applicable, have been carried out on the final bulk
Total protein (2.3.49). The total protein is determined by a vaccine with satisfactory results, they may be omitted on the
validated method. The content is within the limits approved final lot.
for the specific product. Identification
Antigen content and identification. The quantity and
specificity of HBsAg is determined in comparison with the The assay or, where applicable, the electrophoretic profile,
International standard for HBsAg subtype ad or an in-house also serves to identify the vaccine.
reference, by a suitable immunochemical method such as radio
Tests
immunoassay (RIA), enzyme-linked immunosorbent assay
(ELISA), immunoblot (preferably using a monoclonal antibody Aluminium (2.3.9). When hydrated aluminium phosphate or
directed against a protective epitope) or single radial diffusion. aluminium hydroxide is used as the adsorbent, the vaccine

53
INACTIVATED INFLUENZA VACCINE (SPLIT VIRION) IP 2007

complies with the test prescribed in the monograph on recommends new strains corresponding to prevailing
Vaccines. epidemiological evidence.
Test for inactivating agent. The concentration of any The origin and passage history of virus strains shall be
inactivating agent remaining in the final vaccine shall be approved by the National Regulatory Authority.
determined by methods approved by the competent authority.
The concentration shall not exceed a specified upper limit. Substrate for virus propagation

Antimicrobial preservative. Where applicable, determine the Influenza virus seed to be used in the production of vaccine is
amount of antimicrobial preservative by a suitable chemical propagated in fertilised eggs from chicken flocks free from
method. The content is not less than 85.0 per cent and not specified pathogens or in suitable cell cultures, such as chick-
greater than 115.0 per cent of the intended amount. embryo fibroblasts or chick kidney cells obtained from chicken
flocks free from specified pathogens. For production, the virus
Sterility (2.2.11). Complies with the test for sterility.
of each strain is grown in the allantoic cavity of eggs derived
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject from specific pathogen free flocks.
the equivalent of one human dose into each rabbit.
SEED LOT
A validated test for bacterial endotoxins (2.2.3) may be used
instead of the test for pyrogens. The production of vaccine is based on a seed-lot system.
Working seed lots represent not more than fifteen passages
Assay from the approved reassorted virus or the approved virus
The upper fiducial limit (P = 0.95) of the estimated relative isolate. The final vaccine represents one passage from the
potency is not less than 1.0. working seed lot. The haemagglutinin and neuraminidase
antigens of each seed lot are identified as originating from the
Determine the potency by method A (Biological) as described correct strain of influenza virus by suitable methods.
under Hepatitis-B vaccine (rDNA).
Only a working virus seed lot that complies with the following
Labelling. The label states (1) the amount of HBsAg per dose; requirements may be used in the preparation of the monovalent
(2) the name and amount of inactivating agent; (3) the name pooled harvest.
and amount of the adjuvant; (4) that the vaccine must be
shaken before use; (5) that the vaccine must not be frozen. Sterility (2.2.11). Carry out the test for sterility using 10 ml for
each medium.
Mycoplasmas (2.7.4). Carry out the test for mycoplasmas using
10 ml.
Inactivated Influenza Vaccine (Split
Virion) PROPAGATION AND HARVEST

Influenza Vaccine (Split Virion, Inactivated) is a sterile, aqueous An antimicrobial agent may be added to the inoculum. After
suspension of a strain or strains of influenza virus, type A or incubation at a controlled temperature, the allantoic fluids are
B, or a mixture of strains of the two types grown individually harvested and combined to form a monovalent pooled harvest.
in eggs derived from specific pathogen free flock or cell An antimicrobial agent may be added at the time of harvest.
cultures, inactivated and treated so that the integrity of the At no stage in the production, penicillin or streptomycin is
virus particles has been disrupted without diminishing the used.
antigenic properties of the haemagglutinin and neuraminidase
MONOVALENT POOLED HARVEST
antigens. The stated amount of haemagglutinin antigen for
each strain present in the vaccine is 15 µg per dose, unless To limit the possibility of contamination, inactivation is initiated
clinical evidence supports the use of a different amount. as soon as possible after preparation. The virus is inactivated
by a method that has been demonstrated on three consecutive
Production
batches to be consistently effective for the manufacturer. The
The production method is validated to demonstrate that the inactivation process shall have been shown to be capable of
product, if tested, would comply with the test for safety and inactivating the influenza virus without destroying its
efficacy. antigenicity; the process should cause minimum alteration of
the haemagglutinin and neuraminidase antigens. The
Choice of vaccine strain
inactivation process shall also have been shown to be capable
The World Health Organisation reviews the world of inactivating avian leucosis viruses and mycoplasmas. If
epidemiological situation annually and if necessary the monovalent pooled harvest is stored after inactivation, it

54
IP 2007 INACTIVATED INFLUENZA VACCINE (SPLIT VIRION)

is held at a temperature of 5±3°. If formaldehyde solution is Sterility (2.2.11). Carry out the test for sterility using 10 ml for
used, the concentration does not exceed 0.2 g/l of each sterility medium.
formaldehyde at any time during inactivation; if FINAL LOT
betapropiolactone is used, the concentration does not exceed
0.1 per cent v/v at any time during inactivation. The final bulk vaccine is distributed aseptically into sterile,
tamper-proof containers. The containers are closed so as to
Before or after the inactivation procedure, the monovalent
prevent contamination.
pooled harvest is concentrated and purified by high-speed
centrifugation or other suitable method and the virus particles Only a final lot that is satisfactory with respect to each of the
are disrupted into component subunits by the use of approved requirements given below under Tests and Assay may be
procedures. For each new strain, a validation test is carried released for use. Provided that the test for viral inactivation
out to show that the monovalent bulk consists predominantly has been performed with satisfactory results on each
of disrupted virus particles. monovalent pooled harvest and that the tests for free
formaldehyde, ovalbumin and total protein have been
Only a monovalent pooled harvest that complies with the
performed with satisfactory results on the final bulk vaccine,
following requirements may be used in the preparation of the
they may be omitted on the final lot.
final bulk vaccine.
Description. The vaccine is a slightly opalescent liquid.
Haemagglutinin antigen. Determine the content of
haemagglutinin antigen by an immunodiffusion test (2.2.14), Identification
by comparison with a haemagglutinin antigen reference
preparation or with an antigen preparation calibrated against The assay serves to confirm the antigenic specificity of the
it. Carry out the test at 20° to 25°. vaccine.
For some vaccines, the physical form of the haemagglutinin Tests
particles prevents quantitative determination by
Viral inactivation. Inoculate 0.2 ml of the vaccine into the
immunodiffusion after inactivation of the virus. For these
allantoic cavity of each of ten fertilised eggs and incubate at
vaccines, a determination of haemagglutinin antigen is made
33° to 37° for 3 days. The test is not valid unless at least eight
on the monovalent pooled harvest before inactivation. The
of the ten embryos survive. Harvest 0.5 ml of the allantoic
production process is validated to demonstrate suitable
fluid from each surviving embryo and pool the fluids. Inoculate
conservation of haemagglutinin antigen and a suitable tracer
0.2 ml of the pooled fluid into a further ten fertilised eggs and
is used for formulation, for example, protein content.
incubate at 33° to 37° for 3 days. The test is not valid unless at
Neuraminidase antigen. The presence and type of least eight of the ten embryoes survive. Harvest about 0.1 ml
neuraminidase antigen are confirmed by suitable enzymatic or of the allantoic fluid from each surviving embryo and examine
immunological methods (2.2.14) on the first three monovalent each individual harvest for live virus by a haemagglutination
pooled harvests from each working seed lot. test. If haemagglutination is found for any of the fluids, carry
Sterility (2.2.11). Carry out the test for sterility using 10 ml for out for that fluid a further passage in eggs and test for
each medium. haemagglutination; no haemagglutination occurs.

Viral inactivation. Carry out as described below under Tests. Total protein (2.3.49). Not more than six times the total
haemagglutinin content of the vaccine as determined in the
Chemicals used for disruption. Tests are carried out on the assay, but in any case, not more than 100 µg of protein per
monovalent pooled harvest for the chemicals used for virus strain per human dose and not more than a total of 300
disruption, the limits being approved by the National µg of protein per human dose.
Regulatory Authority.
Ovalbumin. Not more than 1 µg of ovalbumin per human dose,
FINAL BULK VACCINE determined by a suitable technique using a suitable reference
Appropriate quantities of the monovalent pooled harvests preparation of ovalbumin.
are blended to make the final bulk vaccine. Antimicrobial preservative. Where applicable, determine the
Only a final bulk vaccine that complies with the following amount of antimicrobial preservative by a suitable chemical
requirements may be used in the preparation of the final lot. method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount.
Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical Sterility (2.2.11). Complies with the test for sterility.
method. The content is not less than 85.0 per cent and not Abnormal toxicity (2.2.1). Complies with the test for abnormal
greater than 115.0 per cent of the intended amount. toxicity.

55
INACTIVATED INFLUENZA VACCINE (SURFACE ANTIGEN) IP 2007

Bacterial endotoxins (2.2.3). Not more than 100 IU of endotoxin approved by the competent authority.
per human dose.
Substrate for virus propagation
Assay
Influenza virus seed to be used in the production of vaccine is
Determine the content of haemagglutinin antigen by an propagated in fertilised eggs from chicken flocks free from
immunodiffusion test (2.2.14), by comparison with an specified pathogens or in suitable cell cultures, such as chick-
appropriate haemagglutinin antigen reference preparation. embryo fibroblasts or chick kidney cells obtained from chicken
Carry out the test at 20 to 25°. The confidence interval (P = flocks free from specified pathogens. For production, the virus
0.95) of the assay is not greater than 80.0 per cent to 125.0 per of each strain is grown in the allantoic cavity of eggs derived
cent of the estimated content. The lower confidence limit (P = from SPF flocks.
0.95) of the estimate of haemagglutinin antigen content is not
less than 80.0 per cent of the amount stated on the label for SEED LOT
each strain.
The production of vaccine is based on a seed-lot system.
For some vaccines, quantitative determination of Working seed lots represent not more than fifteen passages
haemagglutinin antigen with respect to available reference from the approved reassorted virus or the approved virus
preparations is not possible. An immunological identification isolate. The final vaccine represents one passage from the
of the haemagglutinin antigen and a semi-quantitative working seed lot. The haemagglutinin and neuraminidase
determination are carried out instead by suitable methods. antigens of each seed lot are identified as originating from the
correct strain of influenza virus by suitable methods.
Labelling. The label complies with the requirements stated
under Vaccines and also states (a) that the vaccine has been Only a working virus seed lot that complies with the following
prepared on eggs; (b) the strain or strains of influenza virus requirements may be used in the preparation of the monovalent
used to prepare the vaccine; (c) the method of inactivation; pooled harvest.
(d) the haemagglutinin content in µg per virus strain per dose; Sterility (2.2.11). Carry out the test for sterility using 10 ml for
(e) the season during which the vaccine is intended to protect. each medium.
Mycoplasmas (2.7.4). Carry out the test for mycoplasmas using
10 ml.
Inactivated Influenza Vaccine (Surface PROPAGATION AND HARVEST
Antigen)
An antimicrobial agent may be added to the inoculum. After
Influenza Vaccine (Surface Antigen, Inactivated) is a sterile, incubation at a controlled temperature, the allantoic fluids are
aqueous suspension of a strain or strains of influenza virus, harvested and combined to form a monovalent pooled harvest.
type A or B, or a mixture of strains of the two types grown An antimicrobial agent may be added at the time of harvest.
individually in eggs derived from specific pathogen free flocks At no stage in the production penicillin or streptomycin is
or cell cultures, inactivated and treated so that the preparation used.
consists predominantly of haemagglutinin and neuraminidase
antigens, without diminishing the antigenic properties of these MONOVALENT POOLED HARVEST
antigens. The stated amount of haemagglutinin antigen for
To limit the possibility of contamination, inactivation is initiated
each strain present in the vaccine is 15 µg per dose, unless
as soon as possible after preparation. The virus is inactivated
clinical evidence supports the use of a different amount.
by a method that has been demonstrated on three consecutive
Production batches to be consistently effective for the manufacturer. The
inactivation process shall have been shown to be capable of
The production method is validated to demonstrate that the inactivating the influenza virus without destroying its
product, if tested, would comply with the test for safety and antigenicity; the process should cause minimum alteration of
efficacy. the haemagglutinin and neuraminidase antigens. The
Choice of vaccine strain inactivation process shall also have been shown to be capable
of inactivating avian leucosis viruses and mycoplasmas. If
The World Health Organisation reviews the world
the monovalent pooled harvest is stored after inactivation, it
epidemiological situation annually and if necessary
is held at a temperature of 5±3°. If formaldehyde solution is
recommends new strains corresponding to prevailing
used, the concentration does not exceed 0.2 g/l of
epidemiological evidence.
formaldehyde at any time during inactivation; if
The origin and passage history of virus strains shall be betapropiolactone is used, the concentration does not exceed

56
IP 2007 INACTIVATED INFLUENZA VACCINE (SURFACE ANTIGEN)

0.1 per cent v/v at any time during inactivation. Only a final lot that is satisfactory with respect to each of the
Before or after the inactivation process, the monovalent requirements given below under Identification, Tests and
pooled harvest is concentrated and purified by high-speed Assay may be released for use. Provided that the test for viral
centrifugation or other suitable method. Virus particles are inactivation has been performed with satisfactory results on
disrupted into component subunits by approved procedures each monovalent pooled harvest and that the tests for free
and further purified so that the monovalent bulk consists formaldehyde, ovalbumin and total protein have been
mainly of haemagglutinin and neuraminidase antigens. performed with satisfactory results on the final bulk vaccine,
they may be omitted on the final lot.
Only a monovalent pooled harvest that complies with the
Description. The vaccine is a clear liquid.
following requirements may be used in the preparation of the
final bulk vaccine. Identification
Haemagglutinin antigen. Determine the content of The assay serves to confirm the antigenic specificity of the
haemagglutinin antigen by an immunodiffusion test (2.2.14), vaccine.
by comparison with a haemagglutinin antigen reference
preparation or with an antigen preparation calibrated against Tests
it. Carry out the test at 20° to 25°. Viral inactivation. Inoculate 0.2 ml of the vaccine into the
Neuraminidase antigen. The presence and type of allantoic cavity of each of ten fertilised eggs and incubate at
neuraminidase antigen are confirmed by suitable enzymatic or 33° to 37° for 3 days. The test is not valid unless at least eight
immunological methods (2.2.14) on the first three monovalent of the ten embryos survive. Harvest 0.5 ml of the allantoic
pooled harvests from each working seed lot. fluid from each surviving embryo and pool the fluids. Inoculate
0.2 ml of the pooled fluid into a further ten fertilised eggs and
Sterility (2.2.11). Carry out the test for sterility, using 10 incubate at 33° to 37° for 3 days. The test is not valid unless at
ml for each medium. least eight of the ten embryos survive. Harvest about 0.1 ml of
Viral inactivation. Carry out the test described below under the allantoic fluid from each surviving embryo and examine
Tests. each individual harvest for live virus by a haemagglutination
test. If haemagglutination is found for any of the fluids, carry
Purity. The purity of the monovalent pooled harvest is out for that fluid a further passage in eggs and test for
examined by polyacrylamide gel electrophoresis or by other haemagglutination; no haemagglutination occurs.
approved techniques. Mainly haemagglutinin and
neuraminidase antigens shall be present. Total protein (2.3.49). Not more than 40 µg of protein other
than haemagglutinin per virus strain per human dose and not
Chemicals used for disruption and purification. Tests are more than a total of 120 µg of protein other than haemagglutinin
carried out on the monovalent pooled harvest for the chemicals per human dose.
used for disruption and purification, the limits being approved
by the competent authority. Ovalbumin. Not more than 1 µg of ovalbumin per human dose,
determined by a suitable technique using a suitable reference
FINAL BULK VACCINE preparation of ovalbumin.
Appropriate quantities of the monovalent pooled harvests Free formaldehyde (2.3.20). Complies with the test for free
are blended to make the final bulk vaccine. formaldehyde as stated under General Requirements for
Vaccines for Human Use.
Only a final bulk vaccine that complies with the following
requirements may be used in the preparation of the final lot. Antimicrobial preservative. Where applicable, determine the
amount of antimicrobial preservative by a suitable chemical
Antimicrobial preservative. Where applicable, determine the method. The content is not less than 85.0 per cent and not
amount of antimicrobial preservative by a suitable chemical greater than 115.0 per cent of the intended amount.
method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount. Abnormal toxicity (2.2.1). Complies with the test for abnormal
toxicity.
Sterility (2.2.11). Carry out the test for sterility using 10 ml for
each medium. Sterility (2.2.11). Complies with the test for sterility.

FINAL LOT Bacterial endotoxins (2.2.3). Not more than 100 IU of endotoxin
per human dose.
The final bulk vaccine is distributed aseptically into sterile,
tamper-proof containers. The containers are closed so as to Assay
prevent contamination. Determine the content haemagglutinin antigen by an

57
INACTIVATED INFLUENZA VACCINE (WHOLE VIRION) IP 2007

immunodiffusion test (2.2.14), by comparison with an SEED LOT


appropriate haemagglutinin antigen reference preparation. The production of vaccine is based on a seed-lot system.
Carry out the test at 20° to 25°. The confidence interval Working seed lots represent not more than fifteen passages
(P = 0.95) of the assay is not greater than 80.0 per cent to 125.0 from the approved reassorted virus or the approved virus
per cent of the estimated content. The lower confidence limit isolate. The final vaccine represents one passage from the
(P = 0.95) of the estimate of haemagglutinin antigen content is working seed lot. The haemagglutinin and neuraminidase
not less than 80.0 per cent of the amount stated on the label antigens of each seed lot are identified as originating from the
for each strain. correct strain of influenza virus by suitable methods.
Labelling. The label states (1) that the vaccine has been Only a working virus seed lot that complies with the following
prepared on eggs; (2) the strain or strains of influenza virus requirements may be used in the preparation of the monovalent
used to prepare the vaccine; (3) the method of inactivation; pooled harvest.
(4) the haemagglutinin content in micrograms per virus strain
per dose; (5) the season during which the vaccine is intended Sterility (2.2.11). Carry out the test for sterility using 10 ml for
to protect. each medium.
Mycoplasmas (2.7.4). Carry out the test for mycoplasmas using
10 ml.

Inactivated Influenza Vaccine (Whole PROPAGATION AND HARVEST


Virion) An antimicrobial agent may be added to the inoculum. After
incubation at a controlled temperature, the allantoic fluids are
Influenza Vaccine (Whole Virion, Inactivated) is a sterile, harvested and combined to form a monovalent pooled harvest.
aqueous suspension of a strain or strains of influenza virus, An antimicrobial agent may be added at the time of harvest.
type A or B, or a mixture of strains of the two types grown At no stage in the production is penicillin or streptomycin
individually in eggs derived from specific pathogen free flocks used.
or cell culture and inactivated in such a manner that their
antigenic properties are retained. The stated amount of MONOVALENT POOLED HARVEST
haemagglutinin antigen for each strain present in the vaccine To limit the possibility of contamination, inactivation is initiated
is 15 µg per dose, unless clinical evidence supports the use of as soon as possible after preparation. The virus is inactivated
a different amount. by a method that has been demonstrated on three consecutive
batches to be consistently effective for the manufacturer. The
Production
inactivation process shall have been shown to be capable of
The production method is validated to demonstrate that the inactivating the influenza virus without destroying its
product, if tested, would comply with the test for safety and antigenicity; the process should cause minimum alteration of
efficacy. the haemagglutinin and neuraminidase antigens. The
inactivation process shall also have been shown to be capable
Choice of vaccine strain of inactivating avian leucosis viruses and mycoplasmas. If
the monovalent pooled harvest is stored after inactivation, it
The World Health Organisation reviews the world
is held at a temperature of 5±3°. If formaldehyde solution is
epidemiological situation annually and, if necessary,
used, the concentration does not exceed 0.2 g/l of
recommends new strains corresponding to prevailing
formaldehyde at any time during inactivation; if
epidemiological evidence.
betapropiolactone is used, the concentration does not exceed
The origin and passage history of virus strains shall be 0.1 per cent v/v at any time during inactivation.
approved by the competent authority.
Before or after the inactivation process, the monovalent
Substrate for virus propagation pooled harvest is concentrated and purified by high-speed
centrifugation or other suitable method.
Influenza virus seed to be used in the production of vaccine is
Only a monovalent pooled harvest that complies with the
propagated in fertilised eggs from chicken flocks free from
following requirements may be used in the preparation of the
specified pathogens or in suitable cell cultures, such as chick-
final bulk vaccine.
embryo fibroblasts or chick kidney cells obtained from chicken
flocks free from specified pathogens .For production, the virus Haemagglutinin antigen. Determine the content of
of each strain is grown in the allantoic cavity of eggs derived haemagglutinin antigen by an immunodiffusion test (2.2.14),
from specific pathogen free flocks. by comparison with a haemagglutinin antigen reference

58
IP 2007 JAPANESE ENCEPHALITIS VACCINE (HUMAN)

preparation or with an antigen preparation calibrated against the allantoic fluid from each surviving embryo and examine
it. Carry out the test at 20° to 25°. each individual harvest for live virus by a haemagglutination
Neuraminidase antigen. The presence and type of test. If haemagglutination is found for any of the fluids, carry
neuraminidase antigen are confirmed by suitable enzymatic or out for that fluid a further passage in eggs and test for
immunological methods (2.2.14) on the first three monovalent haemagglutination; no haemagglutination occurs.
pooled harvests from each working seed lot. Total protein (2.3.49). Not more than six times the total
haemagglutinin content of the vaccine as determined in the
Sterility (2.2.11). Carry out the test for sterility using 10 ml for
assay, but in any case, not more than 100 µg of protein per
each medium.
virus strain per human dose and not more than a total of 300
Viral inactivation. Carry out the test described below under µg of protein per human dose.
Tests.
Ovalbumin. Not more than 1 µg of ovalbumin per human dose,
FINAL BULK VACCINE determined by a suitable technique using a suitable reference
Appropriate quantities of the monovalent pooled harvests preparation of ovalbumin.
are blended to make the final bulk vaccine. Free formaldehyde (2.3.20). Maximum 0.2 g/l.
Only a final bulk vaccine that complies with the following Antimicrobial preservative. Where applicable, determine the
requirements may be used in the preparation of the final lot. amount of antimicrobial preservative by a suitable chemical
Antimicrobial preservative. Where applicable, determine the method. The content is not less than the minimum amount
amount of antimicrobial preservative by a suitable chemical shown to be effective and is not greater than 115.0 per cent of
method. The content is not less than 85.0 per cent and not the quantity stated on the label.
greater than 115.0 per cent of the intended amount. Abnormal toxicity (2.2.1). Complies with the test for abnormal
Sterility (2.2.11). Carry out test for sterility using 10 ml of toxicity.
bulk for each sterility medium. Sterility (2.2.11). Complies with the test for sterility.
FINAL LOT
Bacterial endotoxins (2.2.3). Not more than 100 IU of endotoxin
The final bulk vaccine is distributed aseptically into sterile, per human dose.
tamper-proof containers. The containers are closed so as to
prevent contamination. Assay

Only a final lot that is satisfactory with respect to each of the Determine the content of haemagglutinin antigen by an
requirements given below under Tests and Assay may be immunodiffusion test (2.2.14), by comparison with an
released for use. Provided that the test for viral inactivation appropriate haemagglutinin antigen reference preparation.
has been performed with satisfactory results on each Carry out the test at 20° to 25°. The confidence interval (P =
monovalent pooled harvest and that the tests for free 0.95) of the assay is not greater than 80.0 per cent to 125.0 per
formaldehyde, ovalbumin and total protein have been cent of the estimated content. The lower confidence limit (P =
performed with satisfactory results on the final bulk vaccine, 0.95) of the estimate of haemagglutinin antigen content is not
they may be omitted on the final lot. less than 80.0 per cent of the amount stated on the label for
each strain.
Description. The vaccine is a slightly opalescent liquid.
Labelling. The label states (1) that the vaccine has been
Identification prepared on eggs; (2) the strain or strains of influenza virus
used to prepare the vaccine; (3) the method of inactivation;
The assay serves to confirm the antigenic specificity of the
(4) the haemagglutinin content in micrograms per virus strain
vaccine.
per dose; (5) the season during which the vaccine is intended
Tests to protect.
Viral inactivation. Inoculate 0.2 ml of the vaccine into the
allantoic cavity of each of ten fertilised eggs and incubate at
33° to 37° for 3 days. The test is not valid unless at least eight Japanese Encephalitis Vaccine
of the ten embryos survive. Harvest 0.5 ml of the allantoic
fluid from each surviving embryo and pool the fluids. Inoculate
(Human)
0.2 ml of the pooled fluid into a further ten fertilised eggs and Japanese Encephalitis Vaccine for human use is a liquid or
incubate at 33° to 37° for 3 days. The test is not valid unless at freeze dried preparation of Japanese encephalitis virus grown
least eight of the ten embryos survive. Harvest about 0.1 ml of in approved substrate and inactivated by a validated method.

59
JAPANESE ENCEPHALITIS VACCINE (HUMAN) IP 2007

Production if present are reduced to an acceptable level by suitable method


of purification. Serum and trypsin used in the preparation of
General provisions cell suspension and media are shown to be free from infectious
extraneous agents. The cell culture media may contain a pH
The vaccine is produced on the basis of virus seed lot system
indicator such as phenol red. Not less than 500 ml of the cell
The production method shall have been shown to yield
cultures employed for vaccine production are set aside as
consistently the vaccines that comply with the tests for
uninfected cell cultures (control cells). The virus suspension
immunogenicity, safety and stability. The production method
is harvested on one or more occasions during incubation.
is validated to demonstrate that the product, if tested, would
Multiple harvests from the same production cell culture may
comply with the test for safety and efficacy.
be pooled and considered as a single harvest.
Substrate for virus propagation
Virus harvests that comply with the tests given under
The virus is propagated in an approved cell substrate like a Identification and Virus concentration are pooled in the
Vero cell line. preparation of the inactivated viral harvest.
SEED LOT Control cells. The control cells of the production cell culture
from which the single harvest is derived should comply with
The strain of Japanese encephalitis virus used shall be
the test for identification and with the tests for extraneous
identified by historical records that include information on
agents (2.7.3).
the origin of the strain and its subsequent manipulation.
The National Regulatory Authority shall determine the Purification and inactivation
acceptable number of passages from the master virus seed lot The virus harvest may be concentrated and/or purified by
to produce working virus seed lots. suitable methods; the virus harvest is inactivated by a validated
Only a working seed lot that complies with the following tests method at a fixed, well defined stage of the process which may
may be used for virus propagation. be before, during or after any concentration or purification.
The method shall have been shown to be capable of
Identification inactivating Japanese encephalitis virus without destruction
of the immunogenic activity. If formalin is used, the
Each working seed lot is identified as Japanese encephalitis concentration shall at no time exceed 1:2000.
virus using specific antibodies by an approved method.
Only an inactivated viral suspension that complies with the
Virus concentration. The virus concentration of each working following tests may be used in the preparation of the final
seed lot is determined by a cell culture method using bulk vaccine.
immunofluoresence or any other approved method.
Inactivation. Inactivation is confirmed by carrying out an
Extraneous agents (2.7.3). The working seed lot complies with amplification test for residual infectious Japanese encephalitis
the tests for the virus seed lots. virus. Inoculate a quantity of inactivated viral suspension
equivalent to not less than 25 doses into cell cultures of the
PROPAGATION AND HARVEST
same type as those used for production of the vaccine. Make
a passage after 7 days. Maintain the cultures for a further
a) Mouse brain vaccine
period of 14 days and then examine the cell cultures for
The vaccine is prepared by using a seed-lot system. An Japanese encephalitis virus using an immunofluorescence test.
approved strain of virus is grown by inoculating intracerebrally No Japanese encephalitis virus is detected. Alternatively, 5 ml
into healthy mice. Virus harvests are pooled, concentrated of each culture fluid is pooled on day 14 and 21 and 0.03 ml is
and inactivated by addition of formalin or any other suitable inoculated intracerebrally into each of the 10 mice weighing
inactivating agent. It may contain a suitable preservative. The between 12 and 15 g. The mice are observed for 14 days for
vaccine may be issued in single or multidose containers. symptoms caused by Japanese encephalitis virus, and mice
showing symptoms of Japanese encephalitis virus are
b) Cell culture vaccine sacrificed and virus presence is confirmed by
All processing of the cell bank and subsequent cell cultures immunofluorescence test. No Japanese encephalitis virus shall
are done under aseptic conditions in an area where no other be detected.
cells are handled. Approved animal (but not human) serum Residual host-cell DNA (2.2.15). The content of residual host-
may be used in the media, but the final medium for maintaining cell DNA, determined using a suitable method, should not be
cell growth during virus multiplication does not contain animal greater than 10 ng per single human dose if cells are used in
serum; the media may contain human albumin. Serum proteins, the production.

60
IP 2007 JAPANESE ENCEPHALITIS VACCINE (HUMAN)

FINAL BULK VACCINE Antimicrobial preservative. Determine the amount of


antimicrobial preservative by a suitable chemical method. The
The final bulk vaccine is prepared from one or more inactivated
content is not less than the minimum amount shown to be
viral suspensions. An approved stabilizer may be added to
effective and is not greater than 115.0 per cent of that stated
maintain the activity of the product. Thiomersal can be used
on the label.
as preservative.
Only a final bulk vaccine that complies with the following Biological assay
requirements may be used in the preparation of the final lot. Potency of Japanese encephalitis virus vaccine is determined
Formaldehyde (2.3.20). Not more than 0.01 per cent w/v. by titrating the neutralizing antibodies produced in the
immunized mice by plaque reduction method or serum
Antimicrobial preservative. Where applicable, determine the
neutralization test (SNT) using appropriate cell culture.
amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not Standard preparation
greater than 115.0 per cent of the intended amount.
The Standard preparation is a freeze-dried Japanese
Sterility (2.2.11). Carry out test for sterility using 10 ml of encephalitis virus vaccine the potency of which has been
bulk for each sterility medium. determined in relation to the Japanese encephalitis reference
FINAL LOT vaccine obtained from the National Institute of Health, Tokyo,
Japan.
The final bulk vaccine is distributed aseptically into sterile
containers. The containers are then sealed so as to prevent Suggested method
contamination.
Preparation of challenge virus suspension
Only a final lot that complies with each of the tests given
under Identification, Tests and Assay may be released for The approved challenge virus strain is stored in freeze-dried
use. Provided that the test for inactivation has been carried form or in liquid form stored below -70° . Prepare a working
out with satisfactory results on the inactivated virus pool of the challenge virus strain by inoculating intracerebrally
suspension and the test for bovine serum albumin has been 0.03 ml of 100 fold dilution of the standard strain in Hanks’
carried out with satisfactory results on the final bulk vaccine, balanced salt solution containing 5 per cent calf serum into a
these tests may be omitted on the final lot. suitable number of 2-day old suckling mice. Sacrifice the
animals after they show characteristic symptoms of
Identification encephalitis and become moribund. Harvest their brains
aseptically, wash them in chilled sterile saline solution to
The vaccine is shown to contain Japanese encephalitis virus
remove blood clots. Homogenize the brains with Hanks’
antigen by a suitable immunochemical method using specific
balanced salt solution containing 5 per cent calf serum to
antibodies, alternatively, the Assay also serves to identify the
make a 10 per cent emulsion. Centrifuge the emulsion at 2000
vaccine.
g for 30 minutes. Dilute the supernatant with Hanks’ balanced
Tests salt solution containing 5 per cent calf serum so as to contain
about 200 Plaque-Forming Units (PFU) of the virus per 0.4 ml.
Complies with the test for Inactivation under final Purification
and Inactivation. Determination of potency

Sterility (2.2.11). Complies with the test for sterility. Prepare appropriate dilutions of the vaccine under examination
and of the Standard Preparation in a suitable medium. Inject
Bacterial endotoxins (2.2.3). Less than 25 IU per single human intraperitoneally in two doses of 0.5 ml each at 7-day interval
dose. into at least 20 mice of 4 weeks of age. Bleed each mouse 7
Abnormal toxicity (2.2.1). Complies with the test for abnormal days after the second injection, pool the separated serum
toxicity. from each group and inactivate the sera by heating at 56° for
30 minutes. The inactivated sera may be stored at -20°, if
Bovine serum albumin (for cell culture vaccine). Not more
necessary.
than 50 ng per single human dose, determined by a suitable
immunochemical method (2.2.14). Dilute the sera appropriately, e.g. 1:40. 1:160, 1:640 etc., mix
with an equal volume of the challenge virus suspension and
Residual host-cell DNA (for continuous cell line vaccines)
incubate at 37° for 90 minutes for neutralization. Inoculate the
(2.2.15). Not more than 10 ng per single human dose.
mixture into cell cultures and overlay the infected cells with 1
Free formaldehyde (2.3.20). Not more than 0.01 per cent w/v. per cent agar. After incubation for an appropriate time (about

61
MEASLES AND RUBELLA VACCINE (LIVE) IP 2007

48 hours), stain the cells and count the number of plaques following test for thermal stability and with each of the tests
formed on the cultures to obtain the plaque reduction rates given below under Identification and Tests and Assay may be
for the vaccine under examination and the Standard released for use. Provided that the test for bovine serum albumin
preparation. Calculate the neutralizing antibody titres for each has been carried out with satisfactory results on the final bulk
group using standard statistical methods (5.7). The test is not vaccine, it may be omitted on the final lot.
valid unless (a) the mean number of plaques obtained with the Thermal stability. Maintain samples of the final lot of freeze-
Standard preparation is between 100 and 150 per dish and (b) dried vaccine in the dry state at 37° for 7 days. Determine the
the potency of the vaccine under examination is not less than virus concentration as described under Assay in parallel for
that of the Standard preparation. the vaccine held at 37° for 7 days and for vaccine stored at 2°
Labelling. The label states (1) the biological origin of the cells to 8°. For each component, the virus concentration of the
used for the preparation of the vaccine; (2) the strain of virus heated vaccine is not more than 1.0 log10 lower than that of the
used. unheated vaccine.
Identification
When the vaccine reconstituted as stated on the label is mixed
Measles and Rubella Vaccine (Live) with antibodies specific for measles virus and rubella virus, it
Measles and Rubella Vaccine (Live) is a freeze-dried preparation is no longer able to infect cell cultures susceptible to these
of suitable attenuated strains of measles virus and rubella viruses. When the vaccine reconstituted as stated on the
virus grown in suitable cell cultures. label is mixed with quantities of specific antibodies sufficient
to neutralize any one viral components, the second viral
The vaccine is reconstituted immediately before use to give a
component infects susceptible cell cultures.
clear liquid that may be coloured owing to the presence of a
pH indicator. Tests
Production Water (2.3.43). Not more than 3.0 per cent, determined by Karl
Fischer, semi-micro determination of water or by any suitable
General provisions validated method.
The two components are prepared as described in the Sterility (2.2.11). The reconstituted vaccine complies with the
monographs on Measles vaccine (live) and Rubella vaccine test for sterility.
(live) and comply with the tests prescribed therein.
Abnormal toxicity (2.2.1). Complies with the test for abnormal
The production method is validated to demonstrate that the toxicity.
product, if tested, would comply with the test for safety and
Bovine serum albumin. Not more than 50 ng per single human
efficacy.
dose, determined by a suitable immunochemical method
FINAL BULK VACCINE (2.2.14).
Virus harvests for each component are pooled and clarified to Assay
remove cells. A suitable stabilizer may be added and the pooled
A. Mix the vaccine with a sufficient quantity of antibodies
harvests diluted as appropriate. Suitable quantities of the
specific for rubella virus.Titrate the vaccine for infective
pooled harvest for each component are mixed.
measles virus at least in triplicate, using at least eight cell
Only a final bulk vaccine that complies with the following cultures for each dilution 0.5 log10 step or by a method of
requirements may be used in the preparation of the final lot. equal precision. Use an appropriate virus reference preparation
Sterility (2.2.11). Carry out test for sterility using 10 ml of to validate each assay. The estimated measles virus
bulk for each sterility medium. concentration is not less than that stated on the label; the
minimum measles virus concentration stated on the label is
FINAL LOT not less than 1x103 CCID50 per single human dose. The assay
is not valid if the confidence limits (P = 0.95) of the logarithm
For each component, a minimum virus concentration for
of the virus concentration are greater than ± 0.3.
release of the product is established such as to ensure, in the
light of stability data, that the minimum concentration stated Measles vaccine (Live) RS is suitable for use as a reference
on the label will be present at the end of the period of validity. preparation.
Only a final lot that complies with the tests for minimum virus B. Titrate the vaccine for infective rubella virus at least in
concentration of each component for release, with the triplicate, using at least eight cell cultures for each 0.5 log10

62
IP 2007 MEASLES VACCINE (LIVE)

dilution step or by a method of equal precision. Use an information on the origin of the strain and its subsequent
appropriate virus reference preparation to validate each assay. manipulation. Virus seed lots are prepared in large quantities
The estimated rubella virus concentration is not less than that and stored at temperatures below -20° if freeze-dried, or below
stated on the label; the minimum rubella virus concentration -60° if not freeze-dried.
stated on the label are not less than 1x103 CCID50 per single Only a seed lot that complies with the following tests may be
human dose. The assay is not valid if the confidence limits used for virus propagation.
(P = 0.95) of the logarithm of the virus concentration are greater
than ± 0.3. Identification
Rubella vaccine (Live) RS is suitable for use as a reference The master and working seed lots are identified as measles
preparation. virus by serum neutralization in cell culture, using specific
Labelling. The label states (1) the strains of virus used in the antibodies.
preparation of the vaccine; (2) the type and origin of the cells Virus concentration. The virus concentration of the master
used for the preparation of the vaccine; (3) the minimum virus and working seed lots is determined to monitor consistency
concentration for each component of the vaccine; (4) the time of production.
within which the vaccine must be used after reconstitution;
Extraneous agents (2.7.3). The working seed lot complies with
(5) that the vaccine must not be given to a pregnant woman
the tests for seed lots.
and that a woman should not become pregnant within two
months after having the vaccine. Neurovirulence (2.7.5). The master/working seed lot complies
with the test for neurovirulence of live virus vaccines. Macaca
and Cercopithecus monkeys susceptible to measles virus are
suitable for the test.
Measles Vaccine (Live)
PROPAGATION AND HARVEST
Measles Vaccine (Live) is a freeze-dried preparation of a
All processing of the cell bank and subsequent cell cultures is
suitable attenuated strain of measles virus. The vaccine is
done under aseptic conditions in an area where no other cells
reconstituted immediately before use, as stated on the label,
are handled. Suitable animal (but not human) serum may be
to give a clear liquid that may be coloured owing to the
used in the growth medium, but the final medium for maintaining
presence of a pH indicator.
cell growth during virus multiplication does not contain animal
Production serum. Serum and trypsin used in the preparation of cell
suspensions and culture media are shown to be free from
General provisions extraneous agents. The cell culture medium may contain a pH
The production of vaccine is based on a virus seed-lot system indicator such as phenol red and suitable antibiotics at the
and, if the virus is propagated in human diploid cells, a cell- lowest effective concentration. It is preferable to have a
bank system. Unless otherwise justified and authorized, the substrate free from antibiotics during production. Not less
virus in the final vaccine shall have undergone no more than 500 ml of the production cell culture is set aside as
passages from the master seed lot than were used to prepare uninfected cell cultures (control cells). The viral suspensions
the vaccine shown in clinical studies to be satisfactory with are harvested at a time appropriate to the strain of virus being
respect to abnormal toxicity and efficacy; even with authorized used.
exceptions, the number of passages beyond the level used for Only a single harvest that complies with the following tests
clinical studies shall not exceed five. may be used in the preparation of the final bulk vaccine.
The production method is validated to demonstrate that the
Identification
product, if tested, would comply with the tests for abnormal
toxicity and efficacy. The single harvest contains virus that is identified as measles
Substrate for virus propagation virus by serum neutralisation in cell culture, using specific
antibody.
The virus is propagated in human diploid cells or in cultures
Virus concentration. The virus concentration in the single
of chick embryo cells derived from a chicken flock free from
harvest is determined as prescribed under Assay to monitor
specified pathogens.
consistency of production and to determine the dilution to be
SEED LOT used for the final bulk vaccine.
The strain of measles virus used in the production of measles Extraneous agents (2.7.3). Complies with the test for extraneous
vaccine shall be identified by historical records that include agents.

63
MEASLES, MUMPS AND RUBELLA VACCINE (LIVE) IP 2007

Control cells. If human diploid cells are used for production, Assay
the control cells comply with the test for identification and
Titrate the vaccine for infective virus at least in triplicate,
extraneous agents.
using at least five cell cultures for each 0.5 log10 dilution step
FINAL BULK VACCINE or by a method of equal precision. Use an appropriate virus
reference preparation to validate each assay. The estimated
Virus harvests that comply with the above tests are pooled virus concentration is not less than that stated on the label;
and clarified to remove cells. A suitable stabilizer may be added the minimum virus concentration stated on the label is not
and the pooled harvests diluted as appropriate. less than 1 × 103 CCID50 per human dose. The assay is not
Only a final bulk vaccine that complies with the following valid if the confidence limits (P = 0.95) of the logarithm of the
requirements may be used in the preparation of the final lot. virus concentration is greater than ± 0.3.
Sterility (2.2.11). The final bulk vaccine complies with the test Measles vaccine (Live) RS is suitable for use as a reference
for sterility carried out using 10 ml for each medium. preparation.
Labelling. The label states (1) the strain of virus used for the
FINAL LOT
preparation of the vaccine; (2) the type and origin of the cells
A minimum virus concentration for release of the product is used for the preparation of the vaccine; (3) the minimum virus
established so as to ensure, in the light of stability data, that concentration; (4) the time within which the vaccine must be
the minimum concentration stated on the label will be present used after reconstitution.
at the end of the period of validity.
Only a final lot that complies with the tests for minimum virus
concentration for release, with the following requirement for Measles, Mumps and Rubella Vaccine
thermal stability and with each of the requirements given below
under Identification, Tests and Assay may be released for
(Live)
use. Provided that the test for bovine serum albumin has been Measles, Mumps and Rubella Vaccine (Live) is a freeze-dried
carried out with satisfactory results on the final bulk vaccine, preparation of suitable attenuated strains of measles virus,
it may be omitted on the final lot. mumps virus and rubella virus grown in suitable cell cultures.
Thermal stability. Maintain samples of the final lot of freeze- The vaccine is reconstituted immediately before use to give a
dried vaccine in the dry state at 37° for 7 days. Determine the clear liquid that may be coloured owing to the presence of a
virus concentration as described under Assay in parallel for pH indicator.
the vaccine held at 37° for 7 days and for vaccine stored at 2°
to 8°. The virus concentration of the heated vaccine is not Production
more than 1.0 log10 lower than that of the unheated vaccine.
General provisions
Identification The three components are prepared as described in the
When the vaccine reconstituted as stated on the label is mixed monographs on Measles Vaccine (Live), Mumps Vaccine (Live)
with specific measles antibodies, it is no longer able to infect and Rubella Vaccine (Live) and comply with the tests
susceptible cell cultures. prescribed therein.
The production method is validated to demonstrate that the
Tests product, if tested, would comply with the test for safety and
Water (2.3.43). Not more than 3.0 per cent, determined by Karl efficacy.
Fischer, semi-micro determination of water or by any suitable FINAL BULK VACCINE
validated method.
Virus harvests for each component are pooled and clarified to
Sterility ( 2.2.11). The reconstituted vaccine complies with remove cells. A suitable stabilizer may be added and the pooled
the test for sterility. harvests diluted as appropriate. Suitable quantities of the
Abnormal toxicity ( 2.2.1). Complies with the test for abnormal pooled harvest for each component are mixed.
toxicity. Only a final bulk vaccine that complies with the following
Bovine serum albumin. Not more than 50 ng per single human requirements may be used in the preparation of the final lot.
dose, determined by a suitable immunochemical method Sterility (2.2.11). Carry out the test for sterility using 10 ml for
(2.2.14). each medium.

64
IP 2007 MENINGOCOCCAL POLYSACCHARIDE VACCINE

FINAL LOT is not less than 1x103 CCID50 per single human dose. The
assay is not valid if the confidence limits (P = 0.95) of the
For each component, a minimum virus concentration for
logarithm of the virus concentration are greater than ± 0.3.
release of the product is established such as to ensure, in the
light of stability data, that the minimum concentration stated Measles vaccine (Live) RS is suitable for use as a reference
on the label will be present at the end of the period of validity. preparation.
Only a final lot that complies with the tests for minimum virus B. Mix the vaccine with a sufficient quantity of antibodies
concentration of each component for release, with the specific for measles virus and rubella virus. Titrate the vaccine
following requirement for thermal stability and with each of for infective mumps virus at least in triplicate, using at least
the requirements given below under Identification and Tests eight cell cultures for each dilution 0.5 log10 step or by a method
may be released for use. Provided that the test for bovine of equal precision. Use an appropriate virus reference
serum albumin has been carried out with satisfactory results preparation to validate each assay. The estimated mumps virus
on the final bulk vaccine, it may be omitted on the final lot. concentration is not less than that stated on the label; the
minimum mumps virus concentration stated on the label is not
Thermal stability. Maintain samples of the final lot of freeze-
less than 5 x 103 CCID50 per single human dose. The assay is
dried vaccine in the dry state at 37° for 7 days. Determine the
not valid if the confidence limits (P = 0.95) of the logarithm of
virus concentration as described under Assay in parallel for
the virus concentration are greater than ± 0.3.
the vaccine held at 37° for 7 days and for vaccine stored at 2 to
8°. The virus concentration of the heated vaccine is not more Mumps vaccine (Live) RS is suitable for use as a reference
than 1.0 log10 lower than that of the unheated vaccine. preparation.
C. Mix the vaccine with a sufficient quantity of antibodies
Identification specific for mumps virus. Titrate the vaccine for infective
rubella virus at least in triplicate, using at least eight cell
When the vaccine reconstituted as stated on the label is mixed
cultures for each dilution 0.5 log10 step or by a method of
with antibodies specific for measles virus, mumps virus and
equal precision. Use an appropriate virus reference preparation
rubella virus, it is no longer able to infect cell cultures
to validate each assay. The estimated rubella virus
susceptible to these viruses. When the vaccine reconstituted
concentration is not less than that stated on the label; the
as stated on the label is mixed with quantities of specific
minimum rubella virus concentration stated on the label is not
antibodies sufficient to neutralize any two viral components,
less than 1x103 CCID50 per single human dose. The assay is
the third viral component infects susceptible cell cultures.
not valid if the confidence limits (P = 0.95) of the logarithm of
Tests the virus concentration are greater than ± 0.3.

Water (2.3.43). Not more than 3.0 per cent, determined by Karl Rubella vaccine (Live) RS is suitable for use as a reference
Fischer, semi-micro determination of water or by any suitable preparation.
validated method. Labelling. The label states (1) the strains of virus used in the
Sterility (2.2.11). The reconstituted vaccine complies with the preparation of the vaccine; (2) the type and origin of the cells
test for sterility. used for the preparation of the vaccine; (3) the minimum virus
concentration for each component of the vaccine; (4) the time
Abnormal toxicity (2.2.1). Complies with the test for abnormal within which the vaccine must be used after reconstitution;
toxicity. (5) that the vaccine must not be given to a pregnant woman
Bovine serum albumin. Not more than 50 ng per single human and that a woman should not become pregnant within two
dose, determined by a suitable immunochemical method months after having the vaccine.
(2.2.14).

Assay
Meningococcal Polysaccharide
A. Mix the vaccine with a sufficient quantity of antibodies
specific for mumps virus and rubella virus.Titrate the vaccine
Vaccine
for infective measles virus at least in triplicate, using at least Meningococcal Polysaccharide Vaccine is a freeze-dried
eight cell cultures for each dilution 0.5 log10 step or by a method preparation of one or more purified capsular polysaccharides
of equal precision. Use an appropriate virus reference obtained from one or more suitable strains of Neisseria
preparation to validate each assay. The estimated measles meningitidis group A, group C, group Y and group W135 that
virus concentration is not less than that stated on the label; are capable of consistently producing polysaccharides known
the minimum measles virus concentration stated on the label to be safe and effective in man.

65
MENINGOCOCCAL POLYSACCHARIDE VACCINE IP 2007

N. meningitidis group A polysaccharide consists of partly medium. The liquid media used and the final medium are
O-acetylated repeating units of N-acetylmannosamine, linked semisynthetic and free from substances precipitated by
with 1α→6 phosphodiester bonds. cetrimonium bromide (hexadecyltrimethylammonium
N. meningitidis group C polysaccharide consists of partly bromide) and do not contain blood-group substances or high-
O-acetylated repeating units of sialic acid, linked with 2α→9 molecular-mass polysaccharides. The bacterial purity of the
glycosidic bonds. culture is verified by microscopic examination of Gram-stained
smears and by inoculation into appropriate media. The cultures
N. meningitidis group Y polysaccharide consists of partly are centrifuged and the polysaccharides precipitated from the
O-acetylated alternating units of sialic acid and D-glucose, supernatant by addition of cetrimonium bromide. The
linked with 2α→6 and 1α→4 glycosidic bonds. precipitate obtained is harvested and may be stored at or
N. meningitidis group W135 polysaccharide consists of partly below -20° awaiting further purification.
O-acetylated alternating units of sialic acid and D-galactose,
PURIFIED POLYSACCHARIDES
linked with 2α→6 and 1α→4 glycosidic bonds.
The polysaccharides are purified, after dissociation of the
Production complex of polysaccharide and cetrimonium bromide, using
suitable procedures to remove successively nucleic acids,
General provisions
proteins and lipopolysaccharides. The purification step
Production of the meningococcal polysaccharides is based consists of ethanol precipitation of the polysaccharides or
on a well defined seed-lot system. The method of production purification with chloroform and n-butanol or by cold phenol
shall have been shown to yield consistently meningococcal treatment, which are then dried and stored at or below -20°.
polysaccharide vaccines of satisfactory immunogenicity and The loss on drying is determined by thermogravimetry, Karl
safety for man. Fischer or any other suitable method and the value is used to
calculate the results of the other chemical tests with reference
The production method is validated to demonstrate that the
to the dried substance.
product, if tested, would comply with the test of abnormal
toxicity for antisera and vaccines. Only purified polysaccharides that tested comply with the
following requirements may be used in the preparation of the
SEED LOT final bulk vaccine.
The strains of N. meningitidis used for the master seed lots Protein (2.7.1). Not more than 10 mg of protein per gram of
shall be identified by historical records that include information purified polysaccharidefor group A and C organisms and less
on their origin and by their biochemical, serological, than 50 mg of protein per gram of polysaccharide for group Y
physicochemical or molecular characteristics. Cultures from and W135 calculated using bovine plasma albumin as a
the working seed lot shall have the same characteristics as the reference or other methods approved by National Regulatory
strain that was used to prepare the master seed lot. The strains Authority.
have the following characteristics: Nucleic acids (2.7.1). Not more than 10 mg of nucleic acids per
a) Colonies obtained from a culture are round, uniform in gram of purified polysaccharide, calculated with reference to
shape and smooth with a mucous, opalescent, greyish the dried substance.
appearance. O-Acetyl groups (2.7.1). Not less than 2 mmol of O-acetyl
b) Gram staining reveals characteristic Gram-negative groups per gram of purified polysaccharide for group A, not
diplococci in ‘coffee-bean’ arrangement. less than 1.5 mmol per gram of polysaccharide for group C,
not less than 0.3 mmol per gram of polysaccharide for groups
c) The oxidase test is positive.
Y and W135, all calculated with reference to the dried
d) The culture utilizes glucose and maltose. substance.
e) Suspensions of the culture agglutinate with specific Phosphorus (2.7.1). Not less than 80 mg of phosphorus per
antisera of known titre. gram of group A purified polysaccharide, calculated with
reference to the dried substance.
PROPAGATION AND HARVEST
Sialic acid (2.7.1). Not less than 800 mg of sialic acid per gram
The working seed lots are cultured on solid media that do not of group C polysaccharide and not less than 560 mg of sialic
contain blood-group substances or ingredients of mammalian acid per gram of purified polysaccharide for groups Y and
origin. The inoculum may undergo one or more subcultures in W135, all calculated with reference to the dried substance.
liquid medium before being used for inoculating the final Use the following reference solutions:

66
IP 2007 MENINGOCOCCAL POLYSACCHARIDE VACCINE

Group C polysaccharide. A 150 mg/l solution of N- Only a final bulk vaccine that complies with the following
acetylneuraminic acid. requirement may be used in the preparation of the final lot.
Group Y polysaccharide. A solution containing 95 mg/l of N- Sterility (2.2.11). Carry out test for sterility using 10 ml of
acetylneuraminic acid and 55 mg/l of glucose. bulk for each sterility medium.
Group W135 polysaccharide. A solution containing 95 mg/l FINAL LOT
of N-acetylneuraminic acid and 55 mg/l of galactose.
The final bulk vaccine is distributed aseptically into sterile
Calcium. If a calcium salt is used during purification, containers. The containers are then closed so as to avoid
determination of calcium is carried out on the purified contamination. Only a final lot that is satisfactory with respect
polysaccharide by a suitable method; the content is within to each of the requirements prescribed below under
the limits approved for the product. Identification, Tests and Assay may be released for use.
Molecular size. Examine by gel filtration or high performance Identification
size-exclusion chromatography (HPSEC) (2.4.16), using
Carry out an identification test for each polysaccharide present
agarose for chromatography or cross-linked agarose for
in the vaccine by a suitable immunochemical method (2.2.14).
chromatography either alone or in combination with light
scattering and refractive index detector (e.g. multiple angle Tests
LASER light scattering, MALLS) or any other suitable method.
Use a column 0.9 m x 15 mm equilibrated with a solvent having Sterility (2.2.11). Complies with the test for sterility.
an ionic strength of 0.2 mol/kg and a pH of 7.0 to 7.5. Apply to Abnormal toxicity (2.2.1). Complies with the test for abnormal
the column about 2.5 mg of polysaccharide in a volume of toxicity.
about 1.5 ml and elute at about 20 ml/h. Collect fractions of
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
about 2.5 ml and determine the content of polysaccharide by
each of the rabbit with 1ml per kg body weight of solution
a suitable method.
containing
At least 65.0 per cent of group A polysaccharide, 75.0 per cent
a) 0.025 µg of polysaccharide for a monovalent vaccine,
of group C polysaccharide, 80.0 per cent of group Y
polysaccharide and 80.0 per cent of group W135 b) 0.050 µg of polysaccharide for a bivalent vaccine,
polysaccharide is eluted before a distribution coefficient (K0) c) 0.075 µg of polysaccharide for a trivalent vaccine,
of 0.50 is reached. In addition, the percentages eluted before
this distribution coefficient are within the limits approved for d) 0.100 µg of polysaccharide for a tetravalent vaccine.
the particular product. Water (2.3.43). Not more than 3.0 per cent, of moisture content
by thermogravimetery, Karl Fischer or any other suitable
Identification and serological specificity method.
The identity and serological specificity are determined by a Molecular size. Examine by gel filtration or size-exclusion
suitable immunochemical method (2.2.14). Identity and purity chromatography (2.4.16). Use a column about 0.9 m long and
of each polysaccharide shall be confirmed; it shall be shown 16 mm in internal diameter equilibrated with a solvent having
that there is not more than 1.0 per cent m/m of group- an ionic strength of 0.2 mol/kg and a pH of 7.0 to 7.5. Apply to
heterologous N. meningitidis polysaccharide. the column about 2.5 mg of each polysaccharide in a volume
of about 1.5 ml and elute at about 20 ml/h. Collect fractions of
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
about 2.5 ml and determine the content of polysaccharide by
each of the rabbit with 1ml per kg body weight of solution
a suitable method.
containing
Use cross-linked agarose for chromatography and apply a
a) 0.025 µg of polysaccharide for a monovalent vaccine,
suitable immunochemical method (2.2.14) to establish the
b) 0.050 µg of polysaccharide for a bivalent vaccine, elution pattern of the different polysaccharide(s). The vaccine
c) 0.075 µg of polysaccharide for a trivalent vaccine, complies with the test if:
a) 65.0 per cent of Group A polysaccharide is eluted before
d) 0.100 µg of polysaccharide for a tetravalent vaccine.
K0 of 0.50,
FINAL BULK VACCINE b) 75.0 per cent of Group C polysaccharide is eluted before
One or more purified polysaccharides of one or more N. K0 of 0.50,
meningitidis groups are dissolved in a suitable solvent that c) 80.0 per cent of Group Y & W135 polysaccharide is eluted
may contain a stabilizer. before K0 of 0.50.

67
MUMPS VACCINE (LIVE) IP 2007

For a tetravalent vaccine (group A + group C + group Y and shown in clinical studies to be satisfactory with respect to
group W135), use cross linked agarose for chromatography safety and efficacy even with authorized exceptions, the
R1 and apply a suitable immunochemical method (2.2.14) to number of passages beyond the level used for clinical studies
establish the elution pattern of the different polysaccharides. shall not exceed five.
The vaccine complies with the test if K0 for the principal peak The production method is validated to demonstrate that the
is product, if tested, would comply with the tests for safety and
a) not greater than 0.70 for group A and group C efficacy.
polysaccharide,
Substrate for virus propagation
b) not greater than 0.57 for group Y polysaccharide,
The virus is propagated in human diploid cells or in primary
c) not greater than 0.68 for group W135 polysaccharide. cultures of chick embryo cells derived from a chicken flock
Assay free from specified pathogens.
Carry out an assay as stated under each polysaccharide SEED LOT
present in the vaccine.
The strain of mumps virus used shall be identified by historical
For a divalent vaccine (group A + group C), use measurement records that include information on the origin of the strain
of phosphorus (2.7.1) to determine the content of and its subsequent manipulation. To avoid unnecessary use
polysaccharide A and measurement of sialic acid (2.7.1) to of monkeys in the test for neurovirulence, Virus seed lots are
determine the content of polysaccharide C. To determine sialic prepared in large quantities and stored at temperatures below
acid, use as reference solution a 150 mg/l solution of N- -20° if freeze-dried, or below -60° if not freeze-dried.
acetylneuraminic acid.
Only a seed lot that complies with the following tests may be
For a tetravalent vaccine (group A + group C + group Y +
used for virus propagation.
group W135) a suitable immunochemical method (2.2.14) is
used with a reference preparation of purified polysaccharide Identification
for each group.
The master and working seed lots are identified as mumps
The vaccine contains not less than 70.0 per cent and not more virus by serum neutralisation in cell culture, using specific
than 130.0 per cent of the quantity of each polysaccharide antibodies.
stated on the label.
Virus concentration. The virus concentration of the master
Labelling. The label states (1) the group or groups of and working seed lots is determined to ensure consistency of
polysaccharides (A, C, Y or W135) present in the vaccine; (2) production.
the number of µg of polysaccharide per human dose.
Extraneous agents (2.7.3). The working seed lot complies with
the tests for seed lots.
Neurovirulence (2.7.5). The master/working seed lot complies
Mumps Vaccine (Live) with the test for neurovirulence of live virus vaccines. Macaca
Mumps Vaccine (Live) is a freeze-dried preparation of a suitable and Cercopithecus monkeys are suitable for the test.
attenuated strain of mumps virus. The vaccine is reconstituted
immediately before use to give a clear liquid that may be PROPAGATION AND HARVEST
coloured owing to the presence of a pH indicator. All processing of the cell bank and subsequent cell cultures is
done under aseptic conditions in an area where no other cells
Production
are handled. Suitable animal (but not human) serum may be
General provisions used in the growth media. Serum and trypsin used in the
preparation of cell suspensions and culture media are shown
The production of vaccine is based on a virus seed-lot system
to be free from extraneous agents. The cell culture medium
and, if the virus is propagated in human diploid cells, a cell-
may contain a pH indicator such as phenol red and suitable
bank system. The production method shall have been shown
antibiotics at the lowest effective concentration. It is preferable
to yield consistently live mumps vaccines of adequate
to have a substrate free from antibiotics during production.
immunogenicity and safety in man.
Not less than 500 ml of the production cell culture is set aside
Unless otherwise justified and authorised, the virus in the as uninfected cell culture (control cells). The viral suspensions
final vaccine shall have undergone no more passages from are harvested at a time appropriate to the strain of virus being
the master seed lot than were used to prepare the vaccine used.

68
IP 2007 PERTUSSIS VACCINE

Only a single harvest that complies with the following Tests


requirements may be used in the preparation of the final bulk
Water (2.3.43). Not more than 3.0 per cent, determined by Karl
vaccine.
Fischer, semi-micro determination of water or by any suitable
Identification validated method.
Sterility ( 2.2.11). Complies with the test for sterility.
The single harvest contains virus that is identified as mumps
virus by serum neutralization in cell culture, using specific Abnormal toxicity (2.2.1). Complies with the test for abnormal
antibodies. toxicity.
Virus concentration. The virus concentration in the single Bovine serum albumin. Not more than 50 ng per single human
harvest is determined as prescribed under Assay to monitor dose, determined by a suitable immunochemical method
consistency of production and to determine the dilution to be (2.2.14).
used for the final bulk vaccine. Assay
Sterility (2.2.11). Single harvest complies with sterility test
Titrate the vaccine for infective virus at least in triplicate,
should be processed further.
using at least five cell cultures for each 0.5 log10 dilution step
Control cells. The control cells comply with a test for or by a method of equal precision. Use an appropriate virus
extraneous agents (2.7.3). reference preparation to validate each assay. The estimated
virus concentration is not less than that stated on the label;
FINAL BULK VACCINE
the minimum virus concentration stated on the label is not
Single harvests that comply with the above tests are pooled less than 5 x 103 CCID50 per human dose. The assay is not
and clarified to remove cells. A suitable stabiliser may be added valid if the confidence limits (P = 0.95) of the logarithm of the
and the pooled harvests diluted as appropriate. virus concentration is greater than 0.3.
Only a final bulk vaccine that complies with the following Mumps vaccine (Live) RS is suitable for use as a reference
requirements may be used in the preparation of the final lot. preparation.
Sterility (2.2.11). The final bulk vaccine complies with the test Labelling. The label states (1) the strain of virus used for the
for sterility, carried out using 10 ml for each medium. preparation of the vaccine; (2) the type and origin of the cells
used for the preparation of the vaccine; (3) the minimum virus
FINAL LOT concentration and; (4) the time within which the vaccine must
A minimum virus concentration for release of the product is be used after reconstitution.
established such as to ensure, in the light of stability data,
that the minimum concentration stated on the label will be
present at the end of the period of validity.
Pertussis Vaccine
Only a final lot that complies with the tests for minimum virus
concentration for release, with the following requirement for Pertussis Vaccine is a sterile saline suspension of inactivated
thermal stability and with each of the requirements given below whole cells of one or more strains of Bordetella pertussis.
under Identification and Tests and Assay may be released for Production
use. Provided that the test for bovine serum albumin has been
carried out with satisfactory results on the final bulk vaccine, General provisions
it may be omitted on the final lot.
Inactivated B. pertussis suspension
Thermal stability. Maintain samples of the final lot of freeze-
dried vaccine in the dry state at 37° for 7 days. Determine the Production is based on a seed-lot system. One or more strains
virus concentration as described under Assay in parallel for of B. pertussis with known origin and history are used. Strains,
the vaccine held at 37° for 7 days and for vaccine stored at 2° culture medium and cultivation method are chosen in such a
to 8°. The virus concentration of the vaccine exposed to 37° way that agglutinogens 1, 2 and 3 are present in the final
for 7 days is not more than 1.0 log10 lower than that of the vaccine. Each strain is grown for 24 to 72 hours in a liquid
unheated vaccine. medium or on a solid medium; the liquid medium used in the
final cultivation stage does not contain blood or blood
Identification products. Human blood or blood products are not used in any
When the vaccine is reconstituted as stated on the label is culture media. The bacteria are harvested, washed to remove
mixed with specific mumps antibodies, it is no longer able to substances derived from the medium and suspended in a
infect susceptible cell cultures. 0.9 per cent w/v solution of sodium chloride or other suitable

69
PERTUSSIS VACCINE IP 2007

isotonic solution. The opacity of the suspension is determined to 16 g for the vaccine group and for the saline control. Use
not later than 2 weeks after harvest by comparison with the mice of the same sex or distribute males and females equally
reference preparation of Opacity and used as the basis of between the groups. Allow the animals access to food and
calculation for subsequent stages in vaccine preparation. water for at least 2 hours before injection and during the test.
Single harvests are not used for the final bulk vaccine unless Inject each mouse of the vaccine group intraperitoneally with
they have been shown to contain B. pertussis cells with the 0.5 ml, containing a quantity of the vaccine equivalent to not
same characteristics with regard to growth and agglutinogens, less than half the single human dose. Inject each mouse of the
as the parent strain and to be free from contaminating bacteria control group with 0.5 ml of a 0.9 per cent w/v sterile solution
and fungi. The bacteria are killed and detoxified in controlled of sodium chloride, preferably containing the same amount
conditions by means of a suitable chemical agent or by heating of antimicrobial preservative as that injected with the vaccine.
or by a combination of these methods. Freedom from live B. Weigh the groups of mice immediately before the injection, 72
pertussis is tested using a suitable culture medium. The hours and 7 days after the injection. The vaccine complies
suspension is maintained at 5 + 3° for a suitable period to with the test if (a) at the end of 72 h the total mass of the group
diminish its toxicity. of vaccinated mice is not less than that preceding the injection;
FINAL BULK VACCINE (b) at the end of 7 days the average increase in mass per
vaccinated mouse is not less than 60.0 per cent of that per
Suitable quantities of the inactivated single harvests are pooled control mouse; and (c) not more than 5.0 per cent of the
to prepare the final bulk vaccine. Suitable antimicrobial vaccinated mice die during the test. The test may be repeated
preservatives may be added. The bacterial concentration of and the results of the tests combined.
the final bulk vaccine does not exceed that corresponding to
an opacity of 20 IU per single human dose. If 2 or more strains Free formaldehyde (2.3.20). Maximum 0.2 g/l.
of B. pertussis are used, the composition of consecutive lots Antimicrobial preservative. Where applicable, determine the
of the final bulk vaccine shall be consistent with respect to amount of antimicrobial preservative by a suitable chemical
the proportion of each strain as measured in opacity units. method. The content is not less than 85.0 per cent and not
more than 115.0 per cent of the intended amount.
Only a final bulk vaccine that complies with the following
requirements may be used in the preparation of the final lot. Sterility (2.2.11). Complies with the test for sterility.
Antimicrobial preservative. Where applicable, determine the Assay
amount of antimicrobial preservative by a suitable chemical Carry out the assay of Pertussis Vaccine as described below :
method. The amount is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount. Biological assay of pertussis vaccine

Sterility (2.2.11). Carry out test for sterility using 10 ml of The potency of PertussisVaccine is determined by comparison
bulk for each sterility medium. of the dose necessary to protect mice against the effects of a
lethal dose of Bordetella pertussis challenge culture,
FINAL LOT administered intracerebrally, with the dose of a reference
The final bulk vaccine is distributed aseptically into sterile, preparation, calibrated in International Units, required to give
tamper-proof containers. The containers are closed so as to the same level of protection. For this comparison, the Standard
prevent contamination. preparation of Pertussis vaccine & a suitable strain of B.
pertussis (e.g.18323, to be used as challenge strain), are
Only a final lot that is satisfactory with respect to each of the required.
requirements given below under Identification, Tests and
Assay may be released for use. Provided the tests for specific Reference preparation
toxicity, free formaldehyde and antimicrobial preservative and The reference preparation is an International standard of
the assay have been carried out with satisfactory results on Pertussis vaccine, consisting of a freeze dried vaccine or
the final bulk vaccine, they may be omitted on the final lot. another suitable preparation, calibrated in comparison to
International standard, from time to time. The International
Identification Unit is the activity contained in a stated amount of the
Identify pertussis vaccine by agglutination of the bacteria in International standard, which consists of a quantity of dried
the vaccine by antisera specific to B. pertussis. pertussis vaccine. The equivalence in International Units of
the International Standard is stated by the World Health
Tests Organization.
Specific toxicity Use healthy mice of a suitable strain, weighing between 13
Use not less than 10 healthy mice each weighing between 14 and 16 g from the same stock. Distribute the mice randomly, in

70
IP 2007 PNEUMOCOCCAL POLYSACCHARIDE VACCINE

six to eight groups of not less than 16 and not more than 24 The test is not valid unless: a) for both the vaccine under
and four groups of 10. The mice should all be of the same sex examination and the reference preparation, the ED50 (protective
or the males and females should be distributed equally between dose), lies between the largest and the smallest doses given
the groups. Half of the groups of 16 to 24 should receive the to the mice; b) the number of animals, which die in the four
reference preparation and the other half should receive the groups of 10 injected with the challenge suspension and its
vaccine under examination. The four groups of 10 each should dilutions indicate that the challenge dose contains 100 to 1000
be used for the LD50 titration of challenge suspension. LD50 and 1 LD50 contains not more than 300 colony forming
Use at least, three dilutions of the reference vaccine and similar units; c) and the statistical analysis shows no deviation from
dilutions of the vaccine under examination. In each case the linearity or parallelism, in terms of significance of the scope of
dilutions are so selected that the dilution protecting 50 per dose response curve; d) the vaccine passes the requirements
cent of the mice (ED50) is as near as possible to middle of the for potency, if the test results of a statistically valid assay
dilution range. For example, suggested dilutions are (1/8, 1/40 show that the estimated potency of the vaccine is not less
and 1/200 of the human dose of the vaccine under examination) than 4.0 IU per single human dose and the lower fiducial limit
and (0.5 IU, 0.1 IU and 0.02 IU or any other suitable (P = 0.95) of estimated potency is not less than 2.0 IU.
standardized dilutions, of the reference preparation), each dose The test may be repeated once, but when more than one test
being contained in a volume, not exceeding 0.5 ml. For each is performed the results of all valid tests must be combined, in
dilution use 16 to 24 mice and inject intraperitoneally, into the estimate of potency.
each mouse one dose of the dilution.
Labelling. The label states (1) the minimum number of
Select a suitable strain of B. pertussis (e.g. 18323), capable of International Units per single human dose; (2) that the vaccine
causing the death of mice within 14 days of intracerebral must be shaken before use; (3) that the vaccine is not to be
injection. Make two subcultures after reviving the strain on a frozen.
suitable medium (e.g. B.G. medium) and suspend the harvested
growth in a solution containing 1 per cent w/v of casein
hydrolysate (e.g. casamino acid) and 0.85 per cent w/v of Pneumococcal Polysaccharide Vaccine
sodium chloride and having a pH of 7.0 to 7.2 or in another
suitable solution. Determine the opacity of the suspension by Pneumococcal Polysaccharide Vaccine consists of a mixture
using 5th International reference preparation for opacity (10 of equal parts of purified capsular polysaccharide of various
OU) and/or spectophotometerically. Alternatively, aliquots of serotype antigens prepared from suitable pathogenic strains
challenge suspension frozen in liquid nitrogen with a suitable of Streptococcus pneumoniae in different desired
preservative like 10 per cent DMSO may be used, to avoid combinations whose capsules have been shown to be made
heterogenicity. After 14 to 17 days of immunization, inject up of polysaccharides that are capable of inducing satisfactory
intracerebrally, a dose of 0.02 to 0.03 ml of the challenge levels of specific antibodies in man. It contains upto 23
dilution randomly, into each immunized mouse. The challenge immunochemically different capsular polysaccharides listed
should contain, approximately 1,00,000 organisms and 100 to in the Table 1.
1000 LD50 per dose, in a volume of not more than 0.03 ml. In the
Production
same way, inject 4 groups of 10 control mice each, for LD50
titration of challenge preparation, prepared by a series of General provisions
dilutions, from the dilution selected for challenge. The
challenge should be completed within 2 to 2.5 hours of Production of the vaccine is based on a well defined seed-lot
preparation. Exclude any mouse from consideration, that dies system for each type. The production method shall have been
within 3 days of challenge. Count the number of mice surviving shown to yield consistently pneumococcal polysaccharide
in each of the groups, after 14 days. On the basis of the numbers vaccines of acceptable immunogenicity and safety in man.
of animals surviving in each of the groups of 16 to 24 mice, The production method is validated to demonstrate that the
calculate the potency of vaccine under examination, against product, if tested, would comply with the tests for abnormal
the potency of reference preparation. Seed a suitable highest toxicity of vaccines for human use, modified as follows for
dilution of the challenge suspension, into each of two B.G the tesonn guinea-pig; inject 10 human dose into each guinea-
medium plates, before and after challenge. Incubate the plates pig and observe for 12 days.
at 37° for 48 to 72 hours and calculate the number of colony
forming units (CFUs). Monovalent bulk polysaccharides

Calculate the potency of the vaccine by Probit analysis and The bacteria are grown in a suitable liquid medium that does
LD 50 of the challenge suspension by Reed and Munch not contain blood-group substances or high-molecular-mass
Method. polysaccharides. The bacterial purity of the culture is verified

71
PNEUMOCOCCAL POLYSACCHARIDE VACCINE IP 2007

Table 1- Specifications on monovalent bulk polysaccharides (per cent contents):


Molecular Proteins Nucleic Total Phosphorus Molecular size K0 Uronic Hexo- Methyl- O-acetyl
Type* acids Nitrogen CL-4B** CL-2B*** acids samines pentoses Groups
1 <2 <2 3.5-6 0-1.5 < 0.15 ≥ 45 ≥ 1.8
2 <2 <2 0-1 0-1.0 < 0.15 ≥ 15 ≥ 38
3 <5 <2 0-1 0-1.0 < 0.15 ≥ 40
4 <3 <2 4-6 0-1.5 < 0.15 ≥ 40
5 < 7.5 <2 2.5-6.0 <2 < 0.60 ≥ 12 ≥ 20
6B <2 <2 0-2 2.5-5.0 < 0.50 ≥ 15
7F <5 <2 1.5-4.0 0-1.0 < 0.20 ≥ 13
8 <2 <2 0-1 0-1.0 < 0.15 ≥ 25
9N <2 <1 2.2-4 0-1.0 < 0.20 ³ 20 ≥ 28
9V <2 <2 0.5-3 0-1.0 < 0.45 ³ 15 ≥ 13
10A <7 <2 0.5-3.5 1.5-3.5 < 0.65 ≥ 12
11A <3 <2 0-2.5 2.0-5.0 < 0.40 ≥9
12F <3 <2 3-5 0-1.0 < 0.25 ≥ 25
14 <5 <2 1.5-4 0-1.0 < 0.30 ≥ 20
15B <3 <2 1-3 2.0-4.5 < 0.55 ≥ 15
17A or 17F <2 <2 0-1.5 0-3.5 < 0.45 ≥ 20
18C <3 <2 0-1 2.4-4.9 < 0.15 ≥ 14
19A <2 <2 0.6-3.5 3.0-7.0 < 0.45 ≥ 12 ≥ 20
19F <3 <2 1.4-3.5 3.0-5.5 < 0.20 ≥ 12.5 ≥ 20
20 <2 <2 0.5-2.5 1.5-4.0 < 0.60 ≥ 12
22F <2 <2 0-2 0-1.0 < 0.55 ≥ 15 ≥ 25
23F <2 <2 0-1 3.0-4.5 < 0.15 ≥ 37
33F < 2.5 <2 0-2 0-1.0 < 0.50
* The different types are indicated using the Danish nomenclature
** Cross linked agarose for chromatography R
*** Cross linked agarose for chromatography R1

and the culture is inactivated with phenol. Impurities are determined by the methods prescribed below, are shown in
removed by such techniques as fractional precipitation, the Table 1.
enzymatic digestion and ultrafiltration. The polysaccharide is The purified polysaccharides comply with the following tests
obtained by fractional precipitation, washed, and dried in a as applicable:
vacuum to a residual moisture content shown to be favourable
to the stability of the polysaccharide. The residual moisture Protein (2.7.1). Comply with the test for protein.
content is determined by drying under reduced pressure over Nucleic acids (2.7.1). Comply with the test for nucleic acids.
diphosphorus pentoxide or by thermogravimetric analysis and
Total nitrogen (2.3.30). Comply with the test for total nitrogen.
the value obtained is used to calculate the results of the tests
shown below with reference to the dried substance. The Phosphorus (2.7.1). Comply with the test for phosphorus.
monovalent bulk polysaccharide is stored at a suitable
Uronic acids (2.7.1). Comply with the test for uronic acids.
temperature in conditions that avoid the uptake of moisture.
Hexosamine (2.7.1). Comply with the test for hexosamine.
Only a monovalent bulk polysaccharide that complies with
the following requirements may be used in the preparation of Methylpentoses (2.7.1). Comply with the test for
the final bulk vaccine. Percentage contents of components, methylpentoses.

72
IP 2007 POLYSACCHARIDE VACCINE (INACTIVATED)

O-Acetyl groups (2.7.1). Comply with the test for O-acetyl on each monovalent bulk polysaccharide used in the
groups. preparation of the final lot.
Sterility (2.2.11). Comply with the test for sterility. Identification
Molecular size. Molecular size is determined by gel filtration The assay also serves to identify the vaccine.
or high performance size-exclusion chromatography (HPSEC)
(2.4.16) using cross linked Agarose for chromatography R or Tests
chromatography Agarose for chromatograph R1, either alone Sterility (2.2.11). Complies with the test for sterility.
or Multiple angle light laser scattering (MALLS) or any other
suitable method. Abnormal Toxicity (2.2.1). Complies with the test for abnormal
toxicity with the following modifications.
Identification
Inject 10 human doses each in two guinea pigs weighing
Confirm the identity of the monovalent bulk polysaccharide between 250 and 350 g by intraperitoneal route and observe
by immunochemical method (2.2.14)(except for polysaccharides for 12 days,
7F, 14 and 33F).
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
Specificity each of the rabbit with 1 ml of a dilution of the vaccine
For establishing the specificity, no reaction should occur, containing 2.5 µg/ml of each polysaccharide.
when the antigens are tested against all the antisera specific Antimicrobial preservative. Where applicable, determine the
for the other polysaccharides of the vaccine, including factor amount of antimicrobial preservative by a suitable chemical
sera for distinguishing types within groups. The method. The content is not less than 85.0 per cent and not
polysaccharides are tested at a concentration of 50 µg/ml using greater than 115.0 per cent of the intended amount.
a method capable of detecting 0.5 µg/ml. Phenol (2.3.36). Not more than 2.5 g/l.
FINAL BULK VACCINE pH (2.4.24). 4.5 to 7.4.
The final bulk vaccine is obtained by aseptically mixing the Assay
different polysaccharide powders. The uniform mixture is
aseptically dissolved in a suitable isotonic solution so that Determine the content of each polysaccharide by a suitable
one human dose of 0.5 ml contains 25 µg of each biochemical, physicochemical or immunochemical method
polysaccharide. An antimicrobial preservative may be added. (2.2.14), using antisera specific for each polysaccharide
The solution is sterilized by filtration through a bacteria- contained in the vaccine, including factor sera for types within
retentive filter. groups, and purified polysaccharides of each type as
standards.
Only a final bulk vaccine that complies with the following
requirements may be used in the preparation of the final lot. The vaccine contains not less than 70.0 per cent and not more
than 130.0 per cent of the quantity stated on the label for each
Antimicrobial preservative. Where applicable, determine the polysaccharide. The confidence interval (P = 0.95) of the assay
amount of antimicrobial preservative by a suitable chemical is not less than 80.0 and not more than 120.0 per cent of the
method. The content is not less than 85.0 per cent and not estimated content.
greater than 115.0 per cent of the intended amount.
Labelling. The label states (1) the number of µg of each
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk polysaccharide per human dose; (2) the total amount of
for each sterility medium. polysaccharide in the container.
FINAL LOT
The final bulk vaccine is distributed and filled aseptically into
sterile containers (vials or ampoules). Only a final lot that is Poliomyelitis Vaccine (Inactivated)
satisfactory with respect to each of the requirements given
below under Identification, Tests and Assay may be released Poliomyelitis Vaccine (Inactivated) is a liquid preparation of
for use. Provided that the tests for phenol and for antimicrobial suitable strains of human polioviruses 1, 2 and 3 grown in
preservative have been carried out with satisfactory results suitable cell cultures and inactivated by a validated method.
on the final bulk vaccine, they may be omitted on the final lot. Production
When consistency of production has been established on a
suitable number of consecutive batches, the assay may be General provisions
replaced by a qualitative test that identifies each The production method should consistently yield vaccines
polysaccharide, provided that an assay has been performed of acceptable safety and immunogenicity in man.

73
POLYSACCHARIDE VACCINE (INACTIVATED) IP 2007

Production of the vaccine is based on a virus seed-lot system. (Cercopithecine herpesvirus 1).
Cell lines are used according to a cell-bank system. If primary, Monkeys from which kidneys are to be removed are thoroughly
secondary or tertiary monkey kidney cells are used, production examined, particularly for evidence of tuberculosis and
complies with the requirements indicated below. herpesvirus B (Cercopithecine herpesvirus 1) infection. If a
Unless otherwise justified and authorised, the virus in the monkey shows any pathological lesion relevant to the use of
final vaccine shall not have undergone more passages from its kidneys in the preparation of a seed lot or vaccine, it is not
the master seed lot than was used to prepare the vaccine to be used nor are any of the remaining monkeys of the group
shown in clinical studies to be satisfactory with respect to concerned unless it is evident that their use will not impair the
safety and efficacy. safety of the product.
The production method is validated to demonstrate that the All the operations described in this section are conducted
product, if tested, would comply with the test for safety and outside the area where the vaccine is produced.
efficacy. Monkey cell cultures for vaccine production. Kidneys that
show no pathological signs are used for preparing cell cultures.
Substrate for virus propagation
Each group of cell cultures derived from a single monkey forms
The virus is propagated in a human diploid cell line (2.7.2), in a separate production cell culture giving rise to a separate
a continuous cell line (2.7.2) or in primary, secondary or tertiary single harvest.
monkey kidney cells.
The primary monkey kidney cell suspension complies with
Primary, secondary or tertiary monkey kidney cells. The the test for mycobacteria ; disrupt the cells before carrying
following special requirements for the substrate for virus out the test.
propagation apply to primary, secondary or tertiary monkey If secondary or tertiary cells are used, it shall be demonstrated
kidney cells. by suitable validation tests that cell cultures beyond the
Monkeys used in the preparation of kidney cell cultures for passage level used for production are free from tumorigenicity.
production and control of the vaccine. The animals used are
SEED LOT
of a species approved by the competent authority, in good
health and, unless otherwise justified and authorised, have Each of the three strains of poliovirus used shall be identified
not been previously employed for experimental purposes. by historical records that include information on the origin of
Kidney cells used for vaccine production and control are the strain and its subsequent manipulation.
derived from monitored, closed colonies of monkeys bred in
Only a working seed lot that complies with the following
captivity, not from animals caught in the wild; a previously
requirements may be used for virus propagation.
approved seed lot prepared using virus passaged in cells from
wild monkeys may, subject to approval by the competent Identification
authority, be used for vaccine production if historical data on
safety justify this. Each working seed lot is identified as human poliovirus 1, 2 or
3 by virus neutralisation in cell culture using specific
Monitored, closed colonies of monkeys. The monkeys are antibodies.
kept in groups in cages. Freedom from extraneous agents is
achieved by the use of animals maintained in closed colonies Virus concentration. The virus concentration of each
that are subject to continuous and systematic veterinary and working seed lot is determined to define the quantity
laboratory monitoring for the presence of infectious agents. of virus to be used for inoculation of production cell
The supplier of animals is certified by the competent authority. cultures.
Each monkey is tested serologically at regular intervals during
Extraneous agents (2.7.3). The working seed lot complies with
a quarantine period of not less than 6 weeks imposed before
the requirements for seed lots for virus vaccines. In addition,
entering the colony and then during its stay in the colony.
if primary, secondary or tertiary monkey kidney cells have
The monkeys used are shown to be tuberculin-negative been used for isolation of the strain, measures are taken to
and free from antibodies to simian virus 40 (SV40) and ensure that the strain is not contaminated with simian viruses
simian immunodeficiency virus. If Macaca spp. monkeys are such as simian immunodeficiency virus, simian virus 40,
used for production, the monkeys are also shown to be free filoviruses and herpesvirus B (Cercopithecine herpesvirus 1).
from antibodies to herpesvirus B (Cercopithecine herpesvirus A working seed lot produced in primary, secondary or tertiary
1) infection. Human herpesvirus 1 has been used as an monkey kidney cells complies with the requirements given
indicator for freedom from herpesvirus B antibodies on below under Virus Propagation and Harvest for single harvests
account of the danger of handling herpesvirus B produced in such cells.

74
IP 2007 POLYSACCHARIDE VACCINE (INACTIVATED)

PROPAGATION AND HARVEST for non-specific, accidental reasons.


All processing of the cell bank and subsequent cell cultures is Identification
done under aseptic conditions in an area where no other cells
or viruses are being handled. Approved animal serum (but not The single harvest is identified as containing human poliovirus
human serum) may be used in the cell culture media. Serum 1, 2 or 3 by virus neutralisation in cell cultures using specific
and trypsin used in the preparation of cell suspensions and antibodies.
media are shown to be free from extraneous agents. The cell Virus concentration. The virus concentration of each single
culture media may contain a pH indicator such as phenol red harvest is determined by titration of infectious virus in cell
and approved antibiotics at the lowest effective concentration. cultures.
Not less than 500 ml of the cell cultures employed for vaccine
production is set aside as uninfected cell cultures (control Sterility (2.2.11). The single harvest complies with the test for
cells); where continuous cell lines in a fermenter are used for sterility, carried out using 10 ml for each medium.
production, 200 × 106 cells are set aside to prepare control Mycoplasmas (2.7.4). The single harvest complies with the
cells; where primary, secondary or tertiary monkey kidney test for mycoplasmas, carried out using 10 ml.
cells are used for production, a cell sample equivalent to at
least 500 ml of the cell suspension, at the concentration Test in rabbit kidney cell cultures. Where primary, secondary
employed for vaccine production, is taken to prepare control or tertiary monkey kidney cells are used for production, test a
cell cultures. sample of at least 10 ml of the single harvest for the absence of
herpesvirus B (Cercopithecine herpesvirus 1) and other viruses
Only a single harvest that complies with the following in rabbit kidney cell cultures as described for the control cells.
requirements may be used in the preparation of the vaccine.
The tests for Identification and Sterility may be carried out Test in Cercopithecus kidney cell cultures. Where primary,
instead on the purified, pooled monovalent harvest. After secondary or tertiary monkey kidney cells are used for
demonstration of consistency of production at the stage of production, test a sample of at least 10 ml of the single harvest
the single harvest, the test for virus concentration may be for the absence of SV40 virus and other extraneous agents.
carried out instead on the purified, pooled monovalent harvest. Neutralise the sample by a high-titre antiserum against the
specific type of poliovirus. Test the sample in primary
Control cells. The control cells of the production cell culture cercopithecus kidney cell cultures or cells that have been
comply with a test for Identification (if a cell-bank system is demonstrated to be at least as susceptible for SV40. Incubate
used for production) and with the requirements for extraneous the cultures at 37° and observe for 14 days. At the end of this
agents, where primary, secondary or tertiary monkey kidney period, make at least one subculture of fluid in the same cell
cells are used, the tests in cell cultures are carried out as culture system and observe both primary cultures and
shown below under Test in Rabbit Kidney Cell Cultures and subcultures for an additional 14 days.
Test in Cercopithecus Kidney Cell Cultures).
PURIFICATION AND PURIFIED MONOVALENT
Test in rabbit kidney cell cultures. Test a sample of at least 10
HARVEST
ml of the pooled supernatant fluid from the control cultures
for the absence of herpesvirus B (Cercopithecine herpesvirus Several single harvests of the same type may be pooled and
1) and other viruses in rabbit kidney cell cultures. The dilution may be concentrated. The monovalent harvest or pooled
of supernatant in the nutrient medium is not greater than 1:4 monovalent harvest is purified by validated methods. If
and the area of the cell layer is at least 3 cm2 per ml of inoculum. continuous cell lines are used for production, the purification
Set aside one or more containers of each batch of cells with process shall have been shown to reduce consistently the
the same medium as non-inoculated control cells. Incubate content of substrate-cell DNA to not more than 500 pg per
the cultures at 37° and observe for at least 2 weeks. The test is single human dose.
not valid if more than 20 per cent of the control cells are
Only a purified monovalent harvest that complies with the
discarded for non-specific, accidental reasons.
following requirements may be used for the preparation of the
Test in Cercopithecus kidney cell cultures. Test a sample of inactivated monovalent harvest.
at least 10 ml of the pooled supernatant fluid from the control
cultures for the absence of SV40 virus and other extraneous Identification
agents by inoculation onto cell cultures prepared from the
The virus is identified by virus neutralisation in cell cultures
kidneys of cercopithecus monkeys, or other cells shown to be
using specific antibodies or by determination of D-antigen.
at least as sensitive for SV40, by the method described under
Test in Rabbit Kidney Cell Cultures. The test is not valid if Virus concentration. The virus concentration is determined
more than 20 per cent of the control cell cultures are discarded by titration of infectious virus.

75
POLYSACCHARIDE VACCINE (INACTIVATED) IP 2007

Specific activity. The ratio of the virus concentration or the D- suitable immunochemical method (2.2.14) is within the limits
antigen content, determined by a suitable immunochemical approved for the particular preparation.
method (2.2.14). to the total protein content (specific activity)
of the purified monovalent harvest is within the limits approved FINAL BULK VACCINE
for the particular product. The final bulk vaccine is prepared directly from the inactivated
monovalent harvests of human polioviruses 1, 2 and 3 or from
INACTIVATION AND INACTIVATED MONOVALENT
a trivalent pool of inactivated monovalent harvests. If a
HARVEST
trivalent pool of inactivated monovalent harvests is used, a
Several purified monovalent harvests of the same type may test for effective inactivation is carried out on this pool instead
be mixed before inactivation. To avoid failures in inactivation of on the final bulk vaccine. A stabiliser and an antimicrobial
caused by the presence of virus aggregates, filtration is carried preservative may be added.
out before and during inactivation; inactivation is started
Only a final bulk vaccine that complies with the following
within a suitable period, preferably not more than 24 h and in
requirements may be used in the preparation of the final lot.
any case not more than 72 h, of the prior filtration. The virus
suspension is inactivated by a validated method that has been Antimicrobial preservative. Where applicable, determine the
shown to inactivate poliovirus without destruction of amount of antimicrobial preservative by a suitable chemical
immunogenicity; during validation studies, an inactivation method. The content is not less than 85.0 per cent and not
curve with at least four points (for example, time 0, 24, 48, and greater than 115.0 per cent of the intended amount.
96 h) is established showing the decrease in concentration of Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
live virus with time. If formaldehyde is used for inactivation, for each sterility medium.
the presence of an excess of formaldehyde at the end of the
inactivation period is verified. Inactivation. Before addition of any antimicrobial preservative,
a sample of at least 1500 ml or, for a purified and concentrated
Only an inactivated monovalent harvest that complies with vaccine, the equivalent of 1500 doses is tested for residual
the following requirements may be used in the preparation of live poliovirus in cell cultures, as described for the inactivated
a trivalent pool of inactivated monovalent harvests or a final monovalent harvest. If the final bulk vaccine is prepared from
bulk vaccine. a trivalent pool of inactivated monovalent harvests, the test
Test for effective inactivation. After neutralisation of the for inactivation is carried out on that pool rather than on the
formaldehyde with sodium bisulphite (where applicable), verify final bulk vaccine.
the absence of residual live poliovirus by inoculation on FINAL LOT
suitable cell cultures of two samples of each inactivated
monovalent harvest, corresponding to at least 1500 human Only a final lot that complies with each of the requirements
doses. Take one sample not later than three-quarters of the given below under Identification, Tests and Assay may be
way through the inactivation period and the other at the end. released for use. Provided that the tests for free formaldehyde
Inoculate the samples in cell cultures such that the dilution of and antimicrobial preservative and the in vivo assay have
vaccine in the nutrient medium is not greater then ¼ and the been performed with satisfactory results on the final bulk
area of the cell layer is at least 3 cm2 per ml of inoculum. Set vaccine, they may be omitted on the final lot. Provided that
aside one or more containers with the same medium as non- the test for bovine serum albumin has been performed with
inoculated control cells. Observe the cell cultures for at least satisfactory results on the trivalent pool of inactivated
3 weeks. Make not fewer than two passages from each monovalent harvests or on the final bulk vaccine, it may be
container, one at the end of the observation period and the omitted on the final lot.
other 1 week before; for the passages, use cell culture Identification
supernatant and inoculate as for the initial sample. Observe
the subcultures for at least 2 weeks. No sign of poliovirus The vaccine is shown to contain human polioviruses 1, 2 and
multiplication is present in the cell cultures. At the end of the 3 by a suitable immunochemical method such as the
observation period, test the susceptibility of the cell culture determination of D-antigen by enzyme-linked immunosorbent
used by inoculation of live poliovirus of the same type as that assay (ELISA).
present in the inactivated monovalent harvest.
Tests
Sterility (2.2.11). The inactivated monovalent harvest complies
with the test for sterility, carried out using 10 ml for each Free formaldehyde (2.3.20). Maximum 0.2 g/l.
medium. Antimicrobial preservative. Where applicable, determine the
D-antigen content. The content of D-antigen determined by a amount of antimicrobial preservative by a suitable chemical

76
IP 2007 POLIOMYELITIS VACCINE, LIVE (ORAL)

method. The content is not less than 85.0 per cent and not number of animals per group must be sufficient to obtain
greater than 115.0 per cent of the intended amount. results that meet the validity criteria; groups of 10 rats are
Protein content (2.3.49). Not more than 10 µg of protein usually sufficient although valid results may be obtained with
nitrogen per human dose. fewer animals per group. The weight of individual animal must
not vary by more than 10.0 per cent from the group mean. An
Bovine serum albumin. Not more than 50 ng per single human inoculum of 0.5 ml per rat is used. The dose range is chosen
dose, determined by a suitable immunochemical method such that a dose response to all 3 poliovirus types is obtained.
(2.2.14). Bleed the animals after 20 to 22 days. Neutralising titres against
Sterility (2.2.11). Complies with the test for sterility. all 3 polivirus types are measured separately using 100 CCID50
of the Sabin strains as challenge viruses. Vero or Hep2 as
Bacterial endotoxins ( 2.2.3). Not more than 5 IU per human indicator cells, and neutralization conditions of 3 h at 35° to
dose. 37° followed by 18 h at 2° to 8°. Results are read following
Assay fixation and staining after 7 days of incubation at 35°. For a
valid antibody assay, the titre of each challenge virus must be
D-antigen content. As a measure of consistency of production, shown to be within the range of 10 to 1000 CCID50 and the
determine the D-antigen content for human polioviruses 1, 2 neutralizing antibody titre of a control serum must be within 2
and 3 by a suitable immunochemical method (2.2.14) using an twofold dilutions of the geometric mean titre of the serum.
appropriate reference preparation calibrated in D-antigen units. The potency is calculated by comparison of the preparation
For each type, the content, expressed with reference to the of responders for the vaccine under examination and the
amount of D-antigen stated on the label, is within the limits reference vaccine by the probit method or, after validation,
approved for the particular product. using a parallel-line model. For the probit method it is
In vivo test. The capacity of the vaccine to induce the formation necessary to establish a cut-off neutralising antibody titre for
of neutralizing antibodies is determined in-vivo by one of the each poliovirus type to define a responder. Due to
following methods: interlaboratory variation, it is not possible to define cut-off
values that could be applied by all laboratories. Rather, the
Test in chicks or guinea-pigs. Prepare a suitable series of at cut-off values are determined for each laboratory based on a
least three dilutions of the vaccine under examination using a minimum series of 3 tests with the reference vaccine. The mid-
suitable buffered saline solution. Inject 0.5 ml of the dilutions point on a log 2 scale of the minimum and maximum geometric
intramuscularly into groups of ten 3-week-old chickens or mean titres of the series of 3 or more tests is used as the cut-
groups of ten guinea-pigs, each weighing between 250 and off value. For each of the 3 poliovirus types, the potency of
350 g, using a separate group for each dilution of vaccine. the vaccine is not significantly less than that of the reference
Bleed the animals on the fifth or sixth day after the injection preparation. The test is not valid unless (1) for both the test
and separate the sera. Examine the sera for the presence of and reference vaccines the ED50 lies between the smallest and
neutralising antibody, at a dilution of 1 in 4, to each of the the largest doses given to the animals; (2) the statistical
human polioviruses 1, 2 and 3. Mix 100 CCID50 of virus with analysis shows no significant deviation from linearity or
the dilution of serum and incubate at 37° for 4 h 30 min to 6 h. parallelism; (3) the fiducial limits of the estimated relative
Keep at 5 ± 3° for 12 to 18 h. Inoculate the mixtures into cell potency fall between 25.0 per cent and 400.0 per cent of the
cultures for the detection of unneutralised virus and read the estimated potency.
results up to 7 days after inoculation. For each group of animals,
Labelling. The label states (1) the types of poliovirus
note the number of sera which have neutralising antibody
contained in the vaccine; (2) the nominal amount of virus of
and calculate the dilution of the vaccine giving an antibody
each type (1, 2 and 3), expressed in units of D-antigen per
response in 50.0 per cent of the animals. Carry out in parallel a
single human dose; (3) the cell substrate used to prepare the
control test using a suitable reference preparation.
vaccine.
The vaccine complies with the test if a dilution of 1 in 100 or
more produces an antibody response for each of the three
types of virus in 50.0 per cent of the animals.
Test on rats. A suitable in vivo assay method consists of
Poliomyelitis Vaccine, Live (Oral)
intramuscular injection into the hind limb(s) of not fewer than Oral Poliomyelitis Vaccine is a preparation of approved strains
3 dilutions of the vaccine under examination and a reference of live attenuated poliovirus type 1, 2 or 3 grown in in vitro
vaccine, using for each dilution a group of 10 specific cultures of approved cells, containing any one type or any
pathogen-free rats of the suitable strain. Use of 4 dilutions is combination of the three types of Sabin strains, prepared in a
often necessary to obtain valid results for all 3 serotypes. The form suitable for oral administration.

77
POLIOMYELITIS VACCINE, LIVE (ORAL) IP 2007

Production Monkeys from which kidneys are to be removed shall be


anaesthetised and thoroughly examined, particularly for
General provisions evidence of tuberculosis and cercopithecid herpesvirus 1 (B
The vaccine strains and the production method should virus) infection.
consistently yield vaccines that are both immunogenic and If a monkey shows any pathological lesion relevant to the use
safe in man. of its kidneys in the preparation of a seed lot or vaccine, it
The production of vaccine is based on a virus seed-lot system. shall not be used, nor shall any of the remaining monkeys of
Cell lines are used according to a cell-bank system. If primary the quarantine group concerned be used unless it is evident
monkey kidney cells are used, production complies with the that their use will not impair the safety of the product.
requirements indicated below. Unless otherwise justified and All the operations described in this section shall be conducted
authorised, the virus in the final vaccine shall not have outside the areas where the vaccine is produced.
undergone more than two passages from the master seed lot.
The monkeys used shall be shown to be free from antibodies
Substrate for virus propagation to simian virus 40 (SV40) and simian immunodeficiency virus.
If Macaca spp. are used for production, the monkeys shall
The virus is propagated in human diploid cells (2.7.2) or in also be shown to be free from antibodies to cercopithecid
continuous cell lines (2.7.2) or in primary monkey kidney cells herpesvirus 1 (B virus). Human herpesvirus has been used as
(including serially passaged cells from primary monkey kidney an indicator for freedom from B virus antibodies on account
cells). Continuous cell lines are approved by the competent of the danger of handling cercopithecid herpesvirus 1 (B virus).
authority.
Monkey kidney cell cultures for vaccine production. Kidneys
Primary monkey cells. The following special requirements that show no pathological signs are used for preparing cell
for the substrate for virus propagation apply to primary cultures. If the monkeys are from a colony maintained for
monkey cells. vaccine production, serially passaged monkey kidney cell
Monkeys used for preparation of kidney cell cultures and for cultures from primary monkey kidney cells may be used for
testing of virus. If the vaccine is prepared in monkey kidney virus propagation, otherwise the monkey kidney cells are not
cell cultures, animals of a species approved by the competent propagated in series. Virus for the preparation of vaccine is
authority, in good health, and not previously employed for grown by aseptic methods in such cultures. If animal serum is
experimental purposes shall be used. used in the propagation of the cells, the maintenance medium
after virus inoculation shall contain no added serum.
The monkeys shall be kept in well-constructed and adequately
ventilated animal rooms in cages spaced as far apart as Each group of cell cultures derived from a single monkey or
possible. Adequate precautions shall be taken to prevent from fetuses from no more than ten near-term monkeys is
cross-infection between cages. Not more than two monkeys prepared and tested as an individual group.
shall be housed per cage and cage-mates shall not be
SEED LOT
interchanged. The monkeys shall be kept in the country of
manufacture of the vaccine in quarantine groups for a period The strains of poliovirus used shall be identified by historical
of not less than 6 weeks before use. A quarantine group is a records that include information on the origin and subsequent
colony of selected, healthy monkeys kept in one room, with manipulation of the strains.
separate feeding and cleaning facilities, and having no contact
with other monkeys during the quarantine period. If at any Working seed lots are prepared by a single passage from a
time during the quarantine period the overall death rate of a master seed lot and at an approved passage level from the
shipment consisting of one or more groups reaches 5 per cent original Sabin virus. Virus seed lots are prepared in large
(excluding deaths from accidents or where the cause was quantities and stored at a temperature below -60°.
specifically determined not to be an infectious disease), Only a virus seed lot that complies with the following
monkeys from that entire shipment shall continue in quarantine requirements may be used for virus propagation.
from that time for a minimum of 6 weeks. The groups shall be
kept continuously in isolation, as in quarantine, even after Identification
completion of the quarantine period, until the monkeys are
Each working seed lot is identified as poliovirus of the given
used. After the last monkey of a group has been taken, the
type, using specific antibodies.
room that housed the group shall be thoroughly cleaned and
decontaminated before being used for a fresh group. If kidneys Virus concentration. Determined by the method described
from near-term monkeys are used, the mother is quarantined below, the virus concentration is the basis for the quantity of
for the term of pregnancy. virus used in the neurovirulence test.

78
IP 2007 POLIOMYELITIS VACCINE, LIVE (ORAL)

Extraneous agents (2.7.3). If the working seed lot is produced pooled harvest.
in human diploid cells (2.7.2) or in continuous cell lines (2.7.2) Virus concentration. The virus concentration of virus
it complies with the requirements for seed lots for virus harvests is determined as prescribed under Assay to monitor
vaccines. If the working seed lot is produced in primary consistency of production and to determine the dilution to be
monkey cells, it complies with the requirements given below used for the final bulk vaccine.
under Propagation and Harvest and Monovalent Pooled
Harvest and with the tests in adult mice, suckling mice and Extraneous agents ( 2.7.3 ). Complies with tests for extraneous
guinea-pigs given under Tests for extraneous agents in viral agents.
vaccines for human use. Control cells. The control cells of the production cell culture
Working seed lot shall be free from detectable DNA sequences from which the virus harvest is derived comply with a test for
from simian virus 40 (SV40) identity and with the requirements for extraneous agents or,
where primary monkey cells are used, as shown below.
Neurovirulence (2.7.6). Each master and working seed lot
complies with the test for neurovirulence of poliomyelitis Primary monkey cells. The following special requirements
vaccine (oral) in monkeys. Furthermore, the seed lot shall cease apply to virus propagation and harvest in primary monkey
to be used in vaccine production if the frequency of failure of cells.
the monovalent pooled harvests produced from it is greater Cell cultures. On the day of inoculation with virus seed, each
than predicted statistically. This statistical prediction is cell culture is examined for degeneration caused by an infective
calculated after each test on the basis of all the monovalent agent. If, in this examination, evidence is found of the presence
pooled harvests tested; it is equal to the probability of false in a cell culture of any extraneous agent, the entire group of
rejection on the occasion of a first test (i.e. 1 per cent), the cultures concerned shall be rejected.
probability of false rejection on retest being negligible. If the
On the day of inoculation with the virus working seed lot, a
test is carried out only by the manufacturer, the test slides are
sample of at least 30 ml of the pooled fluid removed from the
provided to the control authority for assessment.
cell cultures of the kidneys of each single monkey or from
Genetic markers. Each working seed lot is tested for its fetuses from not more than ten near-term monkeys is divided
replicating properties at temperatures ranging from 36° to 40° into two equal portions. One portion of the pooled fluid is
as described under Monovalent Pooled Harvest. tested in monkey kidney cell cultures prepared from the same
PROPAGATION AND HARVEST species, but not the same animal, as that used for vaccine
production. The other portion of the pooled fluid is, where
All processing of the cell-banks and subsequent cell-cultures
necessary, tested in monkey kidney cell cultures from another
is done under aseptic conditions in an area where no other
species so that tests on the pooled fluids are done in cell
cells are handled. Approved animal (but not human) serum
cultures from at least one species known to be sensitive to
may be used in the media, but the final medium for maintaining
SV40. The pooled fluid is inoculated into bottles of these cell
cell growth during virus multiplication does not contain animal
cultures in such a way that the dilution of the pooled fluid in
serum. Serum and trypsin used in the preparation of cell
the nutrient medium does not exceed 1 in 4. The area of the cell
suspensions and media are shown to be free from live
sheet is at least 3 cm2 per ml of pooled fluid. At least one bottle
extraneous agents. The cell-culture medium may contain a pH
of each kind of cell culture remains uninoculated to serve as a
indicator such as phenol red and approved antibiotics at the
control. If the monkey species used for vaccine production is
lowest effective concentration. It is preferable to have a
known to be sensitive to SV40, a test in a second species is
substrate free from antibiotics during production. Not less
not required. Animal serum may be used in the propagation of
than 5 per cent and not more than 1000 ml of the cell cultures
the cells, provided that it does not contain SV40 antibody, but
employed for vaccine production are set aside as uninfected
the maintenance medium after inoculation of test material
cell cultures (control cells); special requirements, given below,
contains no added serum except as described below.
apply to control cells when the vaccine is produced in primary
monkey cells The virus suspension is harvested not later than The cultures are incubated at a temperature of 35° to 37° and
4 days after virus inoculation. After inoculation of the are observed for a total period of at least 4 weeks. During this
production cell culture with the virus working seed lot, observation period and after not less than 2 weeks’ incubation,
inoculated cells are maintained at a fixed temperature, shown at least one subculture of fluid is made from each of these
to be suitable, within the range 33° to 35°; the temperature is cultures in the same cell culture system. The subcultures are
maintained constant to ±0.5°; control cell cultures are also observed for at least 2 weeks.
maintained at 33° to 35° for the relevant incubation periods. Serum may be added to the original culture at the time of
Only a single virus harvest that complies with the following subculturing, provided that the serum does not contain SV40
requirements may be used in the preparation of the monovalent antibody.

79
POLIOMYELITIS VACCINE, LIVE (ORAL) IP 2007

Fluorescent-antibody techniques may be useful for detecting examined for degeneration caused by an infectious agent. If
SV40 virus and other viruses in the cells. this examination or any of the tests required in this section
A further sample of at least 10 ml of the pooled fluid is tested shows evidence of the presence in a control culture of any
for cercopithecid herpesvirus 1 (B virus) and other viruses in extraneous agent, the poliovirus grown in the corresponding
rabbit kidney cell cultures. Serum used in the nutrient medium inoculated cultures from the same group shall be rejected.
of these cultures shall have been shown to be free from Tests for haemadsorbing viruses. At the time of harvest or
inhibitors of B virus. Human herpesvirus has been used as an within 4 days of inoculation of the production cultures with
indicator for freedom from B virus inhibitors on account of the the virus working seed lot, a sample of 4 per cent of the control
danger of handling cercopithecid herpesvirus 1 (B virus). The cell cultures is taken and tested for haemadsorbing viruses.
sample is inoculated into bottles of these cell cultures in such At the end of the observation period, the remaining control
a way that the dilution of the pooled fluid in the nutrient cell cultures are similarly tested. The tests are made as described
medium does not exceed 1 in 4. The area of the cell sheet is at in (2.7.3), Tests for extraneous agents in viral vaccines for
least 3 cm2 per ml of pooled fluid. At least one bottle of the cell human use.
cultures remains uninoculated to serve as a control.
Tests for other extraneous agents. At the time of harvest, or
The cultures are incubated at a temperature of 35° to 37° within 7 days of the day of inoculation of the production
and observed for at least 2 weeks. cultures with the working seed lot, a sample of at least 20 ml of
the pooled fluid from each group of control cultures is taken
A further sample of 10 ml of the pooled fluid removed from the and tested in two kinds of monkey kidney cell culture, as
cell cultures on the day of inoculation with the seed lot virus described above.
is tested for the presence of extraneous agents by inoculation
into human cell cultures sensitive to measles virus. At the end of the observation period for the original control
cell cultures, similar samples of the pooled fluid are taken and
The tests are not valid if more than 20 per cent of the culture the tests referred to in this section in the two kinds of monkey
vessels have been discarded for non-specific accidental kidney cell culture and in the rabbit cell cultures are repeated,
reasons by the end of the respective test periods. as described above under Cell cultures.
If, in these tests, evidence is found of the presence of an
If the presence of Cercopithecid herpesvirus 1 (B virus) is
extraneous agent, the single harvest from the whole group of
demonstrated, the production cell cultures shall not be used
cell cultures concerned is rejected.
and the measures concerning vaccine production described
If the presence of cercopithecid herpesvirus 1 (B virus) is above must be undertaken.
demonstrated, the manufacture of oral poliomyelitis vaccine
The fluids collected from the control cell cultures at the time
shall be discontinued and the competent authority shall be
of virus harvest and at the end of the observation period may
informed. Manufacturing shall not be resumed until a thorough
be pooled before testing for extraneous agents. A sample of 2
investigation has been completed and precautions have been
per cent of the pooled fluid is tested in each of the cell culture
taken against any reappearance of the infection, and then
systems specified.
only with the approval of the competent authority.
If these tests are not done immediately, the samples of pooled Single harvests
cell-culture fluid shall be kept at a temperature of -60° or below,
Tests for neutralised single harvests in monkey kidney cell
with the exception of the sample for the test for B virus, which
cultures. A sample of at least 10 ml of each single harvest is
may be held at 4°, provided that the test is done not more than
neutralised by a type-specific poliomyelitis antiserum prepared
7 days after it has been taken.
in animals other than monkeys. In preparing antisera for this
Control cell cultures. On the day of inoculation with the virus purpose, the immunising antigens used shall be prepared in
working seed lot 25 per cent (but not more than 2500 ml) of the non-simian cells.
cell suspension obtained from the kidneys of each single
monkey or from not more than ten near-term monkeys is taken Half of the neutralised suspension (corresponding to at least
to prepare uninoculated control cell cultures. These control 5 ml of single harvest) is tested in monkey kidney cell cultures
cell cultures are incubated in the same conditions as the prepared from the same species, but not the same animal, as
inoculated cultures for at least 2 weeks and are examined during that used for vaccine production. The other half of the
this period for evidence of cytopathic changes. The tests are neutralised suspension is tested, if necessary, in monkey
not valid if more than 20 per cent of the control cell cultures kidney cell cultures from another species so that the tests on
have been discarded for non-specific, accidental reasons. At the neutralised suspension are done in cell cultures from at
the end of the observation period, the control cell cultures are least one species known to be sensitive to SV40.

80
IP 2007 POLIOMYELITIS VACCINE, LIVE (ORAL)

The neutralised suspensions are inoculated into bottles of Virus concentration


these cell cultures in such a way that the dilution of the
The virus concentration is determined by the method
suspension in the nutrient medium does not exceed 1 in 4. The
described below and serves as the basis for calculating the
area of the cell sheet is at least 3 cm2 per ml of neutralised
dilutions for preparation of the final bulk, for the quantity of
suspension. At least one bottle of each type of cell culture
virus used in the neurovirulence test and to establish and
remains uninoculated to serve as a control and is maintained
monitor production consistency.
by nutrient medium containing the same concentration of the
specific antiserum used for neutralisation. Genetic markers. A ratio of the replication capacities of the
Animal serum may be used in the propagation of the cells, virus in the monovalent pooled harvest is obtained over a
provided that it does not contain SV40 antibody, but the temperature range between 36° and 40° in comparison with
maintenance medium, after the inoculation of the test material, the seed lot or a reference preparation for the marker tests and
contains no added serum other than the poliovirus neutralising with appropriate rct/40- and rct/40+ strains of poliovirus of
antiserum, except as described below. the same type. The incubation temperatures used in this test
are controlled to within ±0.1°. The monovalent pooled harvest
The cultures are incubated at a temperature of 35° to 37° and
passes the test if, for both the virus in the harvest and the
observed for a total period of at least 4 weeks. During this
appropriate reference material, the titre determined at 36° is at
observation period and after not less than 2 weeks’ incubation,
least 5.0 log greater than that determined at 40°. If growth at
at least one subculture of fluid is made from each of these
40° is so low that a valid comparison cannot be established, a
cultures in the same cell-culture system. The subcultures are
temperature in the region of 39.0° to 39.5° is used, at which
also observed for at least 2 weeks.
temperature the reduction in titre of the reference material
Serum may be added to the original cultures at the time of must be in the range 3.0 to 5.0 log of its value at 36°; the
subculturing, provided that the serum does not contain SV40 acceptable minimum reduction is determined for each virus
antibody. strain at a given temperature. If the titres obtained for one or
Additional tests are made for extraneous agents on a further more of the reference viruses are not concordant with the
sample of the neutralised single harvests by inoculation of 10 expected values, the test must be repeated.
ml into human cell cultures sensitive to measles virus. Neurovirulence (2.7.6). Each monovalent pooled harvest
Fluorescent-antibody techniques may be useful for detecting complies with the test for neurovirulence of poliomyelitis
SV40 virus and other viruses in the cells. vaccine (oral). If the test is carried out only by the manufacturer,
the test slides are provided to the competent authority for
The tests are not valid if more than 20 per cent of the culture assessment. The TgPVR21 transgenic mouse model provides
vessels have been discarded for non-specific accidental a suitable alternative to the monkey neurovirulence test for
reasons by the end of the respective test periods. neurovirulence testing of types 1, 2 or 3 vaccines once a
If any cytopathic changes occur in any of the cultures, the laboratory qualifies as being competent to perform the test
causes of these change are investigated. If the cytopathic and the experience gained is to the satisfaction of the
changes are shown to be due to unneutralised poliovirus, the competent authority. The test is carried out using a standard
test is repeated. If there is evidence of the presence of SV40 or operating procedure approved by the competent authority. A
other extraneous agents attributable to the single harvest, suitable procedure (Neurovirulence test of type 1, 2 or 3 live
that single harvest is rejected. poliomyelitis vaccines (oral) in transgenic mice susceptible
to poliovirus) is available from WHO, Quality and Safety of
MONOVALENT POOLED HARVEST Biologicals, Geneva.
Monovalent pooled harvests are prepared by pooling a number Primary monkey cells. The following special requirements
of satisfactory single harvests of the same virus type. apply to monovalent pooled harvests derived from primary
Monovalent pooled harvests from continuous cell lines may monkey cells.
be purified. Each monovalent pooled harvest is filtered through
a bacteria-retentive filter. Retroviruses. The monovalent pooled harvest is examined
using a reverse transcriptase assay. No indication of the
Only a monovalent pooled harvest that complies with the
presence of retrovirus is found.
following requirements may be used in the preparation of the
final bulk vaccine. Test on rabbits. A sample of the monovalent pooled harvest is
tested for cercopithecid herpesvirus 1 (B virus) and other
Identification viruses by injection of not less than 100 ml into not fewer than
Each monovalent pooled harvest is identified as poliovirus of 10 healthy rabbits each weighing between 1.5 and 2.5 kg. Each
the given type, using specific antiserum. rabbit receives not less than 10 ml and not more than 20 ml, of

81
POLIOMYELITIS VACCINE, LIVE (ORAL) IP 2007

which 1 ml is given intradermally at multiple sites, and the FINAL LOT


remainder subcutaneously. The rabbits are observed for at
Only a final lot that complies with the following requirement
least 3 weeks for death or signs of illness.
for thermal stability and is satisfactory with respect to each of
All rabbits that die after the first 24 h of the test and those the requirements given below under Identification, Tests and
showing signs of illness are examined by autopsy, and the Assay may be released for use.
brain and organs removed for detailed examination to establish
Thermal stability. Expose samples of the final lot at 37° for
the cause of death.
48 hours. Determine the total virus concentration as described
The test is not valid if more than 20.0 per cent of the inoculated under Assay in parallel for the heated vaccine and for unheated
rabbits show signs of intercurrent infection during the vaccine. The estimated difference between the total virus
observation period. The monovalent pooled harvest passes concentration of the unheated and heated vaccines is not
the test if none of the rabbits shows evidence of infection greater than 0.5 log10 infectious virus units (CCID50) per single
with B virus or with other extraneous agents or lesions of any human dose.
kind attributable to the bulk suspension.
Identification
If the presence of B virus is demonstrated, the measures
concerning vaccine production described above under Cell The vaccine is shown to contain poliovirus of each type stated
cultures are taken. on the label, using specific antibodies.
Test on guinea-pigs. Administer to not less than five guinea- Tests
pigs, each weighing between 350 and 450 g, 0.1 ml of the
Sterility (2.2.11). Complies with the test for sterility.
monovalent pooled harvest by intracerebral injection and 0.5
ml by intraperitoneal injection. Measure the rectal temperature Assay
of each animal on each working day for 6 weeks. At the end of
the observation period carry out autopsy on each animal. Titrate for infectious virus at least in triplicate using the method
described below. Use an appropriate virus reference
In addition, administer to not fewer than five guinea-pigs 0.5 preparation to validate each assay. If the vaccine contains
ml by intraperitoneal injection and observe as described above more than one poliovirus type, titrate each type separately,
for 2 to 3 weeks. At the end of the observation period, carry using appropriate type-specific antiserum (or preferably a
out a passage from these animals to not fewer than five guinea- monoclonal antibody) to neutralise each of the other types
pigs using blood and a suspension of liver or spleen tissue. present.
Measure the rectal temperature of the latter guinea-pigs for 2
to 3 weeks. Examine by autopsy all animals that, after the first For a trivalent vaccine, the estimated mean virus titres must
day of the test, die or are killed because they show disease or be: not less than 1 × 106.0 infectious virus units (CCID50) per
show for three consecutive days a body temperature higher single human dose for type 1; not less than 1 × 105.0 infectious
than 39°; carry out histological examination to detect infection virus units (CCID50) for type 2; and not less than 1 × 105.8
with Marburg virus; in addition, inject a suspension of liver or infectious virus units (CCID50) for type 3.
spleen tissue or of blood intraperitoneally into not fewer than For monovalent or divalent vaccine, the minimum virus titres
three guinea-pigs. If any signs of infection with Marburg virus are decided by the competent authority.
are noted, confirmatory serological tests are carried out on
Method. Groups of eight to twelve flat-bottomed wells in a
the blood of the affected animals. The monovalent pooled
microtitre plate are inoculated with 0.05 ml of each of the
harvest complies with the test if not fewer than 80.0 per cent
selected dilutions of virus followed by a suitable cell
of the guinea-pigs survive to the end of the observation period
suspension of the Hep-2 (Cincinnati) line. The plates are
and remain in good health and no animal shows signs of
incubated at a suitable temperature. Examine the cultures on
infection with filoviruses virus.
days 7 to 9.
FINAL BULK VACCINE The assay is not valid if (a) the confidence interval (P = 0.95)
The final bulk vaccine is prepared from one or more of the logarithm of the virus concentration is greater than
satisfactory monovalent pooled harvests and may contain ±0.3; (b) the virus concentration of the reference preparation
more than one virus type. Suitable flavouring substances and differs by more than 0.5 log CCID50 from the assigned value.
stabilisers may be added. Labelling. The label states (1) the types of poliovirus
contained in the vaccine; (2) the minimum amount of virus of
Only a final bulk vaccine that complies with the following
each type contained in one single human dose; (3) the cell
requirement may be used in the preparation of the final lot.
substrate used for the preparation of the vaccine; (4) that the
Sterility (2.2.11). Complies with the test for sterility. vaccine is not to be injected.

82
IP 2007 RABIES VACCINE, HUMAN

Rabies Vaccine, Human immunofluoresence or any other approved method.

Rabies Vaccine for Human use is a freeze-dried or liquid Extraneous agents (2.7.3). The working seed lot complies with
(adsorbed) preparation of a suitable approved, strain of fixed the requirements for the virus seed lots.
rabies virus grown in an approved cell culture/ embryos of PROPAGATION AND HARVEST
duck/chicken and inactivated by a validated method.
All processing of the cell bank and subsequent cell cultures
The freeze-dried vaccine is reconstituted immediately before are done under aseptic conditions in an area where no other
use as stated on the label to give a clear or slightly opalescent cells are handled. Approved animal (but not human) serum
solution/suspension. It may be coloured owing to the presence may be used in the media, but the final medium for maintaining
of a pH indicator. cell growth during virus multiplication does not contain animal
The vaccine complies with the General Requirements of serum; the media may contain human albumin. Serum proteins,
Vaccines for Human Use. if present are reduced to an acceptable level by a suitable
method of purification. Serum and trypsin used in the
Production preparation of cell suspension and media are shown to be free
from infectious extraneous agents. The cell culture media may
General provisions contain a pH indicator such as phenol red and approved
antibiotics at the lowest effective concentration. Not less than
The vaccine is produced on the basis of virus seed lot system
500 ml of the cell cultures employed for vaccine production
and if a cell line is used for virus propagation, a cell-bank
are set aside as uninfected cell cultures (control cells). The
system shall be followed. The production method shall have
virus suspension is harvested on one or more occasions
been shown to yield consistently vaccines that comply with
during incubation. Multiple harvests from the same production
the requirements for immunogenicity, safety and stability.
cell culture may be pooled and considered as a single harvest.
Unless otherwise justified and authorized, the virus in the
final vaccine shall not have undergone more passages from When vaccine is prepared in embryonated eggs, the egg
the master seed lot than was used to prepare the vaccine proteins are minimized by an appropriate method of purification.
shown in clinical studies to be satisfactory with respect to The eggs are inoculated with virus seed by the yolk sac route.
safety and efficacy. The infected sterile living embryos are harvested, minced and
emulsified in suitable diluent, and stabilizer with aseptic
The production method is validated to demonstrate that the
precautions. Emulsions are centrifuged, supernatants are
product, if tested, would comply with the test for safety and
collected and stored as raw virus harvest at a suitable
efficacy.
temperature.
Substrate for virus propagation Viral harvests that comply with the following requirements are
The virus is propagated in any suitable approved cell substrate pooled in the preparation of the inactivated viral harvest.
like a human diploid cell line (2.7.2), a continuous cell line, or
in duck embroys or in cultures of chicken embryos derived Identification
from a flock certified as free from specified pathogens(2.7.7). The single harvest contains virus that is identified as rabies
SEED LOT virus using specific antibodies by an approved method.
Virus concentration. Titrate for infective virus in cell cultures
The strain of rabies virus used shall be identified by historical
or by any other approved method. The titre is used to monitor
records that include information on the origin of the strain
consistency of production.
and its subsequent manipulation.
Control cells. The control cells of the production cell culture
Working seed lots are prepared by not more than five passages
from which the single harvest is derived should comply with a
from the master seed lot.
test for identification and with the requirements for extraneous
Only a working seed lot that complies with the following agents (2.7.3).
requirements may be used for virus propagation.
Control eggs. Control eggs shall be tested for freedom from
Identification haemagglutinating agents, and other extraneous agents.

Each working seed lot is identified as rabies virus using specific PURIFICATION AND INACTIVATION
antibodies by an approved method.
The virus harvests may be concentrated and/or purified by
Virus concentration. The virus concentration of each working suitable methods; the virus harvest is inactivated by a validated
seed lot is determined by a cell culture method using method at a fixed, well defined stage of the process which may

83
RABIES VACCINE, HUMAN IP 2007

be before, during or after any concentration or purification. maintain the activity of the product during and after freeze-
The method shall have been shown to be capable of drying. Thiomersal can be used as preservative.
inactivating rabies virus without destruction of the Only a final bulk vaccine that complies with the following
immunogenic activity. If betapropiolactone is used, the requirements may be used in the preparation of the final lot.
concentration shall at no time exceed 1:3500.
Antigen content. Determine the antigen content by a suitable
Cell culture vaccines approved in vitro or in vivo method. The content should be
Only an inactivated viral suspension that complies with the within the limits approved for the particular product.
following requirements may be used in the preparation of the Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
final bulk vaccine. for each sterility medium.
Inactivation. Inactivation is confirmed by carrying out an FINAL LOT
amplification test for residual infectious rabies virus, not more
than 4 days after inactivation or on the sample frozen after The final bulk vaccine is distributed aseptically into sterile
inactivation and stored at -70º. Inoculate a quantity of containers and can be freeze-dried in case of lyophilized
inactivated viral suspension equivalent to not less than 25 products. The containers are then sealed so as to prevent
vaccine doses into cell cultures of the same type as those contamination and the introduction of moisture.
used for production of the vaccine. Make a passage after 7 Only a final lot that complies with each of the requirements
days. Maintain the cultures for a further period of 14 days and given below under Identification, Tests and Assay may be
then examine the cell cultures for rabies virus using an immune released for use. Provided that the test for inactivation has
fluorescence test. No rabies virus is detected. Alternatively, 5 been carried out with satisfactory results on the inactivated
ml of each culture fluid is pooled on days 14 and 21 and 0.03 ml virus suspension and the test for bovine serum albumin has
is inoculated intracerebrally into each of the 10 mice weighing been carried out with satisfactory results on the final bulk
12 to 15 g. The mice are observed for 14 days for symptoms vaccine, these tests may be omitted on the final lot.
caused by rabies virus, and mice showing symptoms of rabies
are sacrificed and virus presence is confirmed by Identification
immunoflurescence test or tested for live virus in cell culture The vaccine is shown to contain rabies virus antigen by a
by immunofluorescence test or method of equal sensitivity. suitable immunochemical method using specific antibodies,
No rabies virus should be detected. alternatively, the Assay also serves to identify the vaccine.
Residual host-cell DNA (2.2.15). If a continuous cell line is Tests
used for virus propagation, the content of residual host-cell
DNA, determined using a suitable method, should not be Inactivation. Inoculate a quantity equivalent to not less than
greater than 10 ng per single human dose. 25 human doses of vaccine into cell cultures of the same type
as those used for production of the vaccine. Make a passage
Embryonated egg vaccine after 7 days. Maintain the culture for a further 14 days and
Only concentrated viral suspensions that comply with the then examine the cell culture for rabies virus using an
test for sterility, antigen content and endotoxin requirements immunofluorescence test. No rabies virus is detected.
may be used for preparation of bulk for inactivation. Alternatively, inject 0.03ml of the vaccine intracerebrally into
each of the 10 mice weighing between 12 and 15g. Neither
Inactivation is confirmed by carrying out Mice Inoculation symptoms of disease in the central nervous system nor death
Test for residual infectious rabies virus, not more than four occurs in any of the animal within 14 days. If the inactivation
days after inactivation or on the sample frozen 4 days after test is already performed on inactivated virus used for final
inactivation are stored at –70°. Inoculate intracerebrally 0.03 lot, it may be omitted from the test on final lot.
ml into each of 20 mice weighing between 12 and 15 g. The
mice are observed for 14 days for symptoms caused by rabies Sterility (2.2.11). Complies with the test for sterility.
virus and mice showing symptoms of rabies are sacrificed and Bacterial endotoxins (2.2.3). Less than 25 IU per single human
virus presence is confirmed by an immunoflurescence test or dose.
tested for live virus in cell culture by immunofluorescence
test or method of equal sensitivity. No rabies virus should be Pyrogens (2.2.8). Complies with the test for pyrogens. Unless
detected. otherwise justified and authorized, inject into each rabbit a
single human dose of the vaccine diluted to ten times its
FINAL BULK VACCINE volume.
The final bulk vaccine is prepared from one or more inactivated Water (2.3.43). Not more than 3.0 per cent determined by an
viral suspensions. An approved stabilizer may be added to approved method.

84
IP 2007 RABIES VACCINE, HUMAN

Abnormal toxicity (2.2.1). Complies with the test for abnormal signs of rabies between the fifth and fourteenth day after
toxicity. challenge are counted as failing to resist the challenge.
Accelerated degradation. The potency determined by method The strain of mice suitable for the test is such that when 0.03
described under “Assay” of a sample of the preparation under ml containing 5 to 50 LD50 of the challenge virus suspension
examination after storage at 37° for 4 weeks is not less than 2.5 is injected intracerebrally per mouse there is 100 per cent
units per single human dose. This may not be mandatory for mortality.
lot release, once the consistency of the product is approved
Standard challenge virus suspension. A working pool of the
by National Regulatory Authority.
challenge virus strain is prepared by injecting intracerebrally
Bovine serum albumin (for cell culture vaccine). Not more 0.03ml of a 10 fold dilution of the CVS strain of rabies virus in
than 50 ng per single human dose, determined by a suitable 2 per cent v/v sterile inactivated normal horse serum in water
immunochemical method (2.2.14). for injection or another suitable diluent approved by the
Aluminium content (for gel absorbed vaccine) (2.3.9). Not competent authority into a suitable number of test animals.
more than 1.25 mg per single human dose. The animals when moribund after showing characteristic signs
of rabies are sacrificed and their brains harvested aseptically.
Ovalbumin (for egg based vaccines). Not more than 1 µg of
They are then washed in chilled saline solution to remove
ovalbumin per human dose, determined by a suitable technique
blood clots. A 10 per cent suspension of the brains is prepared
using a suitable reference preparation of ovalbumin.
in a suitable diluent approved by the competent authority and
Residual host-cell DNA (for continuous cell line vaccines) thoroughly homogenised. After centrifuging lightly, the
(2.2.15). Should not be greater than 10 ng per single human supernatant liquid is distributed into sterile vials and freeze
dose. dried. The sealed and freeze-dried supernatant liquid containing
Antimicrobial preservative. Where applicable, determine the vials are stored at -20°. When stored under prescribed
amount of antimicrobial preservative by a suitable chemical conditions the virus titre of the freeze-dried preparation may
method. The content is not less than 85.0 per cent and not be expected to be maintained for not less than 3 years.
greater than 115.0 per cent of the intended amount. Alternatively, the washed brains are homogenised in a suitable
diluent approved by the competent authority to give 10 per
Assay cent suspension. It is then centrifuged lightly, distributed into
sterile ampoules or sterile plastic vials and sealed. The sealed
The Standard preparation is the international standard or
ampoules or plastic vials can be stored at –60º or below. When
another suitable preparation, the potency of which has been
stored under prescribed conditions, the virus titre may be
determined in relation to the International standard. The
expected to be maintained for not less than one year. Storage
potency of rabies vaccine is determined by comparing the
time needs to be validated by the manufacturer.
dose necessary to protect mice against a lethal intracerebral
dose of rabies vaccine necessary to provide the same Virus titre of the challenge virus. Prepare ten fold serial
protection. For this comparison, a reference preparation of dilutions of the standard challenge virus suspension. Using
rabies vaccine, calibrated in international units, and a suitable the four groups of 10 mice each, inject 0.03 ml of the virus
preparation of rabies virus for use as the challenge preparation suspension intracerebrally into each mouse, using a different
are necessary. group for each suspension. Observe the mice for 14 days.
The international unit is the activity contained in a stated Calculate the virus titre of the standard challenge virus
quantity of the international standard. The equivalence in suspension in LD50 per dose of 0.03 ml by standard statistical
international units of the international standard is stated by methods.
the World Health Organization. Determination of potency of the vaccine. Reconstitute the
The test described below uses a parallel-line model with at standard preparation with a suitable diluent. Prepare at least
least three points for the vaccine under examination and the three 5-fold serial dilutions of the solution of the standard
reference preparation. preparation and three 5-fold serial dilutions of the vaccine
under examination. For both, the standard preparation and
Test animals. Use mice of a suitable strain, drawn from a uniform the preparation under examination, the serial dilutions should
stock three to four weeks old, weighing between 11 and 15 g. be prepared in such a way that the lowest dilution protects
Distribute the mice into six groups of at least 16 mice each and more than 50 per cent of the injected mice. Allocate one dilution
four groups of 10 mice each and must be of the same sex or the to each of the six groups of 16 mice each. Inject
sexes must be equally distributed among the groups. intraperitoneally each mouse in each group with dilutions of
Throughout the test all mice that die before the fifth day after the vaccine and reference preparation and repeat the injections.
challenge are excluded from the test and all mice that die with After 7 days, prepare identical dilutions of the vaccine and

85
RUBELLA VACCINE (LIVE) IP 2007

reference preparation and repeat the injections. information on the origin of the strain and its subsequent
After a further 7 days, inject each vaccinated mouse manipulation.
intracerebrally with 0.03 ml of the standard challenge virus To avoid unnecessary use of monkeys in the test for
suspension such that on the basis of preliminary titration, neurovirulence virus seed lots are prepared in large quantities
0.03 ml contains between 5 to 50 LD50. Observe the mice for 14 and stored at temperatures below -20° if freeze-dried, or below
days and record the number of mice surviving the challenge -60° if not freeze-dried.
in each group. Calculate the potency of the preparation under Only a seed lot that complies with the following tests may be
examination by standard statistical methods. used for virus propagation.
The vaccine complies with the test if the estimated potency is Identification
not less than 2.5 IU per single human dose.
The master and working seed lots are identified as rubella
The test is not valid unless (a) for both the preparation under virus by serum neutralisation in cell culture, using specific
examination and the standard preparation, the 50 per cent antibodies.
protective dose lies between the largest and smallest doses
Virus concentration. The virus concentration of the master
given to the mice; (b) there is not deviation from linearity or
and working seed lots is determined to ensure consistency of
parallelism of the dose response lines, the confidence limit (P
production.
= 0.95) are not less than 25.0 per cent and not more than 400
per cent of the estimated potency; (c) the titre of the challenge Extraneous agents (2.7.3). The working seed lot complies with
virus suspension lies between 5 to 50 LD50. the tests for seed lots.
Labelling. The label states the biological origin of the cells Neurovirulence (2.7.5). The master/working seed lot complies
used for the preparation of the vaccine. with the test for neurovirulence of live virus vaccines. Macaca
and Cercopithecus monkeys are suitable for the test.
PROPAGATION AND HARVEST
Rubella Vaccine (Live) All processing of the cell bank and subsequent cell cultures is
done under aseptic conditions in an area where no other cells
Rubella Vaccine (Live) is a freeze-dried preparation of a suitable are handled. Suitable animal (but not human) serum may be
attenuated strain of rubella virus. The vaccine is reconstituted used in the growth medium, but the final medium for maintaining
immediately before use to give a clear liquid or may be coloured cell growth during virus multiplication does not contain animal
owing to the presence of a pH indicator. serum. Serum and trypsin used in the preparation of cell
suspensions and culture media are shown to be free from
Production
extraneous agents. The cell culture medium may contain a pH
indicator such as phenol red and suitable antibiotics at the
General provisions
lowest effective concentration. It is preferable to have a
The production of vaccine is based on a virus seed-lot system substrate free from antibiotics during production. Not less
and a cell-bank system. The production method shall have than 500 ml of the production cell culture is set aside as
been shown to yield consistently live rubella vaccines of uninfected cell culture (control cells). The temperature of
adequate immunogenicity and safety in man. Unless otherwise incubation is controlled during the growth of the virus. The
justified and authorised, the virus in the final vaccine shall virus suspension is harvested, on one or more occasions,
have undergone no more passages from the master seed lot within 28 days of inoculation. Multiple harvests from the same
than were used to prepare the vaccine shown in clinical studies production cell culture may be pooled and considered as a
to be satisfactory with respect to safety and efficacy. single harvest.
The production method is validated to demonstrate that the Only a single harvest that complies with the following
product, if tested, would comply with the test for safety and requirements may be used in the preparation of the final bulk
efficacy. vaccine.

Substrate for virus propagation Identification


The virus is propagated in human diploid cells (2.7.2). The single harvest contains virus that is identified as rubella
virus by serum neutralization in cell culture, using specific
SEED LOT antibodies.
The strain of rubella virus used in the production of rubella Virus concentration. The virus concentration in the single
vaccine shall be identified by historical records that include harvest is determined as prescribed under Assay to monitor

86
IP 2007 TETANUS VACCINE (ADSORBED)

consistency of production and to determine the dilution to be Abnormal toxicity (2.2.1). Complies with the test for abnormal
used for the final bulk vaccine. toxicity.
Extraneous agents (2.7.3). The single harvest complies with Assay
the tests for extraneous agents.
Titrate the vaccine for infective virus at least in triplicate,
Control cells. The control cells comply with a test for
using at least five cell cultures for each 0.5 log10 dilution step
identification and with the tests for extraneous agents (2.7.3).
or by a method of equal precision. Use an appropriate virus
FINAL BULK VACCINE reference preparation to validate each assay. The estimated
virus concentration is not less than that stated on the label;
Single harvests that comply with the above tests are pooled the minimum virus concentration stated on the label is not
and clarified to remove cells. A suitable stabilizer may be added less than 1 × 103 CCID50 per human dose. The assay is not
and the pooled harvests diluted as appropriate. valid if the confidence limits (P=0.95) of the logarithm of the
Only a final bulk vaccine that complies with the following virus concentration is greater than ± 0.3.
requirements may be used in the preparation of the final lot. Rubella vaccine (Live) RS is suitable for use as a reference
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk preparation.
for each sterility medium. Labelling. The label states (1) the strain of virus used for the
FINAL LOT preparation of the vaccine; (2) the type and origin of the cells
used for the preparation of the vaccine; (3) the minimum virus
A minimum virus concentration for release of the product is concentration; (4) the time within which the vaccine must be
established such as to ensure, in the light of stability data, used after reconstitution; (5) that the vaccine must not be
that the minimum concentration stated on the label will be given to a pregnant woman and that a woman must not become
present at the end of the period of validity. pregnant within two months.
Only a final lot that complies with the tests for minimum virus
concentration for release, with the following requirement for
thermal stability and with each of the requirements given below
under Identification and Tests may be released for use. Tetanus Vaccine (Adsorbed)
Provided that the test for bovine serum albumin has been Tetanus Vaccine (Adsorbed) is a preparation of tetanus formol
carried out with satisfactory results on the final bulk vaccine, toxoid adsorbed on mineral carrier. The formol toxoid is
it may be omitted on the final lot. prepared from the toxin produced by the growth of Clostridium
Thermal stability. Maintain samples of the final lot of freeze- tetani.
dried vaccine in the dry state at 37° for 7 days. Determine the
virus concentration as described under Assay in parallel for Production
the vaccine held at 37° for 7 days and for vaccine stored at 2° General provisions
to 8°. The virus concentration of the heated vaccine is not
more than 1.0 log10 lower than that of the unheated vaccine. The maximum number of Lf per single human dose of tetanus
vaccine is 25.
Identification The production method is validated to demonstrate that the
When the vaccine reconstituted as stated on the label is mixed product, if tested, would comply with the following test.
with specific rubella antibodies, it is no longer able to infect BULK PURIFIED TOXOID
susceptible cell cultures.
For the production of tetanus toxin, from which toxoid is
Tests prepared, seed cultures are managed in a defined seed-lot
system in which toxinogenicity is conserved and, where
Sterility (2.2.11). The reconstituted vaccine complies with the
necessary, restored by deliberate reselection. A highly
test for sterility.
toxinogenic strain of Clostridium tetani with known origin
Water (2.3.43). Not more than 3.0 per cent, determined by Karl and history is grown in a suitable liquid medium. At the end of
Fischer, semi-micro determination of water or by any suitable cultivation, the purity of each culture is tested and
validated method. contaminated cultures are discarded. Toxin-containing culture
Bovine serum albumin. Not more than 50 ng per single human medium is collected aseptically. The toxin content (Lf per ml)
dose, determined by a suitable immunochemical method is checked to monitor consistency of production. Single
(2.2.14). harvests may be pooled to prepare the bulk purified toxoid.

87
TETANUS VACCINE (ADSORBED) IP 2007

The toxin is purified to remove components likely to cause tetanus or dies from tetanus or any other cause within 21
adverse reactions in humans. The purified toxin is detoxified days.
with formaldehyde by a method that avoids destruction of the Antimicrobial preservative. Where applicable, determine the
immunogenic potency of the toxoid and reversion of toxoid to amount of antimicrobial preservative by a suitable chemical
toxin, particularly on exposure to heat. Alternatively, method. The content is not less than 85.0 per cent and not
purification may be carried out after detoxification. greater than 115.0 per cent of the intended amount.
Only bulk purified toxoid that complies with the following
Free formaldehyde (2.3.20). Maximum 0.2 g/l.
requirements may be used in the preparation of the final bulk
vaccine. pH (2.4.24). 6.0 and 7.0
Sterility (2.2.11). Carry out test for sterility using 10 ml of Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
bulk for each sterility medium. for each sterility medium.
Absence of tetanus toxin. Inject subcutaneously at least 500 Abnormal toxicity (2.2.1). Complies with the test for
Lf of purified toxoid in a volume of 1 ml into each of five abnormal toxicity for antisera and vaccine.
healthy guinea-pigs, each weighing 250 to 350 g, that have
not previously been treated with any material that will interfere FINAL LOT
with the test. If within 21 days of the injection any of the
The final bulk vaccine is distributed aseptically into sterile,
animals shows signs of or dies from tetanus, the toxoid does
tamper-proof containers. The containers are closed so as to
not comply with the test. If more than one animal dies from
prevent contamination.
non-specific causes, repeat the test once; if more than one
animal dies in the second test, the toxoid does not comply Only a final lot that is satisfactory with respect to each of the
with the test. requirements given below under Identification, Tests and
Assay may be released for use. Provided the tests for
Irreversibility of toxoid. Using the buffer for the final vaccine,
antimicrobial preservative and the assay have been carried
prepare a dilution of the bulk purified toxoid containing the
out with satisfactory results on the final bulk vaccine, they
same toxoid concentration as the final vaccine. Divide the
may be omitted on the final lot.
dilution into two equal parts. Keep one of them at 2° to 8° and
the other at 37° for 6 weeks. Test both dilutions by a suitable Identification
sensitive assay for active tetanus toxin, such as inoculation
into mice or guinea-pigs. The toxoid complies with the test if Tetanus toxoid is identified by a suitable immunochemical
neither sample produces any sign of a toxic reaction attribut- method (2.2.14). The following method, applicable to certain
able to tetanus toxin. vaccines, is given as an example. Dissolve in the vaccine under
examination sufficient sodium citrate to give a 10 per cent
Antigenic purity. Not less than 1,000 Lf per mg of protein
solution. Maintain at 37° for about 16 hours and centrifuge
nitrogen.
until a clear supernatant liquid is obtained. The clear
FINAL BULK VACCINE supernatant liquid reacts with a suitable tetanus antitoxin,
giving a precipitate.
The final bulk vaccine is prepared by adsorption of a suitable
quantity of bulk purified toxoid onto a mineral carrier such as Tests
hydrated aluminium phosphate or aluminium hydroxide; the
resulting mixture is approximately isotonic with blood. Suitable Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
antimicrobial preservatives may be added. Certain antimicrobial if aluminium hydroxide or hydrated aluminium phosphate is
preservatives, particularly those of the phenolic type, used as the adsorbent.
adversely affect the antigenic activity and must not be used. Free formaldehyde (2.3.20). Maximum 0.2 g/l.
Only final bulk vaccine that complies with the following Antimicrobial preservative. Where applicable, determine the
requirements may be used in the preparation of the final lot. amount of antimicrobial preservative by a suitable chemical
Specific toxicity. Inject five times the dose stated on the label method. The content is not less than 85.0 per cent and not
subcutaneously or intraperitoneally into each of five guinea- greater than 115.0 per cent of the intended amount.
pigs. None of the guinea-pigs shows any symptoms of, or
Sterility (2.2.11). Complies with the test for sterility.
dies from, tetanus within 21 days. If more than one animal dies
from non-specific causes within this period repeat the test. Abnormal toxicity (2.2.1). Complies with the test for abnormal
None of the second group of animals shows symptoms of toxicity for antisera and vaccine.

88
IP 2007 TETANUS VACCINE (ADSORBED)

Potency of tetanus component dose per ml. If the challenge toxin preparation has been shown
to be stable, it is not necessary to verify the paralytic dose for
Assay. Determine by either of the following methods.
every assay.
(1) Inject subcutaneously on each of two occasions separated Preparation of the challenge toxin solution. Immediately prior
by an interval of not more than 4 weeks, one-tenth of the to use, prepare from the challenge toxin by dilution with
stated human dose diluted to 1 ml with saline solution into phosphate buffered saline pH 7.4 a challenge toxin solution
each of 9 normal, healthy guinea-pigs weighing between 250 containing fifty times the 50 per cent paralytic dose per ml. If
and 350 g. Not more than 2 weeks after the second injection, necessary, dilute portions of this challenge toxin solution
collect the serum from each animal and carry out the biological 16-, 50- and 160-fold with the same buffer solution.
test for tetanus antitoxin, described under Tetanus antitoxin
or any other method approved by National Regulatory Determination of potency. Prepare in saline solution three
Authority. dilutions of the vaccine under examination and three dilutions
of a solution of the Standard preparation such that, for each,
Sera of at least 6 guinea-pigs out of 9 should contain not less
the dilutions form a series differing by not more than 2.5-fold
than 0.5 Unit of tetanus antitoxin per ml.
steps and in which the dilutions of intermediate concentration,
(2) Carry out the biological assay of adsorbed Tetanus Vaccine when injected subcutaneously in 1-ml volumes into guinea-
(Adsorbed) described below. pigs, protect approximately 50 per cent of the animals from the
If the lower limit of the 95.0 per cent confidence interval of paralytic effects of the subcutaneous injection of the quantity
estimated potency is less than 40 IU per single human dose of tetanus toxin prescribed for this test. Allocate the six
then the limits of the 95.0 per cent confidence interval of the dilutions one to each of the six groups of sixteen guinea-pigs
estimater of potency shall be within 50 to 200 per cent of the and inject subcutaneously 1.0 ml of each dilution into each
estimated potency unless the lower limit of the 95.0 per cent guinea-pig in the group to which that dilution is allocated.
confidence interval of the estimated potency is greater than After 28 days inject each animal subcutaneously with 1.0 ml
40 IU per single human dose. of the challenge toxin solution containing fifty times the 50
per cent paralytic dose. If necessary, allocate the challenge
Biological assay of adsorbed tetanus vaccine
toxin solution and the three dilutions made from it one to each
The potency of adsorbed tetanus vaccine is determined by of the four groups of five guinea-pigs and inject
comparing the dose of the vaccine required to protect guinea- subcutaneously 1.0 ml of each toxin solution into each guinea-
pigs or mice from the paralytic effects of a subcutaneous pig in the group to which that toxin solution is allocated.
injection of tetanus toxin with the dose of the Standard Examine the guinea-pigs twice daily, remove and kill all animals
preparation needed to give the same protection. For this showing definite signs of tetanus paralysis. Count the number
comparison, the Standard preparation of adsorbed tetanus of guinea-pigs without paralysis 5 days after injection of the
toxoid and a suitable preparation of tetanus toxin, for use as a challenge toxin and calculate the potency of the vaccine under
challenge toxin, are necessary. examination relative to the potency of the Standard preparation
Standard preparation on the basis of the number of animals without paralysis in
each of the six groups of sixteen, using standard statistical
The Standard preparation is International standard for Tetanus methods.
toxoid, adsorbed or another suitable preparation, the potency
of which has been determined in relation to the International The test is not valid unless (a) for both the vaccine under
standard. examination and the Standard preparation, the 50 per cent
protective doses lie between the largest and smallest doses of
Suggested method the preparations given to the guinea-pigs; (b) if applicable,
(a) Test on guinea-pigs the number of paralysed animals among the four groups of
five injected with the challenge toxin solution and its dilutions
Test animals. Use healthy guinea-pigs from the same stock
indicate that the challenge was approximately 50 times the 50
and weighing between 250 and 350 g. Distribute them into six
per cent paralytic dose; (c) the fiducial limits of assay lie
groups of sixteen each. The guinea-pigs should all be of the
between 50.0 per cent and 200.0 per cent of the estimated
same sex or the males and females should be distributed equally
potency; (d) the statistical analysis shows no deviations from
between the groups. If the challenge toxin to be used has not
linearity or parallelism. The test may be repeated any number
been shown to be stable or has not been adequately
of times but when more than one test is performed the results
standardised, include four further groups of five guinea-pigs
of all valid tests must be combined in the estimate of potency.
to serve as unvaccinated controls.
Challenge toxin. Select a preparation of tetanus toxin (b) Test on mice
containing not less than 50 times the 50 per cent paralytic Test animals. Use healthy mice from the same stock, weighing

89
TETANUS VACCINE (ADSORBED) IP 2007

between 14 and 20 g. Distribute them into six groups of sixteen tests must be combined in the estimate of potency.
each. If the challenge toxin to be used has not been shown to (c) Determination of antibodies in guinea-pigs
be stable or has not been adequately standardised, include
four further groups of six mice to serve as unvaccinated Preparation of serum samples. For preparation of serum
controls. The mice should all be of the same sex or the males samples, the following technique has been found suitable.
and females should be distributed equally among the groups. Invert the tubes containing blood samples 6 times and allow
to stand at 37° for 2 h, then at 4° for 2 hours, centrifuge at room
Challenge toxin. Select a preparation of tetanus toxin
temperature at 800 g for 20 min. transfer the serum to sterile
containing not less than 100 times the 50 per cent paralytic
tubes and store at a temperature below -20°. At least 40.0 per
dose per ml.
cent yield of serum is obtained by this procedure.
Preparation of the challenge toxin solutions. Immediately prior Determination of antibody titre. The ELISA and ToBI tests
to use prepare from the challenge toxin by dilution with shown below are given as examples of immunochemical
phosphate buffered saline pH 7.4 a challenge toxin solution methods that have been found suitable for the determination
containing fifty times the 50 per cent paralytic dose in each 0.5 of antibody tire.
ml. If necessary, dilute portions of this challenge toxin solution
16-, 50- and 160-fold with the same buffer solution. Determination of antibody titre in guinea-pig serum by
enzyme-linked immunosorbent assay (ELISA). Dilutions of
Determination of potency. Prepare in saline solution three test and reference sera are made on ELISA plates coated with
dilutions of the vaccine under examination and three dilutions tetanus toxoid. A positive guinea-pig serum control and a
of a solution of the Standard preparation such that, for each, negative guinea-pig serum control are included on each plate
the dilutions form a series differing by not more than 2.5-fold to monitor the assay performance. Peroxidase-conjugated
steps and in which the dilutions of intermediate concentration, rabbit or goat antibody directed against guinea-pig-IgG is
when injected subcutaneously in 0.5 ml volumes into mice, added followed by a peroxidase substrate. Optical density is
protect approximately 50 per cent of the animals from the measured and the relative antibody titre is calculated using
paralytic effects of the subcutaneous injection of the quantity the usual statistical methods.
of tetanus toxin prescribed for this test. Allocate the six
dilutions one to each of the six groups of sixteen mice and Reagents and equipment
inject subcutaneously 0.5 ml of each dilution into each mouse ELISA plates: 96 wells, columns 1-12, rows A-H.
in the group to which the dilution is allocated. After 28 days
Clostridium tetani guinea-pig antiserum (for vaccines-human
inject each animal subcutaneously with 0.5 ml of the challenge
use) reference preparation (positive control serum).
toxin solution containing fifty times the 50 per cent paralytic
dose. If necessary, allocate the challenge toxin solution and Peroxidase conjugate. Peroxidase-conjugated rabbit or goat
the three dilutions made from it one to each of the four groups antibody directed against guinea-pig IgG.
of six mice and inject subcutaneously 0.5 ml of each toxin Tetanus toxoid.
solution into each mouse in the group to which that toxin
solution is allocated. Count the number of mice without Carbonate coating buffer pH 9.6. Dissolve 1.59 g of anhydrous
paralysis 4 days after injection of the challenge toxin and sodium carbonate and 2.93 g of sodium hydrogen carbonate
calculate the potency of the vaccine under examination relative in 1000 ml of water. Distribute into 150 ml bottles and sterilise
to the potency of the Standard preparation on the basis of the by autoclaving at 121° for 15 min.
numbers of animals without paralysis in each of the six groups Phosphate buffered saline pH 7.4 (PBS). Dissolve with stirring
of sixteen, using standard statistical methods. 80.0 g of sodium chloride, 2.0 g of potassium dihydrogen
The test is not valid unless (a) for both the vaccine under phosphate, 14.3 g of disodium hydrogen phosphate dihydrate
examination and the Standard preparation, the 50 per cent and 2.0 g of potassium chloride in 1000 ml of water. Store at
protective doses lie between the largest and smallest doses of room temperature to prevent crystallisation. Dilute to 10 times
the preparations given to the mice; (b) if applicable, the number its volume with water before use.
of paralysed animals among the four groups of six injected Citric acid solution. Dissolve 10.51 g of citric acid in 1000 ml
with the challenge toxin solution and its dilutions indicate of water and adjust the solution to pH 4.0 with a 400 g/l solution
that the challenge was approximately 50 times the 50 per cent of sodium hydroxide.
paralytic dose; (c) the fiducial limits of the assay lie between Washing buffer. PBS containing 0.5 g/l of polysorbate 20.
50.0 per cent and 200.0 per cent of the estimated potency; (d)
the statistical analysis shows no deviation from linearity or Diluent block buffer. PBS containing 0.5 g/l of polysorbate 20
parallelism. The test may be repeated any number of times but and 25 g/l of dried skimmed milk.
when more than one test is performed the results of all valid Peroxidase substrate. Shortly before use, dissolve 10 mg of

90
IP 2007 TETANUS VACCINE (ADSORBED)

diammonium 2,2'-azinobis(3-ethylbenzothiazoline-6- Flat-bottomed ELISA plates.


sulphonate) (ABTS) in 20 ml of citric acid solution. Tetanus toxin or tetanus toxoid.
Immediately before use add 5 µl of strong hydrogen peroxide
solution. Clostridium tetani guinea-pig antiserum (for vaccines-human
use) reference preparation.
Method
Equine anti-tetanus IgG.
The description below is given as an example of a suitable
plate lay-out but others may be used. Wells 1A-H are for Peroxidase-conjugated equine anti-tetanus IgG.
negative control serum and wells 2A-H and 3A-H are for Carbonate buffer pH 9.6. Dissolve 1.5 g of anhydrous sodium
positive control serum for assay monitoring. Wells 4-12A-H carbonate, 2.39 g of sodium hydrogen carbonate and 0.2 g of
are for test samples. sodium azide in 1000 ml of water, adjust to pH 9.6 and autoclave
Coat each well of the ELISA plates with 100 µl of tetanus at 121° for 20 min.
toxoid solution (0.5 Lf/ml in carbonate coating buffer). Sodium acetate buffer pH 5.5. Dissolve 90.2 g of anhydrous
Allow to stand overnight at 4° in a humid atmosphere. To sodium acetate in 900 ml of water, adjust to pH 5.5 using a
avoid interference from temperature gradient, do not stack saturated solution of citric acid monohydrate and dilute to
more than 4 plates high. On the following day, wash the plates 1000 ml with water.
thoroughly with washing buffer. Block the plates by addition Phosphate buffered saline pH 7.2 (PBS). Dissolve 135.0 g of
of 100 µl of diluent block buffer to each well. Incubate in a sodium chloride, 20.55 g of disodium hydrogen phosphate
humid atmosphere at 37° for 1 h. Wash the plates thoroughly dihydrate and 4.80 g of sodium dihydrogen phosphate
with washing buffer. Place 100 µI of diluent block buffer in monohydrate in water and dilute to 15 litres with the same
each well of the plates, except those of row A. Prepare suitable solvent. Autoclave at 100° for 60 min.
dilutions of negative control serum, positive control serum
(from about 0.01 IU/ml) and test sera. Allocate the negative Diluent buffer. PBS containing 5 g/l of bovine albumin and 0.5
control serum to column 1, positive control serum to columns g/l of polysorbate 80.
2 and 3 and test sera to columns 4-12 and add 100 µI of each Block buffer. PBS containing 5 g/l of bovine albumin.
serum to the first 2 wells of the column to which it is allocated.
Tetramethylbenzidine solution. 6 g/l solution of
Using a multi channel micropipette, make twofold serial
tetramethylbenzidine in alcohol. The substance dissolves
dilutions from row B down the plate to row H by transferring
within 30-40 min at room temperature.
100 µl to the following well. Discard 100 µl from the last row so
that all wells contain 100 µI. Incubate at 37° for 2 h. Wash Peroxidase substrate. Mix 90 ml of water, 10 ml of sodium
thoroughly with washing buffer. Prepare a suitable dilution (a acetate buffer pH 5.5, 1.67 ml of tetramethylbenzidine solution
1 in 2000 dilution has been found suitable) of peroxidase and 20 µI of strong hydrogen peroxide solution.
conjugate in diluent block buffer and add 100 µI to each well. Washing solution. Tap water containing 0.5 g/l of polysorbate
Incubate at 37° in a humid atmosphere for 1 h. Wash the plates 80.
thoroughly with washing buffer. Add 100 µI of peroxidase
substrate to each well. Allow to stand at room temperature, Method
protected from light, for 30 min. Read the plates at 405 nm in Block the round-bottomed polystyrene microtitre plates by
the same order as addition of substrate was made. placing in each well 150 µI of block buffer. Cover the plates
Determination of antibody titre in guinea-pig serum by toxin- with a lid or sealer. Incubate in a humid atmosphere at 37° for
or toxoid-binding inhibition (ToBI). Tetanus toxin or toxoid is 1 h. Wash the plates thoroughly with washing solution. Place
added to serial dilutions of test and reference sera; the serum/ 100 µI of PBS in each well. Place 100 µl of reference guinea-
antigen mixtures are incubated overnight. To determine pig tetanus antitoxin in the first well of a row. Place 100 µl of
unbound toxin or toxoid, the mixtures are transferred to an undiluted test sera in the first well of the required number of
ELISA plate coated with tetanus antitoxin. Peroxidase- rows. Using a multichannel micropipette, make it two fold serial
conjugated equine anti-tetanus IgG is added followed by a dilutions across the plate (up to column 10), by transfer of 100
peroxidase substrate. Optical density is measured and the µI to the following well. Discard 100 µl from the last column so
antibody titre is calculated using the usual statistical methods. that all wells contain 100 µl. Prepare 0.1 Lf/ml solution of tetanus
A positive control serum and a negative control serum are toxin or toxoid using PBS a diluent. Add 40 µl of this solution
included on each plate to monitor assay performance. to all wells except those column 12. The wells of row 11 are a
positive control. Add 40 µl of PBS to the wells of column 12
Reagents and equipment
(negative control). Shake the plates gently and cover them
Round-bottomed, rigid polystyrene microtitre plates. with lids. Coat the ELISA plates: immediately before use make

91
TICK-BORNE ENCEPHALITIS VACCINE (INACTIVATED) IP 2007

a suitable dilution of equine anti-tetanus IgG in carbonate consistently vaccines comparable with the vaccine of proven
buffer pH 9.6 and add 100 µl to all wells. Incubate the 2 series clinical efficacy and safety in man. The virus in the final vaccine
of plates overnight in a humid atmosphere at 37°. To avoid shall not have undergone more passages from the master seed
temperature gradient effects, do not stack more than 4 plate lot than the virus in the vaccine used in clinical trials.
high. Cover the plates with lids. On the following day, wash The production method is validated to demonstrate that the
the ELISA plates thoroughly with washing solution. Block product, if tested, would comply with the test for abnormal
the plates by placing in each well 125 µl of block buffer. Incubate toxicity and immunogenicity.
at 37° in a humid atmosphere for 1 h. Wash the plates
thoroughly with washing solution. Transfer 100 µl of the pre- Substrate for virus propagation
incubation mixture from the polystyrene plates to the
corresponding wells of the ELISA plates, starting with column The virus is propagated in chick embryo cells prepared from
12 and then from 1 to 11. Cover the plates with a lid. Incubate eggs derived from a chicken flock free from specified pathogens
at 37° in a humid atmosphere for 2 h. Wash the ELISA plates (2.7.7) or in other suitable cell cultures.
thoroughly with washing solution. Make a suitable dilution
SEED LOT
(a 1 in 4000 dilution has been found suitable) of the peroxidase-
conjugated equine anti-tetanus IgG in diluent buffer. Add The strain of virus used is identified by historical records that
100 µl of the dilution to each well and cover the plates with a include information on the origin of the strain and its
lid. Incubate at 37° in a humid atmosphere for 1.5 h. Wash the subsequent manipulation. Virus seed lots are stored at or
ELISA plates thoroughly with washing solution. Add 100 µl below -60°.
of peroxidase substrate to each well. A blue colour develops.
Only a seed lot that complies with the following requirements
Incubate the plates at room temperature. Stop the reaction at
may be used for virus propagation.
a given time (within 10 min) by the addition of 100 µl of 2 M
sulphuric acid to each well in the same order as the addition Identification
of substrate. The colour changes from blue to yellow. Measure
Each seed lot is identified as containing the vaccine strain of
the absorbance (2.4.7) at 450 nm immediately after addition of
tick-borne encephalitis virus by a suitable immunochemical
the sulphuric acid or maintain the plates in the dark until
method, preferably using monoclonal antibodies.
reading.
Virus concentration. The virus concentration of each seed
(d) Any other validated serological assay in guinea-pigs/mice
lot is determined by titration in suitable cell cultures to monitor
approved by National Regulatory Authority.
consistency of production.
Labelling. The label states (1) the human dose (ml); (2) the
minimum units per single human dose or the minimum Extraneous agents (2.7.3). Each seed lot complies with the
International Units per single human dose if potency test done requirements for extraneous agents in viral vaccines for human
by challenge method or both; (3) the name and the amount of use; the tests in cell cultures are carried out in human and
the adsorbent and preservative; (4) that the vaccine must be simian cells only.
shaken before use; (5) that the vaccine is not to be frozen. PROPAGATION AND HARVEST
All processing of the cell cultures if performed under aseptic
conditions in an area where no other cells are being handled.
Tick-borne Encephalitis Vaccine Serum and trypsin used in the preparation of cell suspensions
(Inactivated) and media used must be shown to be free from extraneous
agents. The cell culture media may contain a pH indicator
Tick-borne Encephalitis Vaccine (Inactivated) is a liquid such as phenol red and approved antibiotics at the lowest
preparation of a suitable strain of tick-borne encephalitis virus effective concentration. At least 500 ml of the cell cultures
grown in cultures of chick-embryo cells or other suitable cell employed for vaccine production is set aside as uninfected
cultures and inactivated by a suitable, validated method. cell cultures (control cells).
Production Only a single harvest that complies with the following
requirements may be used in the preparation of the inactivated
General provisions
harvest.
The vaccine complies with the General Requirements of
Vaccines for Human Use. Identification
Production of the vaccine is based on a virus seed lot system. The single harvest is shown to contain tick-borne encephalitis
The production method shall have been shown to yield virus by a suitable immunochemical method, preferably using

92
IP 2007 TICK-BORNE ENCEPHALITIS VACCINE (INACTIVATED)

monoclonal antibodies, or by virus neutralization in cell Specific activity. Determine the antigen content of the purified,
cultures. inactivated harvest by a suitable immunochemical method
Sterility (2.2.11). Complies with the test for sterility carried (2.2.14). Determine the total protein content by a suitable
out using 10 ml for each medium. method. The specific activity, calculated as the antigen content
per unit mass of protein, is within the limits approved for the
Mycoplasma (2.7.4). Complies with the test for mycoplasmas specific product.
carried out using 1 ml for each medium.
FINAL BULK VACCINE
Control cells. The control cells comply with the tests for
extraneous agents (2.7.3). If the vaccine is produced using a The final bulk vaccine is prepared from one or more purified,
cell-bank system, the control cells comply with a test for inactivated harvests.
identification.
Only a final bulk vaccine that complies with the following
Virus concentration. Determine the virus concentration by requirement may be used in the preparation of the final lot.
titration in suitable cell culture to monitor consistency of
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
production.
for each sterility medium.
Inactivation
FINAL LOT
To avoid interference, viral aggregates are removed by filtration
immediately before the inactivation process. The virus Only a final lot that is satisfactory with respect to each of the
suspension is inactivated by a validated method; the method requirements given below under Identification, Tests and
shall have been shown to be consistently capable of Assay may be released for use. Provided that the tests for free
inactivating tick-borne encephalitis virus without destroying formaldehyde, bovine serum albumin (where applicable) and
the antigenic and immunogenic activity; as part of the pyrogens and the assay have been carried out satisfactory
validation studies, an inactivation curve is plotted representing results on the final bulk vaccine, they may be omitted on the
residual live virus concentration measured on not fewer than final lot.
three occasions. If formaldehyde is used for inactivation, the
presence of an excess of free formaldehyde is verified at the Identification
end of the inactivation process. The vaccine is shown to contain tick-borne encephalitis virus
Only an inactivated harvest that complies with the following antigen by a suitable immunochemical method using specific
requirements may be used in the preparation of the final bulk antibodies or by the mouse immunogenicity test described
vaccine. under Assay.
Residual infective virus. Inoculate a quantity of the inactivated
Tests
harvest equivalent to not less than ten human doses of vaccine
in the final lot into primary chicken fibroblast cell cultures, or Aluminium (2.3.9). Maximum 1.25 mg per single human dose,
other cells shown to be at least as sensitive to tick-borne if aluminium hydroxide or hydrated aluminium phosphate is
encephalitis virus with not less than 3 cm2 of cell sheet per ml used as the adsorbent.
of inoculum. Incubate at 37 ± 1° for 14 days. No cytopathic
effect is detected at the end of the incubation period. Collect Free formaldehyde (2.3.20). Maximum 0.1 g/l.
the culture fluid and inoculate 0.03 ml intracerebrally into each Bovine serum albumin. If bovine serum albumin has been
of not fewer than ten mice about 4 weeks old. Observe the used during production, the vaccine contains not more than
mice for 14 days. They show no evidence of tick-borne 50 ng per single human dose, determined by a suitable
encephalitis virus infection. immunochemical method (2.2.14).
Purification Sterility (2.2.11). Complies with the test for sterility.
Several inactivated single harvests may be pooled before Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
concentration and purification by suitable methods, preferably into each rabbit, per kg of body mass, one dose of vaccine.
by continuous-flow, sucrose density-gradient centrifugation.
Only a purified, inactivated harvest that complies with the Assay
following requirements way be used in the preparation of final The potency is determined by comparing the dose necessary
bulk vaccine. to protect a given proportion of mice against the effects of a
Sterility ( 2.2.11). Complies with the test for sterility, carried lethal dose of tick-borne encephalitis virus, administered
out using 10 ml for each medium. intraperitoneally, with the quantity of a reference preparation

93
TUBERCULIN PURIFIED PROTEIN DERIVATIVE IP 2007

of tick-borne encephalitis vaccine necessary to provide the mean potency and the fiducial limits (P = 0.95) for the mean
same protection. For this comparison an approved reference potency; compute weighed means with the inverse of the
preparation and a suitable preparation of tick-borne squared standard error as weights. The vaccine complies with
encephalitis virus from an approved strain for use as the the test if the estimated potency is not less than that approved
challenge preparation are necessary. by the competent authority, based on data from clinical efficacy
The following is cited as an example of a method that has been trials.
found suitable for a given vaccine. Labelling. The label states (1) the strain of virus used in
preparation; (2) the type of cells used for production of the
Selection and distribution of test animals. Use healthy mice
vaccine.
weighing between 11 and 17 g derived from the same stock.
Distribute the mice into not less than six groups of a suitable
size to meet the requirements for validity of the test; for titration
of the challenge suspension, use not fewer than four groups Tuberculin Purified Protein Derivative
of ten mice. Use mice of the same sex or distribute males and
females equally between groups. Tuberculin PPD; Tuberculin Purified Protein Derivative for
Human Use
Determination of potency of the vaccine. Prepare not less
than three suitable dilutions of the vaccine under examination Tuberculin Purified Protein Derivative is a preparation made
and of the reference preparation; in order to comply with from the heat-treated products of growth and lysis of one or
validity criteria four to five dilutions will usually be necessary. more strains of Mycobacterium tuberculosis that reveal
Prepare dilutions such that the most concentrated suspension delayed hypersensitivity in animals sensitised by a micro-
is expected to protect more than 50 per cent of the animals and organism of the same species.
the least concentrated suspension less than 50.0 per cent.
Production
Allocate each dilution to a different group of mice and inject
subcutaneously into each mouse 0.2 ml of the dilution allocated It is prepared from the water-soluble fraction obtained by
to its group. Seven days later make a second injection using heating in free-flowing steam or in an autoclave and
the same dilution scale. 14 days after the second injection subsequently filtering cultures of the mycobacteria grown in
prepare a suspension of the challenge virus containing not a suitable liquid medium. The active fraction in the filtrate,
less than 100 LD50 in 0.2 ml. Inject 0.2 ml of this virus which is predominantly protein, is separated by precipitation,
suspension intraperitoneally into each vaccinated mouse. To washed and redissolved. The preparation is free from
verify the challenge dose, prepare a series of not fewer than mycobacteria. An antimicrobial preservative that does not give
three dilutions of the challenge virus suspension at not greater rise to false positive reactions, such as 0.5 per cent w/v of
than one-hundredfold intervals. Allocate the challenge phenol, and a suitable stabiliser may be added. Phenol is not
suspension and the four dilutions, one to each of the five added to preparations that are to be freeze-dried. The final
groups of ten mice, and inject intraperitoneally into each mouse sterile product is distributed into sterile glass containers which
0.2 ml of the challenge suspension or the dilution allocated to are then sealed so as to prevent microbial contamination or
its group. Observe the animals for 21 days after the challenge alternatively it is freeze-dried and the containers subsequently
and record the number of mice that die in the period between sealed.*
7 and 21 days after the challenge.
* To ensure availability of a preparation of uniform potency, Tuberculin
Calculations. Calculate the results by the usual statistical Purified Protein Derivative is produced and issued by the Statens
methods for an assay with quantal responses (5.7). Serum Institute ,Denmark as a powder to be reconstituted as stated
on the label.
Validity criteria. The test is not valid unless (1) the
concentration of the challenge virus is not less than 100 LD50; The preparation may be issued either as a sterile liquid or as a
(2) for both the vaccine under examination and the reference freeze-dried product. If issued as a liquid, it is in a ready-to-
preparation the 50.0 per cent protective dose (PD50) lies use form and 0.1 ml constitutes one intradermal dose
between the largest and smallest doses given to the mice; (3) containing appropriate number of Units. If issued as a freeze-
the statistical analysis shows a significant slope and no dried product, it should yield a ready-to-use preparation when
significant deviation from linearity and parallelism of the dose- reconstituted as per manufacturer’s instructions.
response lines; (4) the fiducial limits (P = 0.95) are not less Description. A colourless or pale, straw-coloured liquid, or
than 33.0 per cent and not more than 300.0 per cent of the dry cream-coloured powder, or pellet.
estimated potency.
The preparation, reconstituted if necessary as stated on the
Potency requirement. Include all valid tests to estimate the label, complies with the following requirements.

94
IP 2007 TUBERCULIN PURIFIED PROTEIN DERIVATIVE

Identification derivative with the dose of the appropriate Standard


Preparation necessary to give the same effect.
A. When progressively increasing doses are injected
intradermally into specifically sensitised guinea-pigs, reactions The estimated potency is not less 80 per cent and not more
occur at the points of injection, varying from erythema to than 125 per cent of the stated potency. The fiducial limits of
necrosis. When similar injections are administered to non- error are not less than 64 per cent and not more than 156 per
sensitised guinea-pigs no such reactions occur. cent of the stated potency.

B. The potency test described below serves as a test for Standard Preparation
identity if it is performed on material from the final containers.
The Standard Preparation is the 1st International Standard for
Tests Tuberculin, purified protein derivative (PPD), mammalian,
established in 1951, consisting of PPD derived from cultures
pH (2.4.24). 6.5 to 7.5. of M. tuberculosis (supplied in ampoules containing 5,00,000
Phenol (if present) (2.3.36). Not more than 0.5 per cent w/v. Units) or another suitable preparation the potency of which
has been determined in relation to the International Standard.
Sterility (2.2.11). Complies with the tests for sterility.
Method
Potency. Carry out the biological assay of tuberculin purified
protein derivative described below. Sensitise 12 guinea-pigs each weighing not less than 400 g by
the intramuscular injection of a total of about 0.5 ml of a
Tuberculin Purified Protein Derivative in freeze-dried form
suspension in a suitable mineral oil with or without emulsifier
complies with the following additional requirements.
and containing 0.1 mg per ml of heat-inactivated, dried
Live mycobacteria mycobacteria of the same type as that used in the preparation
of tuberculin. Use phosphate-buffered saline pH 7.4
A. Inject 5.0 ml intraperitoneally or subcutaneously into each containing 0.005 per cent w/v of polysorbate 80 to prepare
of two guinea-pigs weighing between 300 and 400 g. Observe the dilutions of the Standard preparation and of the
the animals for not less than 42 days. Kill the animals and preparation under examination and to reconstitute freeze-dried
carry out an autopsy. No guinea-pig shows signs of infection preparations containing no stabiliser. Not less than 1 month
with mycobacteria. and not more than 6 months later carry out the following
B. Carry out tests for live mycobacteria in the preparation test :
under examination using suitable culture media. No growth of Shave the flanks to provide space for at least three reactions
mycobacteria should occur. on each side, but not more than a total of 12 injection sites per
Sensitising effect. Inject intradermally into each of three animal. Use at least three doses of the Standard preparation
guinea-pigs three times, at intervals of 5 days, a dose of the and at least three doses of the preparation under examination,
preparation under examination containing about 500 Units in the highest dose being about 10 times as strong as the lowest.
a volume of 0.1 ml. Two to three weeks after the third injection Dilute the preparations so that the lesions produced are 8 to
inoculate the same dose intradermally into these animals and 25 mm in diameter. Allocate the doses to the available sites in
into a control group of three guinea-pigs of the same weight a random manner, in a Latin square design. Inject each dose
but that have not had any previous injections of tuberculin. intradermally in the same volume (0.1 or 0.2 ml) at the sites to
The reactions of the two groups are not significantly different which it has been allocated. Measure the diameters of the
after 48 to 72 hours. lesions after 24 to 48 hours and calculate the result of the test
using standard statistical methods on the basis that the lesion
Toxicity. Inject subcutaneously into 2 healthy guinea-pigs,
diameters are directly proportional to the logarithm of the
weighing not less than 250 g and which have not previously
concentration of the tuberculin.
been treated with any material which will interfere with the
test, 0.5 ml of a solution containing 1,00,000 Units per ml. No Storage. Store in light-resistant containers at a temperature
harmful effects are produced within 7 days. between 2° and 8°. It should not be allowed to freeze.
Labelling. The label states (1) the number of Units per dose of
Biological assay
0.1 ml or per ml or per mg; (2) the total volume in the container
The potency of tuberculin purified protein derivative is (for liquid preparation); (3) the nature and quantity of the
determined by comparing the dose necessary to reveal delayed reconstituting liquid (for the freeze-dried preparation); (4) the
hypersensitivity in guinea-pigs or other animals name and proportion of any added substances; (5) the species
hypersensitised with mycobacteria of the same type as that or strain used; (6) the storage conditions; (7) the date after
used in the preparation of the tuberculin purified protein which the contents are not intended to be used; (8) that care

95
TYPHOID (STRAIN TY 21A) VACCINE, LIVE (ORAL) IP 2007

should be taken to avoid inhaling the powder (for the freeze- the growth conditions, strain Ty 21a does not agglutinate to
dried preparation). Vi antiserum. Strain Ty 21a agglutinates to H:d flagellar
antiserum.
Biochemical markers
Typhoid (Strain Ty 21a) Vaccine, Live Strian Ty 21a does not produce hydrogen sulphide on Kligler
(Oral) iron agar. This property serves to distinguish Ty 21a from
other galactose-epimerase-negative S. typhi strains.
Typhoid Vaccine (Live, Oral, strain Ty 21a) is a freeze-dried
preparation of the live Salmonella typhi strain Ty 21a grown Cell growth
in a suitable medium. When presented in capsules, the vaccine Strain Ty 21a cells lyse when grown in the presence of 1.0 per
complies with the tests stated under Capsules. cent of galactose.

Production PROPAGATION AND HARVEST


The bacteria from the working seed lot are multiplied in a
Choice of vaccine strain preculture, subcultured once and are then grown in a suitable
medium containing 0.001 per cent of galactose at 30° for 13 to
The main characteristic of the strains is the defect of the
15 hours. The bacteria are harvested. The harvest must be
enzyme uridine diphosphate-galactose-4 epimerase. The
free from contaminating micro-organisms.
activities of galactopermease, galactokinase and galactose-1-
phosphate uridyl-transferase are reduced by 50 to 90 per cent. Only a single harvest that complies with the following
Whatever the growth conditions, the strain does not contain requirements may be used for the preparation of the freeze-
Vi antigen. The strain agglutinates to anti-O:9 antiserum only dried harvest.
if grown in medium containing galactose. It contains the pH (2.4.24). 6.8 to 7.5.
flagellar H:d antigen and does not produce hydrogen sulphide
on Kligler iron agar. The strain is nonvirulent for mice. Cells of Optical density
strain Ty 21a lyse if grown in the presence of 1.0 per cent of
galactose. The optical density of the culture, measured at 546 nm, is 6.5
to 11.0. Before carrying out the measurement, dilute the culture
SEED LOT so that a reading in the range 0.1 to 0.5 is obtained and correct
the reading to take account of the dilution.
The vaccine is prepared using a seed-lot system. The working
seed lots represent not more than one subculture from the Identification
master seed lot. The final vaccine represents not more than
four subcultures from the original vaccine on which were made Culture bacteria on an agar medium containing 1.0 per cent of
the laboratory and clinical tests showing the strain to be galactose and bromothymol blue. Light blue, concave
suitable. colonies, transparent due to lysis of cells, should be found.
No yellow colonies (galactose-fermenting) should be found.
Only a master seed lot that complies with the following
requirements may be used in the preparation of working seed FREEZE-DRIED HARVEST
lots.
The harvest is mixed with a suitable stabilizer and freeze-dried
Galactose metabolism by a process that ensures the survival of at least 10.0 per cent
In a spectrophotometric assay, no activity of the enzyme of the bacteria and to a water content shown to be favourable
uridine diphosphate-galactose-4-epimerase is found in the to the stability of the vaccine. No antimicrobial preservative is
cytoplasm of strain Ty 21a compared to strain Ty 2. added to the vaccine.
Biosynthesis of lipopolysaccharide Only a freeze-dried harvest that complies with the following
tests may be used for the preparation of the final bulk.
Lipopolysaccharides are extracted by the hot-phenol method
and examined by size-exclusion chromatography (2.4.16). Strain Identification
Ty 21a grown in medium free of galactose shows only the
rough (R) type of lipopolysaccharide. Culture bacteria are examined on an agar medium containing
1.0 per cent of galactose and bromoethymol blue. Light blue,
Serological characteristics concave colonies, transparent due to lysis of cells, should be
Strain Ty 21a grown in a synthetic medium without galactose found. No yellow colonies (galactose-fermenting) should be
does not agglutinate to specific anti-O:9 antiserum. Whatever found.

96
IP 2007 TYPHOID POLYSACCHARIDE VACCINE

Number of live bacteria Labelling. The label states (1) the minimum number of live
11
Not less than 1 x 10 live S. typhi strain Ty 21a per gram. bacteria per dosage unit; (2) that the vaccine is for oral use
only.
Water (2.4.43). 1.5 to 4.0 per cent, determined by the semi-
micro determination of water or any other validated method.
FINAL BULK VACCINE Typhoid Polysaccharide Vaccine
The final bulk vaccine is prepared by aseptically mixing one or Typhoid Polysaccharide Vaccine is a preparation of purified
more freeze-dried harvests with a suitable sterile excipient. Vi capsular polysaccharide obtained from Salmonella typhi
Only a final bulk that complies with the following requirement Ty2 strain or some other suitable strain of known origin and
may be used in the preparation of the final lot. history that has the capacity to produce Vi polysaccharide,
and which have been characterized by suitable biochemical,
Number of live bacteria. Not less than 40 × 109 live S. typhi physicochemical, serological or molecular methods.
strain Ty 21a per gram.
Capsular polysaccharide is a partly 3-O-acetylated repeated
FINAL LOT units of 2-acetylamino-2-deoxy-D-galactopyranuronic acid
The final bulk vaccine is distributed under aseptic conditions with á-(1?4)-linkages.
into capsules with a gastro-resistant shell or into suitable
containers.
Production

Only a final lot that is satisfactory with respect to Identification, General provisions
Tests and Number of live bacteria will be released for use,
The production of Vi polysaccharide is based on a defined
except that in the determination of number of live bacteria
seed-lot system. The method of production shall have been
each dosage unit must contain not less than 4 x 109 live
shown to yield consistently Vi polysaccharide typhoid
bacteria.
vaccines of adequate immunogenicity and safety in man. The
Identification production method is validated to demonstrate that the product,
if tested, would comply with the tests for abnormal toxicity.
Culture bacteria from the vaccine under examination on an
agar medium containing 1.0 per cent of galactose and SEED LOT
bromothymol blue. Light blue, concave colonies transparent
due to lysis of cells, should be found. No yellow colonies The strain of S. typhi used for the master seed lot shall be
(galactose-fermenting) should be found. identified by historical records that include information on its
origin and by its biochemical and serological characteristics.
Tests Cultures from the working seed lot shall have the same
Microbial contamination (2.2.9). Carry out the test using characteristics as the strain that was used to prepare the master
suitable selective media. Determine the total viable count using seed lot, shall be demonstrated along with adequate
the plate-count method. The number of contaminating micro- documentation.
organisms per dosage unit is not greater than 102 bacteria and Only a strain that has the following characteristics may be
20 fungi. No pathogenic bacterium, particularly Escherichia used in the preparation of the vaccine: (a) stained smears from
coli, Staphylococcus aureus, Pseudomonas aeruginosa, and a culture are typical of S. typhi; (b) the culture utilises glucose
no Salmonella other than strain Ty 21a are found. without production of gas; (c) colonies on agar are oxidase-
Water (2.4.43). 1.5 to 4.0 per cent. negative; (d) a suspension of the culture agglutinates
specifically with an appropriate Vi antiserum or colonies form
Number of live bacteria haloes on an agar plate containing a suitable Vi antiserum.
Carry out the test using not less than five dosage units. PROPAGATION AND HARVEST
Homogenise the contents of the dosage units in a 0.9 per cent
w/v solution of sodium chloride at 4° using a mixer in a cold The working seed lot is cultured on a solid medium, which
room with sufficient glass beads to emerge from the liquid. may contain blood-group substances, or a liquid medium; the
Immediately after homogenization prepare a suitable dilution inoculum obtained is transferred to a liquid medium, which is
of the suspension using cooled diluent and inoculate brain used to inoculate the final medium. The liquid medium used
heart infusion agar, incubate at 36 ± 1° for 20 to 36 hours. The and the final medium are semi-synthetic, free from substances
vaccine contains not less than 2 x 109 live S. typhi Ty 21a that are precipitated by cetrimonium bromide and do not
bacteria per dosage unit. contain blood-group substances or high-molecular-mass

97
TYPHOID POLYSACCHARIDE VACCINE IP 2007

polysaccharides, unless it has been demonstrated that they Bacterial endotoxins (2.2.3). Determine by a suitable method,
are removed by the purification process. The bacterial purity the content should not be more than 150 IU per mg of
of the culture is verified by microscopic examination of Gram- polysaccharide.
stained smears and by inoculation into appropriate media.
The culture is then inactivated at the beginning of the FINAL BULK VACCINE
stationary phase by the addition of formaldehyde. Bacterial One or more batches of purified Vi polysaccharide are
cells are eliminated by centrifugation; the polysaccharide is dissolved in a suitable solvent, which may contain an
precipitated from the culture medium by addition of antimicrobial preservative, so that the volume corresponding
hexadecyltrimethylammonium bromide (cetrimonium to one dose contains 25 µg of polysaccharide and the solution
bromide). The precipitate is harvested and may be stored at is isotonic with blood (250 mosm/kg to 350 mosm/kg).
or below -20° before purification.
Only a final bulk vaccine that complies with the following
Purified Vi Polysaccharide requirements may be used in the preparation of the final lot;
The polysaccharide is purified, after dissociation of the Sterility (2.2.11). Carry out test for sterility using 10 ml of
polysaccharide/cetrimonium bromide complex, using suitable bulk for each sterility medium.
procedures to eliminate successively nucleic acids, proteins
Antimicrobial preservative. Where applicable, determine the
and lipopolysaccharides. The polysaccharide is precipitated
amount of antimicrobial preservative by a suitable chemical
in its salt form and dried at 5+3°; the powder obtained
method. The content is not less than 85.0 per cent and not
constitutes the purified Vi polysaccharide. The loss on drying
greater than 115.0 per cent of the intended amount. If phenol
is determined by thermogravimetry, Karl Fischer or any other
has been used in the preparation, the content is not more than
suitable method and is used to calculate the results of the
2.5 g/l.
chemical tests shown below with reference to the dried
substance.
FINAL LOT
Only those pools of purified Vi polysaccharide that comply
The final bulk vaccine is distributed and filled aseptically into
with the following requirements may be used in the preparation
sterile containers. The containers are closed so as to prevent
of the final bulk:
contamination.
Protein (2.7.1). Not more than 10 mg per gram of
polysaccharide, calculated with reference to the dried Only a final lot that is satisfactory with respect to each of the
substance. requirements prescribed below under Identification, Tests and
Assay and with the requirements for Bacterial endotoxins may
Nucleic acids (2.7.1). Not more than 20 mg per gram of be released for use. Provided the tests for free formaldehyde
polysaccharide, calculated with reference to the dried and antimicrobial preservative have been carried out on the
substance. final bulk vaccine, they may be omitted on the final lot.
O-Acetyl groups (2.7.1). Not less than 2 mmol per gram of
The vaccine contains minimum of 25 µg purified Vi-
polysaccharide, calculated with reference to the dried
Polysaccharide per dose of 0.5 ml.
substance.
Molecular size. Examine by gel filtration or size-exclusion Identification
chromatography (2.4.16) using cross-linked agarose for
chromatography. Use a column 0.9 m long and 16 mm in internal Carry out an identification test using a suitable
diameter equilibrated with a solvent having an ionic strength immunochemical method (2.2.14).
of 0.2 mol per kg and a pH of 7.0 to 7.5. Apply about 5 mg of
Tests
polysaccharide in a volume of 1 ml to the column and elute at
about 20 ml/h. Collect fractions of about 2.5 ml. Determine the Sterility (2.2.11). Complies with the tests for sterility.
point corresponding to K0 = 0.25 and make two pools
consisting of fractions eluted before and after this point. Abnormal toxicity (2.2.1). Complies with the test for abnormal
Determine O-acetyl groups on the two pools. Not less than 50 toxicity.
per cent of the polysaccharide is found in the pool containing pH (2.4.24). 6.5 to 7.5.
fractions eluted before K0 = 0.25.
O-Acetyl groups (2.7.1). 0.085 (± 25 per cent) µmol per dose
Identification (25 µg of polysaccharide).
Carry out an identification test using a suitable Test solution. Place 3 ml of the vaccine in each of three tubes
immunochemical method (2.2.14). (two reaction solutions and one correction solution).

98
IP 2007 TYPHOID VACCINE (FREEZE DRIED)

Reference solutions. Dissolve 0.150 g of acetylcholine Typhoid Vaccine


chloride in 10 ml of water (stock solution containing 15 g/l of
acetylcholine chloride). Immediately before use, dilute 0.5 ml Typhoid Vaccine is a sterile suspension of inactivated
of the stock solution to 50 ml with water (working dilution Salmonella typhi containing not less than 5 x 108 and not
containing 150 µg/ml of acetylcholine chloride). In ten tubes, more than 1 x 109 bacteria per human dose. The human dose
place in duplicate (reaction and correction solutions) 0.1 ml, does not exceed 1.0 ml.
0.2 ml, 0.5 ml, 1.0 ml and 1.5 ml of the working dilution.
Production
Prepare a blank using 3 ml of purified water.
The vaccine is prepared using a seed-lot system from a
Make up the volume in each tube to 3 ml with water. Add 0.5 suitable strain of S. typhi such as, Ty 2. The final vaccine
ml of a mixture of 1 volume of water and 2 volumes of dilute represents not more than 3 subcultures from the strain on
hydrochloric acid to each of the correction tubes and to the which were made the laboratory and clinical tests that showed
blank. Add 1.0 ml of alkaline hydroxylamine solution to each it to be suitable. The bacteria are inactivated by acetone, by
tube. Allow the reaction to proceed for exactly 2 min and add formaldehyde, by phenol or by heating or by a combination
0.5 ml of a mixture of 1 volume of water and 2 volumes of of the last two methods.
dilute hydrochloric acid to each of the reaction tubes. Add
The production method is validated to demonstrate that the
0.5 ml of a 20 per cent w/vl solution of ferric chloride in 0.2M
product, if tested, would comply with the test for abnormal
hydrochloric acid to each tube, stopper the tubes and shake
toxicity (2.2.1) modified to the extent that 0.5 ml of the vaccine
vigorously to remove bubbles.
is injected subcutaneously or intramuscularly or
Measure the absorbance of each solution at 540 nm using the intraperitoneally into each mouse and 1.0 ml into each guinea-
blank as the compensation liquid. For each reaction solution, pig.
subtract the absorbance of the corresponding correction
solution. Draw a calibration curve from the corrected Identification
absorbance for the five reference solutions and the
corresponding content of acetylcholine chloride and read from It is identified by specific agglutination.
the curve the content of acetylcholine chloride in the test Phenol (2.3.36). If phenol has been used in the preparation,
solution for each volume tested. Calculate the mean of the the concentration is not more than 0.5 per cent w/v.
two values.
Antigenic power. When injected into susceptible laboratory
1 mol of acetylcholine chloride (181.7 g) is equivalent to 1 mol animals, it elicits anti-O, anti-H and, to a lesser extent, anti-Vi
of O-acetyl (43.05 g). agglutinins.

Free formaldehyde (2.3.20). Maximum 0.2 g/l. Sterility (2.2.11). Complies with the test for sterility.
Labelling. The label states (1) the method used to inactivate
Antimicrobial preservative. Where applicable, determine the
the bacteria; (2) the number of bacteria per human dose.
amount of antimicrobial preservative by a suitable chemical
method. The content is not less than 85.0 per cent and not
greater than 115.0 per cent of the intended amount.
If phenol has been used in the preparation, the content is not Typhoid Vaccine (Freeze Dried)
more than 2.5 g/l. Freeze Dried Typhoid Vaccine is a freeze-dried preparation of
inactivated Salmonella typhi. The vaccine is reconstituted as
Assay
stated on the label to give a uniform suspension containing
Determine Vi polysaccharide content by a suitable not less than 5 x 108 and not more than 1 x 109 bacteria per
immunochemical method (2.2.14) using a reference purified human dose. The human dose does not exceed 1.0 ml of the
polysaccharide. The estimated amount of polysaccharide per reconstituted vaccine.
dose is 80.0 per cent to 120.0 per cent of the content stated on
the label. The fiducial limits of error (P = 0.95) of the estimated Production
amount of polysaccharide are not less than 80.0 per cent and
The vaccine is prepared from a seed-lot system from a suitable
not more than 120.0 per cent.
strain of S. typhi, such as Ty 2. The final vaccine represents
Labelling. The label states (1) the number of micrograms of not more than 3 subcultures from the strain on which were
polysaccharide per human dose (25 µg); (2) the total quantity made the laboratory and clinical tests that showed it to be
of polysaccharide in the container. suitable. The bacteria are inactivated either by acetone or by

99
VARICELLA VACCINE, LIVE IP 2007

formaldehyde or by heat. Phenol is not used in the preparation. origin of the strain and its subsequent manipulation. The virus
The vaccine is distributed into sterile containers and freeze- shall at no time have been passaged in continuous cell lines.
dried to a moisture content favourable to the stability of the Seed lots are prepared in the same kind of cells as those used
vaccine. for the production of the final vaccine. To avoid the
The production method is validated to demonstrate that the unnecessary use of monkeys in the test for neurovirulence,
product, if tested, would comply with test for abnormal toxicity virus seed lots are prepared in large quantities and stored at
(2.2.1), modified to the extent that 0.5 ml of the vaccine is temperatures below -20°, if freeze-dried, or below -60°, if not
injected subcutaneously or intramuscularly or freeze-dried.
intraperitoneally into each mouse and 1.0 ml into each guinea- Only a virus seed lot that complies with the following
pig. requirements may be used for virus propagation.
Identification Identification
The vaccine reconstituted as stated on the label is identified The master and working seed lots are identified as varicella
by specific agglutination. virus by serum neutralisation in cell culture, using specific
Antigenic power. When injected into susceptible laboratory antibodies.
animals, the reconstituted vaccine elicits anti-O, anti-H and, Virus concentration. The virus concentration of the master
to a lesser extent, anti-Vi agglutinins. and working seed lots is determined as prescribed under Assay
Sterility (2.2.11). The reconstituted vaccine complies with the to monitor consistency of production.
tests for sterility. Extraneous agents (2.7.3). Complies with the requirements for
Water. Not more than 5.0 percent. seed lots for live virus vaccines; a sample of 50 ml is taken for
the test in cell cultures.
Labelling. The label states (1) the method used to inactivate
the bacteria; (2) the number of bacteria per human dose; (3) Neurovirulence (2.7.5). Complies with the test for
that the vaccine should be used within 8 hours of neurovirulence of live virus vaccines.
reconstitution. PROPAGATION AND HARVEST
All processing of the cell bank and subsequent cell cultures is
done under aseptic conditions in an area where no other cells
Varicella Vaccine, Live are handled. Approved animal (but not human) serum may be
used in the media. Serum and trypsin used in the preparation
Varicella Vaccine (Live) is a freeze-dried preparation of a suitable
of cell suspensions and media are shown to be free from
attenuated strain of Herpesvirus varicellae.
extraneous agents. The cell culture medium may contain a pH
Production indicator such as phenol red and approved antibiotics at the
lowest effective concentration. It is preferable to have a
General provisions substrate free from antibiotics during production. 5.0 per cent,
but not less than 50.0 ml, of the cell cultures employed for
The production of vaccine is based on a virus seed-lot system vaccine production is set aside as uninfected cell cultures
and a cell-bank system. The production method shall have (control cells). The infected cells constituting a single harvest
been shown to yield consistently live varicella vaccines of are washed, released from the support surface and pooled.
adequate immunogenicity and safety in man. The virus in the The cell suspension is disrupted by sonication.
final vaccine shall not have been passaged in cell cultures
beyond the 38th passage from the original isolated virus. Only a virus harvest that complies with the following
requirements may be used in the preparation of the final bulk
The production method is validated to demonstrate that the vaccine.
product, if tested, would comply with the test for safety and
efficacy. Identification
Substrate for virus propagation The virus harvest contains virus that is identified as varicella
The virus is propagated in human diploid cells (2.7.2). virus by serum neutralisation in cell culture, using specific
antibodies.
SEED LOT
Virus concentration. The concentration of infective virus in
The strain of varicella virus shall be identified as being suitable virus harvests is determined as prescribed under assay to
by historical records which shall include information on the monitor consistency of production and to determine the

100
IP 2007 VIPER VENOM

dilution to be used for the final bulk vaccine. Labelling. The label states (1) the strain of virus used for the
Extraneous agents (2.7.3). Use 50 ml for the test in cell cultures. preparation of the vaccine; (2) the type and origin of the cells
used for the preparation of the vaccine; (3) that contact with
Control cells. The control cells of the production cell culture disinfectants is to be avoided; (4) the minimum virus
from which the single harvest is derived comply with a test for concentration; (5) that the vaccine is not to be administered
identity and with the requirements for extraneous agents to pregnant women; (6) the time within which the vaccine
(2.7.3). must be used after reconstitution.
FINAL BULK VACCINE
Virus harvests that comply with the above tests are pooled
and clarified to remove cells. A suitable stabiliser may be added Viper Venom
and the pooled harvests diluted as appropriate.
Daboia Venom
Only a final bulk vaccine that complies with the following
Viper Venom is the dried secretion obtained from the poison
requirements may be used in the preparation of the final lot.
glands of Viperae russelli and other species of Viperae (Fam.
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk Viperidae).
for each sterility medium. Viper Venom contains not less than 50 Mouse Units per mg.
FINAL LOT
Production
The final bulk vaccine is distributed aseptically into sterile,
Immediately after extraction, the poisonous secretion is dried
tamper-proof containers and freeze-dried to a moisture content
from the frozen state. The dried venom is pooled, mixed,
shown to be favourable to the stability of the vaccine. The
dissolved in ice-cold water for injection and then filtered
containers are then closed so as to prevent contamination
through a bacteria-proof filter to give a stock solution. Further
and the introduction of moisture.
dilutions of the stock solution are made with water for
Only a final lot that is satisfactory with respect to each of the injection under aseptic conditions to give solutions with the
requirements given below under Identification, Tests and required number of Mouse Units per ml. These solutions are
Assay may be released for use. Provided that the test for then distributed in single dose sterile glass containers, dried
bovine serum albumin has been carried out with satisfactory from the frozen state and sealed at a pressure not exceeding
results on the final bulk vaccine, it may be omitted on the final 2.75 kPa.
lot.
Description. An almost white or very light yellow, dry powder
Identification which when mixed with water yields a clear solution with
some insoluble residue.
When the vaccine reconstituted as stated on the label is mixed
with specific Herpesvirus varicellae antibodies, it is no longer Identification
able to infect susceptible cell cultures.
A. Produces almost immediate coagulation of blood and
Tests citrated human plasma.
Sterility (2.2.11) Complies with the test for sterility. B. Mix the soluble fraction from at least 0.6 mg with 1 ml of
Abnormal toxicity (2.2.1). Complies with the test for abnormal polyvalent antisnake venom serum and incubate the mixture
toxicity. at 37o for 30 minutes. Inject 0.5 ml of the mixture intravenously
into a group of mice weighing between 18 and 20 g. Observe
Bovine serum albumin. Not more than 0.5 µg per human dose, the animals for 24 hours; no animal dies.
determined by a suitable immunochemical method (2.2.14).
Water (2.3.43). Not more than 3.0 per cent, determined by the Tests
semi-micro determination of water. Sterility (2.2.11). Complies with tests for sterility.
Assay Assay. Carry out the biological assay of snake venom
described below:
Titrate for infective virus, using at least ten cell cultures for
each fourfold dilution or by a technique of equal precision. Biological assay of snake venom
Use a suitable virus reference preparation to validate each
assay. The virus concentration is not less than the minimum Dissolve a quantity of the freeze-dried venom equivalent to
stated on the label. 50 Mouse Units in 25 ml of saline solution. Inject 0.5 ml

101
YELLOW FEVER VACCINE (LIVE) IP 2007

intravenously into each of 10 mice weighing between 18 and be only one passage from a master seed lot. A working seed
20 g and observe the animals for 24 hours. Not less than 3 and lot shall be used without intervening passage as the inoculum
not more than 8 of the mice die in 2 to 24 hours. If the number for infecting the tissues used in the production of a vaccine
of deaths is not within this range, change the dilution of the lot, so that no vaccine virus is more than one passage from a
venom suitably. seed lot that has passed all the safety tests.
Express the result in terms of number of Mouse Units per mg. Only a virus seed lot that complies with the following
requirements may be used for virus propagation.
NOTE — The quantity in mg of the venom which will kill in
2 to 24 hours not less than 3 and not more than 8 mice Identification
represents one Mouse Unit.
The master and working seed lots are identified as containing
Storage. Store in single dose, light-resistant containers. yellow fever virus by serum neutralisation in cell culture, using
Labelling. The label states (1) the number of Mouse Units per specific antibodies.
container; (2) the volume of water for injection to be used for Extraneous agents (2.7.3). Each working seed lot complies
reconstitution. with the test for extraneous agents.
PROPAGATION AND HARVEST

Yellow Fever Vaccine (Live) All processing of the fertilised eggs is done under aseptic
conditions in an area where no other infectious agents or cells
Yellow Fever Vaccine (Live) is a freeze-dried preparation of are handled at the same time. Two per cent but not less than
the 17D strain of yellow fever virus grown in fertilised hen twenty and not more than fifty eggs are set aside as uninfected
eggs. control eggs. After inoculation and incubation at a controlled
temperature, only living and typical chick embryos are
Production harvested. The age of the embryo at the time of virus harvest
General provisions is reckoned from the initial introduction of the egg into the
incubator and shall not be more than 12 days. After
The production of vaccine is based on a virus seed-lot system. homogenisation and clarification by centrifugation, the extract
The production method shall have been shown to yield of embryonic pulp is tested as described below and kept at
consistently the yellow fever vaccine (live) of acceptable -70° or colder until further processing. Virus harvests that
immunogenicity and safety for man. comply with the prescribed tests may be pooled. No human
The production method is validated to demonstrate that the protein is added to the virus suspension at any stage during
product, if tested, would comply with the test for safety and production. If stabilisers are added, they shall have been shown
efficacy. to have no antigenic or sensitising properties for man.

Reference preparation. In the test for neurotropism, a suitable Only a single harvest that complies with the following tests
batch of vaccine known to have satisfactory properties in may be used in the preparation of the final bulk vaccine.
man is used as the reference preparation.
Identification
Substrate for virus propagation
The single harvest contains virus that is identified as yellow
Virus for the preparation of master and working seed lots and fever virus by serum neutralisation in cell culture, using
for all vaccine batches is grown in the tissues of chick embryos specific antibodies.
from a flock free from specified pathogens (2.7.7).
Extraneous agents (2.7.3). Complies with the tests for
SEED LOT extraneous agents.
The 17D strain shall be identified by historical records that Control eggs. Complies with the tests for extraneous agents
include information on the origin of the strain and its (2.7.3).
subsequent manipulation. Virus seed lots are prepared in large Virus concentration. In order to calculate the dilution for
quantities and stored at a temperature below -60°. Master and formulation of the final bulk, each single harvest is titrated as
working seed lots shall not contain any human protein or described under Assay.
added serum.
FINAL BULK VACCINE
Unless otherwise justified and authorised, the virus in the
final vaccine shall be between passage levels 204 and 239 Single harvests that comply with the tests prescribed above
from the original isolate of strain 17D. A working seed lot shall are pooled and clarified again. A test for protein nitrogen

102
IP 2007 YELLOW FEVER VACCINE (LIVE)

content is carried out. A suitable stabiliser may be added and The virus concentration is not less than the equivalent in
the pooled harvests diluted as appropriate. TCID50 or PFU of 1 × 103 mouse LD50 per human dose. The
Only a final bulk vaccine that complies with the following relationship between mouse LD50 and TCID50 or PFU is
tests may be used in the preparation of the final lot. established by each laboratory and approved by the competent
authority.
Sterility (2.2.11). Carry out test for sterility using 10 ml of bulk
for each sterility medium. The method shown below, or another suitable technique, may
be used to determine the mouse LD50.
Protein nitrogen content (2.3.30). The protein nitrogen
content, before the addition of any stabiliser, is not more than Mouse LD50. The statistically calculated quantity of virus
0.25 mg per human dose. suspension that is expected to produce fatal specific
encephalitis in 50 per cent of mice of a highly susceptible
FINAL LOT strain, 4 to 6 weeks of age, after intracerebral inoculation.
The final bulk vaccine is distributed aseptically into sterile, Appropriate serial dilutions of the reconstituted vaccine are
tamper-proof containers and freeze-dried to a moisture content made in diluent for yellow fever virus (0.75 per cent solution
shown to be favourable to the stability of the vaccine. The of bovine albumin in phosphate-buffered saline pH 7.4, or
containers are then closed so as to prevent contamination any other diluent that has been shown to be equivalent for
and the introduction of moisture. maintaining the infectivity of the virus).
Only a final lot that is satisfactory with respect to thermal Mice of a highly susceptible strain, 4 to 6 weeks of age, are
stability and each of the tests given under Identification, Tests injected intracerebrally under anaesthesia with 0.03 ml of the
and Assay may be released for use. Provided that the test for vaccine dilution. Groups of not less than eight mice are used
ovalbumin has been performed with satisfactory results on for each dilution; the series of dilutions is chosen so as to
the final bulk vaccine, it may be omitted on the final lot. cover the range 0 to 100.0 per cent mortality of the mice.
Thermal stability. Maintain samples of the final lot of freeze- Injection of the mice is performed immediately after the
dried vaccine in the dry state at 37° for 14 days. Determine the dilutions have been made. The mice are observed for 21 days
virus concentration as described under Assay in parallel for and all deaths are recorded. Only survivors and deaths caused
the heated vaccine and for unheated vaccine. The difference by typical yellow fever infections are counted in the
in the virus concentration between unheated and heated computations. Mice paralysed on the twenty-first day of
vaccine does not exceed 1.0 log10, and the virus concentration observation are counted as survivors.
of the heated vaccine is not less than the number of TCID50 or Tests in monkeys for Yellow Fever Vaccine
plaque-forming units (PFU) equivalent to 1 × 103 mouse LD50
per human dose. Each master and working seed lot complies with the following
tests in monkeys for viraemia (viscerotropism),
Identification immunogenicity and neurotropism.
When the vaccine reconstituted as stated on the label is mixed The monkeys shall be Macaca spp. susceptible to yellow
with specific yellow fever virus antibodies, there is a significant fever virus and shall have been shown to be non-immune to
reduction in its ability to infect susceptible cell cultures. yellow fever at the time of injecting the seed virus. They shall
be healthy and shall not have received previously intracerebral
Tests
or intraspinal inoculation. Furthermore, they shall not have
Sterility (2.2.11). Complies with the test for sterility. been inoculated by other routes with neurotropic viruses or
Ovalbumin. Not more than 5 µg of ovalbumin per human dose, with antigens related to yellow fever virus. Not fewer than ten
determined by a suitable immunochemical method (2.2.14). monkeys are used for each test.
Abnormal toxicity (2.2.1). Complies with the test for abnormal Use a test dose of 0.25 ml containing the equivalent of not
toxicity. less than 5000 mouse LD50 and not more than 50,000 mouse
Bacterial endotoxins (2.2.3). Not more than 5 IU of bacterial LD50, determined by a titration for infectious virus and using
endotoxin per human dose. the established equivalence between virus concentration and
mouse LD50 (see under Assay). Inject the test dose into one
Water (2.3.43). Not more than 3.0 per cent, determined by the
frontal lobe of each monkey under anaesthesia and observe
semi-micro determination of water.
the monkeys for not less than 30 days.
Assay
Viraemia (Viscerotropism). Viscerotropism is indicated by the
Titrate for infective virus in cell cultures. Use an appropriate amount of virus present in serum. Take blood from each of the
virus reference preparation to validate each assay. test monkeys on the second, fourth and sixth days after

103
YELLOW FEVER VACCINE (LIVE) IP 2007

inoculation and prepare serum from each sample. Prepare 1:10, monkey in the test is given a score based on the scale:
1:100 and 1:1000 dilutions from each serum and inoculate each Grade 1 — rough coat, not eating,
dilution into a group of at least six cell culture vessels used for
the determination of the virus concentration. The seed lot Grade 2 — high-pitched voice, inactive, slow moving,
complies with the test if none of the sera contains more than Grade 3 — shaky, tremors, unco-ordinated, limb weakness,
the equivalent of 500 mouse LD50 in 0.03 ml and at most one
Grade 4 — inability to stand, limb paralysis or death (a dead
serum contains more than the equivalent of 100 mouse LD50 in
monkey receives a daily score of 4 from the day
0.03 ml.
of death until day 30).
Immunogenicity. Take blood from each monkey 30 days after
the injection of the test dose and prepare serum from each A clinical score for a particular monkey is the average of its
sample. The seed lot complies with the test if at least 90.0 per daily scores; the clinical score for the seed lot is the mean of
cent of the test monkeys are shown to be immune, as the individual monkey scores. The seed lot is not acceptable
determined by examining their sera in the test for neutralisation if the mean of the clinical severity scores for the group of
of yellow fever virus described below. monkeys inoculated with it is significantly greater (P = 0.95)
than the mean for the group of monkeys injected with the
It has been shown that a low dilution of serum (for example,
reference preparation. In addition, special consideration is
1:10) may contain non-specific inhibitors that influence this
given to any animal showing unusually severe signs when
test; such serum shall be treated to remove inhibitors. Mix
deciding on the acceptability of the seed lot.
dilutions of at least 1: 10, 1:40 and 1: 160 of serum from each
monkey with an equal volume of 17D vaccine virus at a dilution Histological evaluation. Five levels of the brain are examined
that will yield an optimum number of plaques with the titration including :
method used. Incubate the serum-virus mixtures in a water- Block 1 — the corpus striatum at the level of the optic
bath at 37° for 1 h and then cool in iced water; add 0.2 ml of chiasma,
each serum-virus mixture to each of four cell-culture plates
and proceed as for the determination of virus concentration. Block II — the thalamus at the level of the mamillary bodies,
Inoculate similarly ten plates with the same amount of virus Block III — the mesencephalon at the level of the superior
plus an equal volume of a 1:10 dilution of monkey serum known colliculi,
to contain no neutralising antibodies to yellow fever virus. At
Block IV — the pons and cerebellum at the level of the
the end of the observation period, compare the mean number
superior olives,
of plaques in the plates receiving virus plus non-immune serum
with the mean number of plaques in the plates receiving virus Block V — the medulla oblongata and cerebellum at the
plus dilutions of each monkey serum. Not more than 10 per level of the mid-inferior olivary nuclei.
cent of the test monkeys have serum that fails to reduce the Cervical and lumbar enlargements of the spinal cord are each
number of plaques by 50.0 per cent at the 1:10 dilution. divided equally into six blocks; 15 µm sections are cut from
Neurotropism. Neurotropism is assessed from clinical the tissue blocks embedded in paraffin wax and stained with
evidence of encephalitis, from incidence of clinical gallocyanin. Numerical scores are given to each hemisection
manifestations and by evaluation of histological lesions, in of the cord and to structures in each hemisection of the brain
comparison with ten monkeys injected with the reference as listed below. Lesions are scored as follows:
preparation. The seed lot is not acceptable if either the onset
Grade 1. Minimal: 1 to 3 small focal inflammatory infiltrates.
and duration of the febrile reaction or the clinical signs of
Degeneration or loss of a few neurons.
encephalitis and pathological findings are such as to indicate
a change in the properties of the virus. Grade 2. Moderate: 4 or more focal inflammatory infiltrates.
Clinical evaluation. The monkeys are examined daily for 30 Degeneration or loss of neurons affecting not more than one
days by personnel familiar with clinical signs of encephalitis third of cells.
in primates (if necessary, the monkeys are removed from their Grade 3. Severe: moderate focal or diffuse inflammatory
cage and examined for signs of motor weakness or spasticity). infiltration. Degeneration or loss of up to two third of the
The seed lot is not acceptable if in the monkeys injected with neurons.
it the incidence of severe signs of encephalitis, such as
Grade 4. Overwhelming: variable but often severe inflammatory
paralysis or inability to stand when stimulated, or mortality is
reaction. Degeneration or loss of more than 90.0 per cent of
greater than for the reference vaccine. These and other signs
neurons.
of encephalitis, such as paresis, in-coordination, lethargy,
tremors or spasticity are assigned numerical values for the It has been found that inoculation of yellow fever vaccine
severity of symptoms by a grading method. Each day each into the monkey brain causes histological lesions in different

104
IP 2007 YELLOW FEVER VACCINE (LIVE)

anatomical formations of the central nervous system with


varying frequency and severity (I. S. Levenbook et al., Journal
of Biological Standardization, 1987, 15, 305-313). Based on
these two indicators, the anatomical structures can be divided
into target, spared and discriminator areas. Target areas are
those which show more severe specific lesions in a majority
of monkeys irrespective of the degree of neurovirulence of
the seed lot. Spared areas are those which show only minimal
specific lesions and in a minority of monkeys. Discriminator
areas are those where there is a significant increase in the
frequency of more severe specific lesions with seed lots having
a higher degree of neurovirulence. Discriminator and target
areas for Macaca cynomolgus and Macaca rhesus monkeys
are shown in the table below:
Table 1. The discriminator and target areas for monkey.
Type of monkey Discriminator areas Target areas
Macaca cynomolgus globus pallidus substantia nigra
putamen anterior/
median thalamic
nucleus lateral
thalamic nucleus
Macaca rhesus caudate nucleus substantia nigra
globus pallidus cervical
putamen anterior/ lumbar
median thalamic enlargement
nucleus lateral
thalamic nucleus
cervical enlargement
lumbar enlargement
enlargement

Scores for discriminator and target areas are used for the final
evaluation of the seed lot. The individual monkey score is
calculated from the sum of individual target area scores in
each hemisection divided by the number of areas examined. A
separate score is calculated similarly for the discriminator areas.
Mean scores for the test group are calculated in two ways: (1)
by dividing the sum of the individual monkey discriminator
scores by the number of monkeys and (2) by dividing the sum
of the individual monkey target and discriminator scores by
the number of monkeys. These two mean scores are taken
into account when deciding on the acceptability of the seed
lot. The seed lot is not acceptable if either of the mean lesion
scores is significantly greater (P = 0.95) than for the reference
preparation.
Labelling. The label states (1) the strain of virus used in
preparation; (2) that the vaccine has been prepared in chick
embryos; (3) the minimum virus concentration; (4) that contact

105
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

HERBS AND HERBAL PRODUCTS

General Requirements .....................................................................................................


Monographs .....................................................................................................................
Acacia ..................................................................................................................
Amalaki .................................................................................................................
Amra ....................................................................................................................
Arachis Oil ............................................................................................................
Arjuna ....................................................................................................................
Artemisia ...............................................................................................................
Ashwagandha ........................................................................................................
Belladonna Dtry Extract .......................................................................................
Belladonna Leaves ..................................................................................................
Bhibhitaki ................................................................................................................
Bhringraj ................................................................................................................
Bhuiamla ................................................................................................................
Brahmi .................................................................................................................
Castor Oil ...............................................................................................................
Clove Oil ...............................................................................................................
Coleus ..................................................................................................................
Eucalyptus Oil ........................................................................................................
Garcinia .................................................................................................................
Gokhru .................................................................................................................
Guar Gum .............................................................................................................
Gudmar ................................................................................................................
Guduchi ...............................................................................................................
Guggul resin ........................................................................................................
Gugulipid ...............................................................................................................
Gugulipid Tablets ....................................................................................................
Haridra ..................................................................................................................
Haritaki .................................................................................................................
Hydrogenated Castor Oil .......................................................................................

1377
MONOGRAPHS INDIAN PHARMACOPOEIA 2007

Isapgol Husk .........................................................................................................


Kalmegh ................................................................................................................
Kunduru ................................................................................................................
Kutki ......................................................................................................................
Lausna ...................................................................................................................
Malt Extract ............................................................................................................
Mendukaparni ........................................................................................................
Manjistha ..............................................................................................................
Maricha ................................................................................................................
Mentha Oil .............................................................................................................
Opium ...................................................................................................................
Opium Powder .......................................................................................................
Papain ....................................................................................................................
Peppermint Oil ......................................................................................................
Pippali Large ..........................................................................................................
Pippali Small .........................................................................................................
Punarnava .........................................................................................................
Sarpagandha .........................................................................................................
Senna Pods .........................................................................................................
Senna Leaf .........................................................................................................
Shatavari ................................................................................................................
Shati ......................................................................................................................
Shellac ..................................................................................................................
Shunti .....................................................................................................................
Starch ....................................................................................................................
Tolu Blasam ...........................................................................................................
Tragacanth .........................................................................................................
Tulasi ...................................................................................................................
Vasaka ...............................................................................................................
Yasti ..................................................................................................................

1378
IP 2007 HERBS AND HERBAL PRODUCTS

Herbs and Herbal Products which may figure in a regulated list under appropriate forest
and other laws, may still be taken up for a monograph for
Herbs and products containing herb(s) have been in trade inclusion in pharmacopoeia, if there is knowledge of efforts
and commerce and are currently used for a variety of purposes. to cultivate or take care of sustainability issues and / or specific
As a country, India has a rich history of use of herbs, processed permission is available under law for use of the herb. As already
herbs and formulations containing herbs both from traditional specified under “General Notices” in the pharmacopeia,
wisdom as well as cultural usage. Herbs and herbals products appearance of a monograph does not mean its approval as a
are also regulated by various laws. For the purposes of drug under the law. Monograph of a herb in the pharmacopoeia
pharmacopoeial standards various considerations have been is to provide qualitative and quantitative standards of quality
given. This monograph provides a general outline and policies for the herb for its use either as a food item or food ingredient
towards the same. or food supplement/ nutraceuticals, as a drug, and / or as an
ingredient in cosmetics. Each such use would need to comply
Crude Herbs with applicable regulations. Each herb is regarded as one active
This term means, unless specified otherwise, mainly whole, substance, irrespective of the knowledge about the active
fragmented or cut, plants, parts of plants, algae, fungi, and constituents of the herb is available of not.
lichen in a form which is not processed. Herbs are usually in * Ayurvedic Pharmacopeia of India, Vol. 1-6, Ministry of Health and
dried form, but sometimes, when specified, may also be in a Family Welfare, Govt. of India.
fresh form. In specific cases exudates which have not been Herbs may be exposed to low dose gamma radiation for
processed further also are covered under the term herbs. purposes of reducing their microbial contamination. Herbs
Processing, does not include, normally expected value treated with low dose gamma radiation shall meet national
addition steps like grading, sizing, removal of weeds or parts laws related to such treatment and shall be labeled as per law.
of plants other than those specified herb and removal of
adulterants. The term herbs, though botanically generally refer Processed Herbs
to plants of specified height and nature, for the purposes of
pharmacopoeial reference, shall mean and include plants and Processed herbs means preparations obtained by subjecting
parts of plants not necessarily from herbs and shrubs, but herbs to treatment such as extraction, distillation, expression,
cover the entire range namely creepers, climbers, trees etc. fractionation, purification, concentration and partial or full
Each monograph of a herb in the pharmacopoeia shall specify fermentation. Processing may also be done by way of
the botanical scientific name according the binomial system powdering herbs, preparing tincture, preparing extract,
specifying the genus, species, variety and author. In cases distilling to get essential oils, fatty oils (either expressed or
where there are controversial botanical identity, as is seen solvent extracted or a blend of both) expressed juices, extracted
with mainly herbs known in the Indian traditional system, the exudates, gums and oleo resins, liquid extract where the
monograph shall specify the official name of the herb along solvent is evaporated to yield concentrated semi solid mass
with its botanical scientific name and guidance is taken from or dried mass. Extraction may be performed by means of
Ayurvedic Pharmacopoiea of India* to decide the same. In appropriate technology such as infusion, maceration,
cases where, the same herb is available in different grades or soxhleting, boiling under ambient or higher pressure, with or
sizes, if found appropriate and necessary, separate without specified enzymes, with or without agitation and
monographs may be introduced in the pharmacopoeia to cover combination thereof. Drying of liquid extracts for removal of
each of them with appropriate standards. For example- Pippali the solvent may be done by using various appropriate
(large) and Pippali (small). technologies like air drying, sun drying, drying under vacuum
or with forced air circulation, drying at low temperature with
While deciding to introduce a monograph for a herb in the
air circulation, by way of lyophilization or freeze drying.
pharmacopeia, the criteria that would be kept in mind, but not
Extracts of herbs may also be prepared by using carbon di-
limited to are – herbs with specific name and a definitive
oxide as a solvent-super critical fluid extraction.
botanical identity up to species, availability and usage in trade
and commerce, regulatory interest, knowledge of and Extracts may be liquid extracts and tinctures, semi solid (soft
availability of a specific chemical compound of well extracts) or solid dry extracts of known consistency obtained
characterised structure [either responsible for the biological from herbs. Standardized extract, a term commonly employed,
activity of the herb (bio-marker) or a chemical compound would for pharmacopoeial purposes, mean an extract adjusted
known to be present in the herb even if not responsible for with in an acceptable tolerance to a given content of bio-
biological activity(chemical/analytical marker)], availability of marker or chemical/ analytical marker. Standardization may be
a quantitative method for estimation of such a compound, achieved by adjusting the extracts with approved inert material
knowledge of safety of the herb, and its sustainability. Herbs or by blending one or more batches of extracts. Wherever

1379
HERBS AND HERBAL PRODUCTS IP 2007

possible, extracts shall specify the defined range of the Herbs may also be extracted using vegetable oils (approved
constituents (bio-marker or chemical/ analytical marker). by Food Law) for extraction purposes and such extracts shall
Extracts not covered in the above description would be defined specify the oil used for processing.
by the process of production of the herb to the extract, solvent Approved preservatives or preservatives system may be used
used and technology applied. The difference between extracts during preparation of extracts. The names of such
and tinctures would be, in the type of solvent used for preservatives used which would remain in the final extract
extracting a herb, and tincture would normally mean an extract shall be listed on the label of such extract, and the proportion
where aqueous-ethanol is used as a solvent for extraction. of preservatives used shall not exceed normally accepted safe
Dry extracts usually have a loss on drying or water content limits of their usage as per relevant laws or pharmacopoeial
not greater than 5 per cent w/w, unless specified otherwise in standards. No artificial colours may be used in extracts of
any monograph. It is normal to extrapolate safety aspects and herbs.
history of use information for extracts as long as the process,
solvents, extraction ratios are comparable to the processes Extracts may be exposed to ethylene oxide fumigation or low
dose gamma radiation for purposes of reducing their microbial
used in documented traditional knowledge. All extracts should
contamination. In cases where they are fumigated, the final
specify the extractive ratios for example 15: 1 meaning 15 parts
extracts exposed shall meet residual levels of ethylene oxide
of the herb provides one part of the extracts, either as w/w or
limits as applicable. Herbs treated with low dose gamma
w/v as the case may be. Additionally in cases of standardized
radiation shall meet national laws related to such treatment
extracts the inert excipients(s), if any used for standardization
and shall be labelled as per law.
or adjustment of the content of constituents should also be
declared on the label of such extracts. Extracts shall be free Appearance of a monograph of an extract in the pharmacopeia
from solvent used for extraction and shall comply with a limit does not mean its approval as a drug under the law. Monograph
of not more than 10 ppm incases where the solvent is either of an extract in the pharmacopoeia is to provide qualitative
acetone, iso-propyl alcohol, or methanol and not more than and quantitative standards of quality for the extract for its use
100 ppm of hexane, if hexane is the solvent used, in the final either as a food item or food ingredient or food supplement/
extract. Harmful and carcinogenic solvents shall not be used nutraceuticals, as a drug and / or as an ingredient in cosmetics.
for extraction purposes. Solvents and solvent systems may Each such use would need to comply with applicable
include use of propylene glycol, glycerin, sorbitol and such regulations. Each extract is regarded as one active substance
irrespective of the knowledge about the active constituents
other polyhydroxy alcohols, as long as the content of such
of the herb is available or not.
polyhydroxy alcohol are within safe limit in the final product.
In cases where extraction and fractionation process leads to Herbal Formulations
preparation of an extract, which consists of a single chemical Herbal formulation shall mean a dosage form consisting of
compound of more than of at least 70 per cent purity, such one or more herbs or processed herb(s) in specified quantities
extracts shall be treated as an active pharmaceutical ingredient to provide specific nutritional, cosmetic benefits, and/or other
or a food additive or a cosmetic ingredient and would be benefits meant for use to diagnose treat, mitigate diseases of
required to meet relevant laws. human beings or animals and/or to alter the structure or
Extracts may also be offered as purified or enriched extracts. physiology of human beings or animals. Dosage forms
Such extract of a herb is processed in such a way to provide commonly employed for food or cosmetic or pharmaceuticals
higher than normal proportion of the active constituent (s) of may be employed to formulate one or more herb or processed
the herb as long as the active constituent (s) is/are are known. herbs. Dosage forms known in traditional medicines may also
Such purified or enriched extracts may contain additional be adopted for preparing herbal formulations, either for external
valuable components which may provide specific properties use or for internal administration. Adequate consideration for
like enhanced efficacy or stability or solubility and availability uniform distribution of herb or processed herbs as well as
of the active constituent (s). Purified and enriched extracts stability of the same in the dosage form shall be provided
may also be prepared to reduce or remove other specific during formulation development.
compound or group of compounds that is scientifically Herbal formulation shall be labeled to comply with relevant
considered undesirable in the herb extracts. Pharmacological, labelling requirements under food or drug or cosmetics laws
toxicological, pharmaceutical considerations need to be as applicable. Additionally, adequate information shall be
applied while preparing such purified or enriched extracts. provided on label of such formulations to include the name of
Mixed extracts may also be offered which would cover the herb, parts used, nature and type of extract or processed
combination of more than one herb extract for purposes of herb used, extraction ratios, quantity per unit dose or per
providing simplification or economical way to manufacture serving, name (s) of inert excipients used and any preservatives
herbal formulations. added shall be provided on the label.

1380
IP 2007 ACACIA

Appearance of a monograph of a herbal formulation in the addition of 0.5 ml of acetic acid, gives a white precipitate.
pharmacopoeia does not mean its approval as a drug under Filter and add to the clear filtrate 50 ml of ammonium oxalate
the law. Monograph of a herbal formulation in the solution; the filtrate becomes cloudy.
pharmacopoeia is to provide qualitative and quantitative
C. A 10 per cent w/v solution is either dextro-rotatory or slightly
standards of quality for the formulation for its use either as a
laevo-rotatory.
food item or food ingredient or food supplement/
nutraceuticals, as a drug and / or as a cosmetic. Each such use Tests
would need to comply with applicable regulations.
Sterculia gum and agar. To 50 mg of the powdered substance
under examination add 0.2 ml of freshly prepared ruthenium
red solution and examine microscopically; the particles do
not acquire a red colour after irrigation with water.
Acacia Agar and tragacanth. To 10 ml of a 10 per cent w/v solution
Gum Acacia; Indian Gum add 0.2 ml of lead acetate solution; no precipitate is produced.
Starch and dextrin. Boil 10 ml of a 10 per cent w/v solution
and cool, add 0.1 ml of 0.05 M iodine; no blue or brown colour
is produced.
Tannins. To 10 ml of a 10 per cent w/v solution add 0.1 ml of
ferric chloride test solution; a gelatinous precipitate is formed,
but neither the precipitate nor the liquid shows a dark blue
colour.
Sucrose and fructose. To 1 ml of a 10 per cent w/v solution
add 4 ml of water, 0.1 g of resorcinol and 2 ml of hydrochloric
acid and heat on a water-bath; no yellow or pink colour
develops.
Water-insoluble matter. Dissolve 5 g, in fine powder, in 100
ml of water in a 250-ml flask, add 10 ml of dilute hydrochloric
acid and boil gently for 15 minutes. Filter by suction while hot
Acacia is the air-hardened, gummy exudation from the stem through a sintered-glass crucible, previously tared, wash
and branches of Acacia nilotica (Linn.) Del. subsp. indica thoroughly with hot water, dry at 105º and weigh; the residue
(Benth.) Brenan (syn. A. arabica Willd. var. indica Benth.) does not exceed 50 mg.
(Fam. Leguminosae), or other species of Acacia. It is available Microbial contamination (2.2.9). 1.0 g is free from Escherichia
as pieces (tears) or in the form of a powder. coli.

Description Sulphated ash (2.3.18). Not more than 5.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 1.0 per cent,
Tears — Irregular and broken pieces of varying size, yellowish-
determined on 1.0 g by Method C.
white, yellow or amber in colour, with numerous minute
fissures; brittle fractured surface, glassy and occasionally Loss on drying (2.4.19). Not more than 15.0 per cent, determined
iridescent; odourless. on 1.0 g by drying in an oven at 105º.
Powder — A white or yellowish-white powder; odourless; on Storage. Store protected from heat, moisture and against
treatment with water it dissolves to give a mucilaginous liquid attack by insects and rodents.
which is colourless or yellowish, dense, viscous, adhesive
and translucent.

Identification
A. An aqueous solution is gelatinised by the addition of lead
subacetate sol3either dextro-rotatory or slightly laevo-rotatory.
B. To 5 ml of a 10 per cent w/v solution add gradually, while
shaking, 10 ml of ethanol (95 per cent). The cloudy liquid, on

1381
AMALAKI IP 2007

Amalaki and filter. Reflux the residue further for two times with 75 ml of
methanol, cool and filter. Combine all the filtrates and
Emblic Myrobalan; Indian Gooseberry concentrate under vacuum to 10 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with anisaldehyde sulphuric acid reagent. Heat at 100º
for 5-10 minutes and examine the plate in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 3 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 30 per cent.
Water-soluble extractive (2.6.3). Not less than 40 per cent by
Method 1.
Amalaki consists of the dried fruit pericarp of Emblica Ash (2.3.19). Not more than 5.0 per cent.
officinalis Gaertn. (Phyllanthus emblica Linn.) (Fam. Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Euphorbiaceae).
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Amalaki contains not less than 1.0 per cent w/w gallic acid heavy metals, Method B ( 20 ppm).
calculated on the dried basis.
Loss on drying (2.4.19). Not more than 12.0 per cent,
Description. The dried fruit has a highly shriveled and wrinkled determined on 5 g by drying in an oven at 105º.
external surface. The taste is sour and astringent followed by
Microbial contamination (2.2.9). Complies with the microbial
delicately sweet tinge.
contamination tests.
Identification Assay. Determine by liquid chromatography (2.4.14).
A. Macroscopic — The dried fruit shows a broad, highly Test solution. Weigh accurately about 0.5 g of coarsely
shriveled and wrinkled external convex surface, lateral surface powdered substance under examination, add 50 ml of water,
transversely wrinkled, external surface exhibits few whitish sonicate for 3 minutes and heat on a boiling water bath for
specks, occasionally some pieces show a portion of stony 15 minutes, cool and dilute to 100.0 ml with water and filter.
testa. Reference solution. A 0.01 per cent w/v solution of gallic
B. Microscopic — The epicarpic cells are rectangular in shape acid RS in water.
and their walls are highly cuticularized. Anomocytic type of Chromatographic system
stomata is found rarely. Collateral fibrovascular bundles are – a stainless steel column 25 cm x 4.6 mm packed with
scattered throughout the inner mesocarp. Pitted and helical octadecylsilane bonded to porous silica (5 µm),
tracheids with tapering ends are seen. At places in the phloem, – mobile phase: a gradient mixtures of acetonitrile and a
large cavities filled with crystal mass are present. buffer solution prepared by dissolving 0.136 g of
C. Determine by thin-layer chromatography (2.4.17), coating potassium di-hydrogen orthophosphate in 500 ml of
the plate with silica gel. water, add 0.5 ml of orthophosphoric acid and make
upto 1000 ml with water,
Mobile phase. A mixture of 20 volumes of toluene, 45 volumes
– flow rate. 1.5 ml per minute,
of ethyl acetate, 20 volumes of glacial acetic acid and 5
– spectrophotometer set at 270 nm,
volumes of formic acid.
– a 20 µl loop injector.
Test solution. Reflux 2 g of the coarsely powdered substance
Time Buffer solution Acetonitrile
under examination with 50-75 ml of methanol for 15 minutes,
(min) (per cent v/v) (per cent v/v)
cool and filter. Reflux the residue further for two times with
75 ml of methanol, cool and filter. Combine all the filtrates and 0 100 0
concentrate under vacuum to 50 ml. 18 55 45
Reference solution. Reflux 0.4 g of the coarsely powdered 25 20 80
amalaki RS with 50-75 ml of methanol for 15 minutes, cool 30 100 0

1382
IP 2007 AMRA

Inject the reference solution. The relative standard deviation Test solution. To 5 g of the coarsely powdered substance
for the replicate injections is not more than 2.0 per cent. under examination, add 50 ml of methanol and reflux for
Inject the test solution and reference solution. 15 minutes, cool and filter. Reflux the residue further for three
times with 50 ml of methanol, cool and filter. Combine all the
Calculate the content of gallic acid. filtrates and concentrate under vacuum to 10 ml.
Storage. Store protected from light, heat, moisture and against Reference solution. Weigh about 2 g of amra RS, add 50 ml of
attack by insects and rodents. methanol and reflux for 15 minutes, cool and filter. Reflux the
residue further three times with 50 ml of methanol, cool and
filter. Combine all the filtrates and concentrate under vacuum
to 4 ml.
Amra
Apply to the plate 10 µl of each solution as bands 10 mm by
Mango; Mangifera 2 mm. Allow the mobile phase to rise 8 cms. Dry the plate in air,
spray with vanilin sulphuric acid reagent. Heat at 100º for
5-10 minutes and examine the plate in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 10.0 per cent.
Water-soluble extractive (2.6.3). Not less than 10.0 per cent
by method I.
Ash (2.3.19). Not more than 16.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Amra consists of dried stem bark of Mangifera indica L.(Fam. heavy metals, Method B (20 ppm).
Anacardiaceae).
Loss on drying (2.4.19). Not more than 12.0 per cent,
Amra contains not less than 1.5 per cent of mangiferin determined on 5 g by drying in an oven at 1050.
calculated on the dried basis.
Microbial contamination (2.2.9). Complies with the microbial
Description. The dried stem bark occurs in pieces of variable contamination tests.
size and thickness, surface rough. Odour pleasant and taste
Assay. Determine by liquid chromatography (2.4.14).
astringent.
Test solution. Weigh 2 g of coarsely powdered substance
Identification under examination, add 10 ml of dimethylformamide, sonicate
for 5 minutes and add 50 ml of methanol and boil on a water
A. Macroscopic — The surface is rough due to longitudinal
bath for 10 minutes, cool and dilute to 100.0 ml with methanol
cracks, fissures and scattered, raised lenticels, greyish to dark
and filter. Dilute 5.0 ml of this solution to 50.0 ml in methanol.
brown externally and yellowish-white to reddish internally.
Reference solution. Dissolve 10 mg of mangiferin RS in 10 ml
B. Microscopic — The mature bark shows a wide cork which of dimethylformamide and dilute to 100.0 ml with methanol.
has tangentially elongated cells a few outer layers are brown
and inner lighter in colour, resin canals and yellow coloured Chromatographic system
tannin sacs are found in the phloem region, stone cells are – a stainless steel column 25 cm x 4.6 mm packed with
thick walled and lignified, prismatic calcium oxalate crystals octadecylsilane bonded to porous silica (5 µm),
are present. – mobile phase: filtered and degassed mixture of 15
volumes of acetonitrile and 85 volumes of buffer pH
C. Determine by thin-layer chromatography (2.4.17), coating 2.8 prepared by dissolving 1.36 g of potassium di-
the plate with silica gel. hydrogen orthophosphate in 950 ml of water, adjust
Mobile phase. A mixture of 100 volumes of ethyl acetate, the pH 2.8 with orthophosphoric acid and make upto
11 volumes of formic acid, 11 volumes of acetic acid and | 1000 ml,
25 volumes of water. – flow rate. 1 ml per minute,

1383
IP 2007
ARACHIS OIL

– spectrophotometer set at 366 nm, Tests


– a 20 µl loop injector.
Weight per ml (2.4.29). 0.908 g to 0.920 g.
Inject the reference solution. The relative standard deviation
for the replicate injections is not more than 2.0 per cent. Refractive index (2.4.27). 1.467 to 1.470.
Inject the test solution and reference solution. Alkaline impurities. Mix 10 ml of recently distilled acetone
and 0.3 ml of water in a test-tube, add 0.05 ml of a 0.04 per cent
Calculate the content of mangiferin.
w/v solution of bromophenol blue in ethanol (95 per cent)
Storage. Store protected from moisture and against attack by and neutralise the solution, if necessary, with 0.01 M
insects and rodents. hydrochloric acid or 0.01 M sodium hydroxide. Add 10 ml of
the substance under examination, shake and allow to stand.
Not more than 0.1 ml of 0.01 M hydrochloric acid is required
Arachis Oil to change the colour of the upper layer to yellow.
Groundnut Oil; Peanut Oil Semi-drying oils. Boil 1 g in a flask under a reflux condenser
for 5 minutes with 5 ml of a mixture of 3 volumes of 2 M
Arachis Oil is the refined fixed oil obtained from the seed
ethanolic potassium hydroxide and 1 volume of ethanol
kernels of one or more of the cultivated varieties of Arachis
(95 per cent), add 1.5 ml of 6 M acetic acid and 50 ml of
hypogaea Linn. (Fam. Leguminosae) and may contain suitable
ethanol (70 per cent), warm until the solution is clear. Cool
antioxidants as stabilisers.
slowly with a thermometer in the liquid; the temperature at
Description. A clear, colourless or pale yellow oily liquid; which the solution becomes turbid is not lower than 36º.
odour, faint and nutlike.
Peroxide value (2.3.35). Not more than 5.0.
Identification Acid value (2.3.23). Not more than 0.5.
Determine by the thin layer chromatography (2.4.17), coating Iodine value (2.3.28). 85 to 105.
the plate with kieselguhr G. Saponification value (2.3.37). 185 to 196.
Mobile phase. Glacial acetic acid. Rancidity. Shake 1 ml of a 10 per cent v/v solution in ether
Test solution. Dissolve 20 mg (one drop) of the substance with 1 ml of hydrochloric acid, add 1 ml of a 0.1 per cent w/v
under examination in 4 ml of chloroform. solution of phloroglucinol in ether; no red or pink colour is
produced.
Reference solution (a). Dissolve 20 mg (one drop) of arachis
oil in 4 ml of chloroform. Unsaponifiable matter (2.3.39). Not more than 1.0 per cent.
Reference solution (b). Dissolve 20 mg (one drop) of Cottonseed oil. In the Identification test, the chromatogram
cottonseed oil in 4 ml of chloroform. obtained with the test solution does not correspond to that
obtained with reference solution (b)
Reference solution (c). Dissolve 20 mg (one drop) of sesame
oil in 4 ml of chloroform. Sesame oil. In the Identification test, the chromatogram
obtained with the test solution does not correspond to that
Impregnate the plate by placing it to a depth of about 5 mm in
obtained with reference solution (c).
a tank containing a shallow layer of a mixture of 95 volumes of
light petroleum (60º to 80º) and 5 volumes of liquid paraffin, Heavy metals (2.3.13). 2.0 g complies with the limit test for
allowing the solvent to rise at least 14 cm, removing the plate heavy metals, Method B (10 ppm).
and allowing it to dry in air for 5 minutes; use with the flow of Arachis Oil intended for use in the manufacture of parenteral
the mobile phase in the direction in which impregnation was preparations complies with the following additional
carried out. requirement.
Apply to the plate 2 µl of each solution. Allow the mobile Water (2.3.43). Not more than 0.3 per cent, determined on 3.0 g.
phase to rise 12 cm. Dry the plate at 110º for 10 minutes, allow
Storage. Store protected from light and moisture, in a well-
to cool and expose it to iodine vapour in a saturated chamber.
filled container. Arachis Oil intended for use in the manufacture
Remove the plate and allow it to stand for a few minutes until
of parenteral preparations should be stored in a glass
the brown background colour has disappeared. Spray the plate
container.
with starch solution; blue spots appear which become brown
on drying and become blue again on spraying with water. The Labelling. The label states (1) whether the contents are
spots in the chromatogram obtained with the test solution suitable for use in the manufacture of parenteral preparations;
correspond to those in the chromatogram obtained with (2) when the addition of antioxidants is authorised, the name
reference solution (a). and quantity of the added antioxidants.

1384
IP 2007
ARJUNA

Arjuna Apply to the plate 10 µl of each solution as bands 10 mm by


2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
Terminalia Bark spray with 10 per cent w/v solution of sulphuric acid in
methanol and heat at 1100 for 5 minutes. The chromatographic
profile of the test solution is similar to that of the reference
solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 20.0 per cent.
Water-soluble extractive (2.6.3). Not less than 20 per cent by
method I.
Ash (2.3.19). Not more than 30.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 2 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Arjuna consists of dried stem bark of Terminalia arjuna (Roxb) Loss on drying (2.4.19). Not more than 10.0 per cent,
Wight & Arn (Fam. Combretaceae) determined on 5 g by drying in an oven at 1050.
Arjuna contains not less than 0.02 per cent of arjungenin Microbial contamination (2.2.9). Complies with the microbial
calculated on the dried basis. contamination tests.
Description. A flat or minutely curved thick pieces of bark Assay. Determine by liquid chromatography (2.4.14).
with reddish gray colour and astringent taste.
Test solution. Reflux 5 g of coarsely powdered substance under
Identification examination with 50 ml of chloroform for 15 minutes, cool and
filter. Reflux the residue further with 50 ml of chloroform, cool
A. Macroscopic — Stem bark pieces, flat or minutely curved, and filter. Combine the filtrates and concentrate under vacuum
with reddish gray external surface and darker inner surface. to dryness, then extract dried residue with 10 ml of ethanol at
Internal surface has longitudinal minute ridges. Fractures 50° for 10 minutes and filter.
longitudinal.
Reference solution. A 0.1 per cent w/v solution of arjungenin
B. Microscopic — Cork consisting of 6-10 layers of elongated RS in ethanol.
cells, phloem broad, medullary rays uniseriate. Calcium oxalate
clusters abundant. Few of the parenchyma cells contain Chromatographic system
colouring matter. – a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (10 µm),
C. Determine by thin-layer chromatography (2.4.17), coating – mobile phase (A). acetonitrile (70 per cent) in water,
the plate with silica gel G. (B). acetonitrile (30 per cent) in water,
Mobile phase. A mixture of 5 volumes of toluene, 5 volumes – flow rate. 1.2 ml per minute,
of ethyl acetate and 0.5 volume of acetic acid. – spectrophotometer set at 210 nm,
– a 20 µl loop injector.
Test solution. Reflux 2 g of coarsely powdered substance under
examination with 50 ml of chloroform for 15 minutes, cool and Time Mobile phase A Mobile phase B
filter. Reflux the residue further with 50 ml of chloroform. (min) (per cent v/v) (per cent v/v)
Combine the filtrate and concentrate under vacuum to dryness. 0 30 70
Dissolve the residue in 10 ml of ethanol at 50° for 10 minutes 10 50 50
and filter. 30 70 30
Reference solution. Reflux 1 g of arjuna RS with 50 ml of 50 30 70
chloroform for 15 minutes, cool and filter. Reflux the residue Inject the reference solution. The test is not valid unless the
further with 50 ml of chloroform. Combine the filtrate and relative standard deviation for the replicate injection for the
concentrate under vacuum to dryness. Dissolve the residue analyte peak corresponding to arjungenin is not more than 2.0
in 5 ml of ethanol at 50° for 10 minutes and filter. per cent.

1385
ARTEMISIA IP 2007

Inject the test solution and reference solution. because florets / achenes come out of it. Receptacle globular
Calculate the contents of arjungenin. to oblong. Achenes minute; possess striated surface,
yellowish brown in colour, 0.7-1.0 mm long, and oval to elliptic
Storage. Store protected from light, heat, moisture and against in shape. Stomatal index: 8-10-12.
attack by insects and rodents.
B. The powder of the herb is grayish green to greenish yellow.
Examined under a microscope using chloral hydrate solution.
The powder shows the following diagnostic characteristics:
Artemisia T-shaped and globular trichomes, epidermal cells with wavy
walls, anomocytic to anisocytic stomata, minute druse crystals,
tricolpate rounded pollen grains, stone cells, annular vessels,
rod shaped palisade cells and stigma with small and club
shaped papillae.
C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Mobile phase. A mixture of 1 volume of hexane and 1 volume
of diethyl ether.
Test solution. Boil 0.1 g of the coarsely powdered substance
under examination with 10 ml of hexane for 10 minutes and
filter. Evaporate the filterate to 1 ml.
Reference solution. A 0.1 per cent w/v solution of
artemisia RS in hexane.

Artemisia consists of dried leaves or the dried leaf and Apply to the plate 5 µl of each solution. Allow the mobile
flowering tops of Artemisia annua L. (Fam. Asteraceae), phase to rise 5 cm. Dry the plate in air and spray with a mixture
known as Qinghao. of 50 volumes of glacial acetic acid, 1 volume of sulphuric
acid and 0.5 volume of anisaldehyde. Heat the plate at 100º
Artemisia contains not less than 0.8 per cent of artemisinin, for 15 minutes. The chromatogram having pink colour spot
calculated on the dried basis. obtained with the test solution corresponds to the reference
Description. Slightly camphoraceous odour and bitter taste. solution.

Identification Tests
A. The leaves grayish green, slightly darker upper surface, Foreign organic matter (2.6.1). Not more than 2.0 per cent.
glabrous to sparsely hairy, break easily into small fragments,
Ash (2.3.19). Not more than 11.0 per cent.
3-pinnatipartite, petiolate and much variable in size (2.5-
10.0 cm long). Leaf lobes narrow, oblong to elliptical with Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
acuminate tip, about 1.0 mm wide. Petioles up to 1.0 cm long, Assay. Determine by high performance thin-layer
base amplexicaul. Inflorescence panicled raceme. Dry capitula chromatography (2.4.17), coating the plate with silica gel
yellowish brown, pedicellate, heterogamous globose to sub- GF254.
globose or disc shaped, 2.0 mm in diameter, flower heads
arranged in lax or drooping, involucral bracts 3- seriate, Mobile phase. A mixture of 1 volume of hexane and 1 volume
greenish yellow, glabrous and oblong in shape, measure of diethyl ether.
1.0-1.2 x 0.5 mm in size. Inner involucral bracts elliptic having Test solution. To 0.1 g of the coarsely powdered substance
a median greenish streak on its outer surface. Ray florets under examination, add 10 ml of hexane and keep for 12 hours,
pistillate, 6-8 in number per capitulum and 1.0-1.2 mm long. filter. Repeat the process of extraction 3 times. Combine the
Disc florets hermaphrodite, 20-36 florets per capitulum and extracts, evaporate and dissolve in 1.0 ml of hexane.
0.8-1.0 mm long. All florets possess capitate oil glands on the
Reference solution. A 0.1 per cent w/v solution of artemisinin
middle of the outer surface that are 54-83 µm in diameter.
RS in hexane.
Stamens 0.7 mm long attached to the corolla base, anther
appendages lanceolate to triangular with acuminate tip. Anthers Apply to the plate 5 µl of each solution as bands 10 mm by
oblong and introrse. Pollen grains tricolpate, rounded 21-33 2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
µm in diameter. In dry condition, capitula become empty and spray with a mixture of 50 volumes of glacial acetic acid,

1386
IP 2007 ASHWAGANDHA

1 volume of sulphuric acid and 0.5 volume of anisaldehyde. Reference solution. Reflux 0.6 g of coarsely powdered
Heat the plate at 100º for 15 minutes, scan the plate in ashwagandha RS with 10 ml methanol for 15 minutes, cool
absorbance mode at 540 nm. Record the chromatograms and and filter.
measure the responses for the analyte peak.
Apply to the plate 10 µl of each solution as bands 10 mm by
Calculate the content of artemisinin. 2 mm. Allow the mobile phase to rise 8 cms above the line of
Storage. Store protected from light and moisture and against application. Dry the plate in air, spray with solution of
attack by insects and rodents. anisaldehyde reagent. Heat at 100º for 5-10 minutes and
examine the plate in day light. The chromatographic profile of
the test solution is similar to that of the reference solution.

Tests
Ashwagandha
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Indian Ginseng; Withania
Ethanol-soluble extractive (2.6.2). Not less than 10.0 per cent.
Water-soluble extractive (2.6.3). Not less than 15 per cent by
Method I.
Ash (2.3.19). Not more than 7.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 1.2 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Ashwagandha consists of the dried mature roots of Withania
somnifera Dunal (Fam. Solanaceae). Test solution. Reflux about 5 g of the coarsely powdered
substance under examination with 50 ml of methanol on a
Ashwagandha contains not less than 0.02 per cent of total
water bath for 15 minutes, cool and filter. Reflux the residue
withanolide A and withaferin A, calculated on the dried basis.
further with methanol till the last extract turns colorless, cool
Description. Buff to greyish-yellow roots. Taste, slightly and filter. Combine all the filtrates and concentrate to 50.0 ml.
mucilaginous, bitter and acrid.
Reference solution (a). A solution containing 0.02 per cent
Identification w/v each of withanolide A RS and withaferin A RS in
methanol, prepared by heating gently on a water bath.
A. Macroscopic — Primary roots are straight, conical or finger
Reference solution (b). A 0.02 per cent w/v solution of
like in shape, variable in thickness with the age. Secondary
withanolide A RS in methanol, prepared by heating gently on
roots are thin and fibrous. Surface buff to greyish-yellow
a water bath, for peak identification.
with longitudinal wrinkles.
B. Microscopic — Vessels with bordered pits and horizontal Chromatographic system
perforations. Fibres aseptate with pointed ends. Wood – a stainless steel column 25 cm x 4.6 mm packed with
elements lignified. Starch grains abundant, simple, mostly octadecylsilane bonded to porous silica (10 µm),
spherical, reniform – oval with central hilum. Microcrystals in – mobile phase: a gradient mixtures of acetonitrile and a
parenchyma cells. buffer solution prepared by dissolving 1.36 g of
potassium dihydrogen phosphate in 900 ml water, adjust
Mobile phase. A mixture of 9 volumes of chloroform and pH to 2.8 with dilute phosphoric acid and diluting it to
1 volume of methanol. 1000 ml with water,
Test solution. Reflux 3 g of coarsely powdered substance under – flow rate. 1.5 ml per minute,
examination with 50 ml methanol for 15 minutes, cool and – spectrophotometer set at 227nm,
filter. – a 20 µl loop injector.

1387
BELLADONNA DRY EXTRACT IP 2007

Time Buffer solution Acetonitrile acid and discard the chloroform. Combine the acid solutions,
(min) (per cent v/v) ( per cent v/v) neutralise with dilute ammonia solution, add 5 ml in excess
0 90 10 and shake with successive quantities, each of 25 ml, of
chloroform until complete extraction of the alkaloids is effected,
18 55 45
washing each chloroform solution with the same 10 ml of water
25 20 80 and filtering into a flask through a plug of cotton wool
27 90 10 previously moistened with chloroform. Distil most of the
37 90 10 chloroform from the combined extracts and transfer the
remainder of the chloroform to a shallow open dish. Evaporate
Inject the reference solution (b) and determine the retention
the remainder of the chloroform without the aid of a current of
time of withanolide A. Inject the reference solution (a). Identify
air, heat the residue in an oven at 100o for 15 minutes, dissolve
the retention time of withaferin A. The test is not valid unless
in a little chloroform, evaporate to dryness without the aid of
the relative standard deviation for the replicate injections for
a current of air and again heat in an oven at 100o for 15 minutes.
both the analyte peaks corresponding to withanolide A and
Dissolve the residue in 2 ml of chloroform, add 5.0 ml of
withaferin A is not more than 2.0 per cent and resolution is not
0.025 M sulphuric acid, warm to remove the chloroform, cool
less than 2.0.
and titrate the excess of acid with 0.05 M sodium hydroxide
Inject the test solution. using methyl red solution as indicator.
Calculate the contents of withanolide A and withaferin A. 1 ml of 0.025 M sulphuric acid is equivalent to 0.01447 g of
Storage. Store protected from heat, moisture and against attack alkaloids, calculated as hyoscyamine.
by insects and rodents. Storage. Store in small, wide-mouthed, tightly-closed
containers in a cool place.

Belladonna Dry Extract


Belladonna Dry Extract is a dried and powdered ethanolic
Belladonna Leaf
extract of Belladonna Herb. Belladonna Leaf consists of the dried leaf and flowering tops
Belladonna Dry Extract contains not less than 0.95 per cent of Atropa belladonna Linn. or of A. acuminata Royle ex
and not more than 1.05 per cent w/w of alkaloids, calculated as Lindley (Fam. Solanaceae)‘ or a mixture of both species.
hyoscyamine. Belladonna Herb contains not less than 0.30 per cent of total
alkaloids, calculated as hyoscyamine with reference to the
Tests material dried at 100o to 105o.
Loss on drying (2.4.19). Not more than 5.0 per cent, determined Description. Green to greenish-brown leaves, slightly darker
on 0.5 g by drying in an oven at 105o. on the upper surface, often crumpled and rolled and partly
Assay. Weigh accurately about 3 g and wash into a separating matted together in the drug. When whole, the lamina is 5 to
funnel with 12 ml of a mixture of equal volumes of ethanol 25 cm long and 3 to 12 cm wide, elliptical to ovate; acuminate
(95 per cent) and water, shake-well and frequently for at the apex, narrowing at the base; margin entire. Petiole 0.5 to
30 minutes, add 2 ml of dilute ammonia solution and 25 ml of 4 cm in length. The young leaves are highly pubescent, the
chloroform. Shake well, allow to separate and filter the older leaves are slightly pubescent along the veins. In the
chloroform layer into a second separating funnel through a flowering tops, the stems are hollow and flattened, with leaves
plug of absorbent cotton moistened with chloroform. Continue in pairs of unequal size, in the axils of which are single flowers
the extraction with further quantities, each of 25 ml, of with campanulate corolla, about 2 cm long and 1.5 cm wide,
chloroform until complete extraction of the alkaloids is effected purple or yellow-brown in colour, with five short, reflexed lobes,
(2.6.4), running each chloroform solution through the same five epipetalous stamens and one bilocular ovary with
plug of absorbent cotton. Extract the combined chloroform numerous ovules.
solutions with successive quantities of a mixture of 3 volumes
Identification
of 0.1 M sulphuric acid and 1 volume of ethanol (95 per
cent) until complete extraction of the alkaloids is effected, A. When examined under a microscope it shows epidermal
filtering each extract through a plug of absorbent cotton cells with sinuate anticlinal walls and cuticle which is often
previously moistened with water. Wash the mixed acid striated and furrowed. Covering and glandular hairs infrequent,
solutions with 10, 5 and 5 ml of chloroform, extracting each though more frequent in the young leaves and around the
chloroform solution with the same 20 ml of 0.05 M sulphuric veins; covering hairs, multicellular, uniseriate, with thin smooth

1388
IP 2007 BELLADONNA LEAF

walls; glandular hairs; short clavate glands with multicellular chromatogram obtained with 10 µl of test solution. Spray the
heads and glands with a long uniseriate stalk and ovoid plate with a freshly prepared 10 per cent w/v solution of
unicelluar head. Stomata, anisocytic, more frequent on the sodium nitrite until transparent and examine after 15 minutes.
lower epidermis. The midrib is characterised by an open arc of The colours due to hyoscyamine in the chromatogram change
vascular bundles with isolated groups of perimedullary phloem. from brown to reddish-brown but not to greyish-blue
Mesophyll dorsiventral with a single palisade layer. (atropine); any secondary bands are no longer visible.
Throughout the parenchyma and particularly just below the Foreign organic matter (2.6.1). Not more than 3 per cent.
palisade layer are cells containing microsphenoidal crystals
of calcium oxalate or, very rarely, cluster crystals. The stems Acid-insoluble ash (2.3.19). Not more than 3 per cent.
show pericyclic fibres and perimedullary bundles of phloem, Assay. Powder 50 g and determine the loss on drying (2.4.19 ),
few trichomes; the cortical parenchymatous cells and the pith by drying 2 g, accurately weighed, in an oven at 105o. From
cells contain microsphenoidal crystals of calcium oxalate. the remaining sample, weigh accurately about 10 g, moisten
B. Powder 1 g and shake for 2 minutes with 10 ml of 0.05 M with a mixture of 5 ml of dilute ammonia solution, 10 ml of
sulphuric acid. Filter and add to the filtrate 1 ml of strong ethanol (95 per cent) and 30 ml of ether and mix thoroughly.
ammonia solution and 5 ml of water. Extract with 15 ml of Transfer the mixture to a percolator with the aid of an extracting
ether, taking care to prevent the formation of an emulsion. Dry solvent mixture consisting of 3 volumes of ether and 1 volume
the ether extract over anhydrous sodium sulphate and filter. of chloroform. Allow to macerate for 4 hours and percolate
Evaporate the filtrate to dryness, add 0.5 ml of fuming nitric with the solvent mixture until complete extraction of the
acid and evaporate to dryness. Add 10 ml of acetone and, alkaloids is effected, ( 2.6.4).
dropwise, a 3 per cent w/v solution of potassium hydroxide in Concentrate the percolate to about 50 ml by distilling off the
ethanol (95 per cent); a deep violet colour develops. solvent mixture on a water-bath, and transfer to a separator,
C. Determine by thin-layer chromatography (2.4.17), coating previously rinsed with ether. Add a quantity of ether at least
the plate with silica gel G.. equal to 2.1 times the volume of the percolate and extract with
three quantities, each of 20 ml, of 0.5 M sulphuric acid.
Mobile phase. A mixture of 90 volumes of acetone, 7 volumes Transfer each acid extract to another separating funnel.
of water and 3 volumes of strong ammonia solution. Combine the acid extracts, make the solution alkaline with
Test solution. Add 15 ml of 0.05 M sulphuric acid to 0.6 g of dilute ammonia solution and extract with chloroform until
the material under examination, in fine powder, shake for complete extraction of the alkaloids has been effected. Wash
15 minutes, filter and wash the filter with 0.05 M sulphuric the combined chloroform extracts with 10 ml water, discard
acid until 20 ml of filtrate is obtained; add 1 ml of strong the water, evaporate the chloroform layer to dryness and heat
ammonia solution to the filtrate, extract with two quantities, the residue for 15 minutes on a water-bath. Redissolve the
each of 10 ml, of peroxide-free ether, separate the ether layer, residue in successive small quantities of chloroform,
by centrifugation if necessary, dry the combined ether extracts evaporating to dryness on a water-bath each time before
over anhydrous sodium sulphate, filter, evaporate to dryness adding the solvent. Heat for 15 minutes on a water-bath and
on a water-bath and dissolve the residue in 0.5 ml of methanol. dissolve the residue in 5 ml of chloroform. Add 20.0 ml of
0.01 M sulphuric acid, remove the chloroform by evaporation
Reference solution. Dissolve 50 mg of hyoscyamine sulphate
on a water-bath and titrate the excess of acid with 0.02 M
in 9 ml of methanol (solution A) and dissolve 15 mg of hyoscine
sodium hydroxide using methyl red solution as indicator.
hydrobromide in 10 ml of methanol (solution B); mix 8 ml of
solution A with 1.8 ml of solution B. 1 ml of 0.01 M sulphuric acid is equivalent to 0.005788 g of
total alkaloids calculated as hyoscyamine. Calculate the
Apply to the plate 10 µl and 20 µl of each solution as bands.
content of total alkaloids with reference to the dried material.
After development, dry the plate at 105o for 15 minutes, allow
to cool and spray with 10 ml of modified potassium Storage. Store protected from light and moisture.
iodobismuthate solution until the bands become visible as
orange or brown on a yellow background. The bands in the
chromatogram obtained with test solution have similar Rf
values to those in the chromatograms obtained with reference
solution (hyoscyamine in the lower third of the chromatogram;
hyoscine in the upper third) and are similar in colour and
atleast equal in size. Faint secondary bands may appear,
particularly in the middle of the chromatogram obtained with
20 µl of the test solution or near the line of application in the

1389
BHIBHITAKI IP 2007

Bhibhitaki 75 ml of methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 50 ml.
Belliric Myrobalan; Terminalia
Reference solution. Reflux 0.4 g of the coarsely powdered
bhibhitaki RS with 50-75 ml of methanol for 15 minutes, cool
and filter. Reflux the residue further for two times with 75 ml of
methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 10 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with anisaldehyde sulphuric acid reagent. Heat at 100º
for 5-10 minutes and examine the plate in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 25 per cent.
Water-soluble extractive (2.6.3). Not less than 35 per cent by
Bhibhitaki consists of the dried fruit pericarp of Terminalia Method 1.
belerica Roxb. (Fam. Combretaceae).
Ash (2.3.19). Not more than 8 per cent.
Bhibhitaki contains not less than 0.3 per cent w/w of ellagic
Acid-insoluble ash (2.3.19). Not more than 2 per cent.
acid and 0.75 per cent w/w of gallic acid, calculated on the
dried basis. Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Description. The dried pericarp appears as curved pieces of
irregular shapes, the external surface is velvety, wrinkled grey Loss on drying (2.4.19). Not more than 12.0 per cent,
to brown in colour and has astringent taste determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
Identification
contamination tests.
A. Macroscopic – The dried pericarp of the ripe fruit occurs Assay. Determine by liquid chromatography (2.4.14).
as curved pieces of irregular shapes with convex external
surface. The external surface appears velvety, slightly wrinkled Test solution. Weigh 0.5 g of coarsely powdered substance
grey to brown in colour. Internal surface is pale yellow. The under examination, add 50 ml of water, sonicate for 3 minutes
cut surface is with occasional projecting threads, representing and heat on a boiling water bath for 15 minutes, cool and
the vascular bundles. dilute to 100.0 ml with water and filter.

B. Microscopic – The cells of the epidermis has a characteristic Reference solution. A solution containing 0.01 per cent w/v
and a slightly bulged based with a hair like prolongation. each of gallic acid RS and ellagic RS in water.
Several vascular strands traverse the mesocarp in various NOTE — Use freshly prepared solution and protected from
directions. Peripheral layers of mesocarp have tangentially light.
elongated cells, devoid of starch grains, containing rosettes Chromatographic system
of calcium oxalate crystals and few small stone cells. – a stainless steel column 25 cm x 4.6 mm packed with
C. Determine by thin-layer chromatography (2.4.17), coating octadecylsilane bonded to porous silica (5 µm),
the plate with silica gel. – mobile phase: a gradient mixtures of acetonitrile and a
buffer solution prepared by dissolving 0.136 g of
Mobile phase. A mixture of 20 volumes of toulene, 45 volumes
potassium di-hydrogen orthophosphate in 500 ml of
of ethyl acetate and 20 volumes of glacial acetic acid and
water, add 0.5 ml of orthophosphoric acid and make
5 volumes of formic acid.
upto 1000 ml with water,
Test solution. Reflux 2 g of the coarsely powdered substance – flow rate. 1.5 ml per minutes,
under examination with 50-75 ml of methanol for 15 minutes, – spectrophotometer set at 270 nm,
cool and filter. Reflux the residue further for two times with – a 20 µl loop injector.

1390
IP 2007 BHRINGRAJ

Time Buffer solution Acetonitrile Leaf. Opposite, sessile to sub sessile, usually oblong,
(min) (per cent v/v) (per cent v/v) lanceolate, sub-entire, sub -acute or acute, strigose with
0 95 5 appressed hairs on both surfaces.
18 65 35 Flower. Solitary or 2, together on unequal axillary peduncles,
25 45 55 involucral bracts about 8, ovate, obtuse or acute, herbaceous,
strigose with oppressed hairs; ray flowers ligulate,
30 95 5
ligule small, spreading, scarcely as long as bracts, not toothed;
Inject the reference solution. The relative standard deviation
white disc flowers tubular, corolla often 4 toothed; pappus
for the replicate injections is not more than 2.0 per cent.
absent, except occasionally very minute teeth on the top of
Inject the test solution and reference solution. achene; stamen 5, filaments epipetalous, free, anthers united
Calculate the content of gallic acid and ellagic acid. into a tube with base obtuse; pistill bicarpellary; ovary inferior;
unilocular with one basal ovule.
Storage. Store protected from light, heat, moisture and against
attack by insects and rodents. Fruit. Achenial cypsella, one seeded, cuneate, with a narrow
wing, covered with warty excrescences, brown.
Seed. Dark brown, hairy and non endospermic.
B. Microscopic — Powder. Dark green; shows vessels in
Bhringraj large groups or single broken pieces with pitted walls,
Eclipta numerous fibres entire or in pieces, trichomes entire or in
pieces, warty, a few attached with epidermal and subsidiary
cells, anomocytic and anisocytic stomata.
Root. The cells of outer one or two rows of secondary cortex,
elongated or rounded with air cavities, while cells of inner
secondary cortex, elongated to irregular in shape. Stone cells
scattered in secondary cortex. Phloem rays broader towards
the periphery, cells rounded. Xylem rays distinct, run straight
in tangential section, rarely uniseriate and biseriate, cells pitted.
Stem. A few epidermal cells elongate to form characteristic
non-glandular trichomes. Secondary cortex composed of large,
rounded parenchymatous cells having wide air space. Vascular
bundle in a ring, collateral, endarch, of varying size. Vessels
barrel-shaped, some elongated with simple perforations, pitted
with spiral thickening. A few xylem fibres bifurcate. Xylem
rays uniseriate or biseriate.
Leaf. Anomocytic and anisocytic stomata and non-glandular
Bhringraj consists of the dried whole plant of Eclipta alba hairs are present on both surface, more abundant on lower
(L.) Hassk. (Fam. Asteraceae). side. Vascular bundle, fine in mid rib, central one largest while
Bhringraj contains not less than 0.1 per cent of wedelolcatone, four other small flanking either side of central bundle.
calculated on the dried basis. C. Determine by thin-layer chromatography (2.4.17), coating
Description. A green to greenish brown colour when the plate with silica gel F254.
completely dry. Mobile phase. A mixture of 9 volumes of toluene, 6 volumes
of acetone and 1 volume of formic acid.
Identification
Test solution. Reflux 1g of the coarsely powdered substance
A. Macroscopic — Root. Well developed, a number of under examination with 25 ml of methanol for 30 minutes, cool
secondary branches arise from main root up to about 7 mm in and filter. Reflux the residue further with 3 × 25 ml of methanol,
dia, cylindrical, greyish. cool and filter. Combine all the filtrates and concentrate under
Stem. Herbaceous, branched occasionally rooting at nodes, vacuum to 10 ml.
cylindrical or flat, rough due to oppressed white hairs, node Reference solution. Reflux 0.5 g bhringraj RS with 25 ml of
distinct, greenish, occasionally brownish. methanol for 30 minutes, cool and filter. Reflux the residue

1391
BHUIAMLA IP 2007

further with 3 × 25 ml of methanol, cool and filter. Combine all Bhuiamla


the filtrates and concentrate under vacuum to 5 ml.
Phyllanthus
Apply to the plate 10 µl of each solution. Allow the mobile
phase to rise 8 cm. Dry the plate in air and examine in ultra
violet light at 254 nm. The chromatographic profile of the test
solution is similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2 .0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 5.0 per cent.
Water- soluble extractive (2.6.3). Not less than 15.0 per cent
by Method I.
Ash (2.3.19). Not more than 22 per cent.
Acid-insoluble ash (2.3.19). Not more than 11 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 15.0 per cent,
Bhuiamla consists of the dried aerial parts of Phyllanthus
determined on 5 g by drying in an oven at 105º.
amarus Schum. & Thom. (Fam. Euphorbiaceae).
Microbial contamination (2.2.9). Complies with the microbial
Bhuiamla contains not less than 0.25 per cent of total
contamination tests.
phyllanthin and hypophyllanthin, calculated on the dried
Assay. Determine by liquid chromatography (2.4.14). basis.
Test solution. Reflux 5 g of the coarsely powdered substance Description. A green to greenish yellow in colour, taste,
under examination with 30 ml of methanol on a water- bath for slightly bitter.
30 minutes, cool and filter. Reflux the residue further with
methanol till the last extract turns colourless, cool and filter. Identification
Combine all the filtrates and concentrate to 100 ml.
A. Macroscopic — Stem teret, 1-4 mm in diameter. Leaves
Reference solution. 0.01 per cent w/v solution of oblong 5 × 3 mm, short stalked, greenish brown in colour.
wedelolactone RS in methanol.
B. Microscopic — Stem, inner cortex chlorenchymatous; xylem
Chromatographic system rays 1-2-seriate. Leaf stomata mostly paracytic; epidermal cell
– a stainless steel column 25 cm x 4.6 mm packed with wall markedly sinuous; rosette and prismatic crystals of calcium
octadecylsilane bonded to porous silica (5 µm), oxalate along the veins and midrib.
– mobile phase: 35 volumes of acetonitrile and 60 volumes
C. Determine by thin-layer chromatography (2.4.17), coating
of 0.1 per cent v/v phosphoric acid prepared by diluting
the plate with silica gel GF 254.
1ml of phosphoric acid to 1000 ml with water,
– flow rate. 1 ml per minute, Mobile phase. A mixture of 6 volumes of toluene, 2 volumes
– spectrophotometer set at 249 nm, of ethyl acetate, 1 volume of formic acid and 0.2 volume of
– a 20 µl loop injector. methanol.
Inject the reference solution. The test is not valid unless the Test solution. Reflux 2 g of coarsely powdered substance under
relative standard deviation for the replicate injections is not examination with 50 ml methanol on a boiling water-bath for
more than 2.0 per cent. 30 minutes, cool and filter. Reflux the residue further with 2 ×
50 ml of methanol, cool and filter. Combine all the filtrates and
Inject the test solution and reference solution.
concentrate under vacuum to 10 ml.
Calculate the content of wedelolactone.
Reference solution. Reflux 1 g of bhuiamla RS with 50 ml
Storage. Store protected from heat, moisture and against attack methanol on a boiling water-bath for 30 minutes, cool and
by insects and rodents. filter. Reflux the residue further with 2 × 50 ml of methanol,
cool and filter. Combine all the filtrates and concentrate under
vacuum to 5 ml.

1392
IP 2007 BRAHMI

Apply to the plate 10µl of each solution as bands 10 mm by Brahmi


2 mm. Allow the mobile phase to rise 8 cms above the line of
application. Dry the plate in air and spray with methanolic Bacopa
sulphuric acid (10 per cent) Heat at 120º for 5-10 minutes and
examine the plate in day light. The chromatographic profile of
the test solution is similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 15.0 per cent
by Method I.
Ash (2.3.19). Not more than 8.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent, Brahmi consists of the dried whole plant, preferably leaves
determined on 5 g by drying in an oven at 1050. and stem of Bacopa monnieri (Linn.) Pennell (Fam.
Scrophulariaceae).
Microbial contamination (2.2.9). Complies with the microbial
contamination tests. Brahmi contains not less than 2.5 per cent of bacoside A,
calculated on the dried basis.
Assay. Determine by liquid chromatography (2.4.14).
Description. A brown to reddish brown colour when
Test solution. Reflux about 2 g of the coarsely powdered completely dried or green colour when partially dried with
substance under examination with 50 ml of methanol on a slightly bitter taste.
water bath for 15 minutes, cool and filter. Reflux the residue
further with methanol till the last extract turns colorless, cool Identification
and filter. Combine all the filtrates and concentrate to 10.0 ml.
A. Macroscopic — Herbaceous comprising of stems, runner
Reference solution (a). A 0.020 per cent w/v solution of stems and leaves. Stems glabrous, leafless towards the base;
phyllanthin RS in methanol. internodes long. Leaves spatulate-obovate, sessile, and
Reference solution (b). A 0.020 per cent w/v solution of glabrous.
hypophyllanthin RS in methanol.
B. Microscopic — Cortex in stem composed of parenchyma
Chromatographic system cells enclosing large air spaces; xylem vessels radially arranged
– a stainless steel column 25 cm x 4.6 mm packed with xylem rays uniseriate; pith parenchyma collapsed. In leaf,
octadecylsilane bonded to porous silica (5µm), midrib indistinct, mesophyll isobilateral of spongy cells, a few
– mobile phase: a mixture of 65 volumes of methanol and prismatic crystals of calcium oxalate in mesophyll; stomata
35 volumes of water, anomocytic on both the surfaces of leaf.
– flow rate. 1.5 ml per minute,
C. Determine by thin-layer chromatography (2.4.17), coating
– spectrophotometer set at 230 nm,
the plate with silica gel G.
– a 20 µl loop injector.
Mobile phase. A mixture of 70 volumes of chloroform and
Inject the reference solution (a) and (b). The test is not valid 30 volumes of methanol.
unless the relative standard deviation for the replicate
injections for both the analyte peaks corresponding to Test solution. Reflux 2 g of coarsely powdered substance under
phyllanthin and hypophyllanthin is not more than 2.0 per cent. examination with 25 ml methanol for 15 minutes, cool and
filter. Reflux the residue further with 2 x 25 ml of methanol,
Inject the test solution, reference solutions (a) and (b).
cool and filter. Combine all the filtrates and concentrate under
Calculate the contents of phyllanthin and hypophyllanthin. vacuum to 25 ml.
Storage. Store protected from heat, moisture and against attack Reference solution. Reflux 0.4 g of coarsely powdered brahmi
by insects and rodents. RS with 5 ml methanol for 15 minutes, cool and filter.

1393
CASTOR OIL IP 2007

Apply to the plate 10 µl of each solution as bands 10 mm by Inject the reference solution. The relative retention times are
2 mm. Allow the mobile phase to rise 8 cms above the line of 1.00 for bacoside A3, about 1.04 for bacopaside II, 1.13 for
application. Dry the plate in air, spray with 20 per cent v/v jujubogenin isomer of bacopasaponine C and 1.19 for
sulphuric acid in methanol. Heat at 100º for 5-10 minutes and bacopasaponine C as bacoside A. The test is not valid unless
examine the plate in day light. The chromatographic profile of the relative standard deviation for the replicate injections is
the test solution is similar to that of the reference solution. not more than 2.0 per cent.
D. In the Assay, the chromatogram obtained with test solution Inject the test solution and reference solution.
corresponds to the chromatogram obtained with reference
Calculate the content of bacoside A.
solution.
Storage. Store protected from heat, moisture and against attack
Tests by insects and rodents.
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 22 per cent by Castor Oil
Method I. Castor Oil is the fixed oil obtained by cold expression from the
Ash (2.3.19). Not more than 18 per cent. seeds of Ricinus communis Linn. (Fam. Euphorbiaceae). It
may contain suitable antioxidants.
Acid-insoluble ash (2.3.19). Not more than 6.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for Description. A pale yellowish or almost colourless,
heavy metals, Method B (20 ppm). transparent, viscid liquid; odour, slight and characteristic.

Loss on drying (2.4.19). Not more than 12.0 per cent, Tests
determined on 5 g by drying in an oven at 1050.
Light absorption (2.4.7). Absorbance of a 1.0 per cent w/v
Microbial contamination (2.2.9). Complies with the microbial solution in ethanol (95 per cent) at the maximum at about 269
contamination tests. nm, not more than 1.0.
Assay. Determine by liquid chromatography (2.4.14).
Weight per ml (2.4.29). 0.945 g to 0.965 g.
Test solution. Reflux about 2 g of the coarsely powdered
Refractive index (2.4.27). 1.4758 to 1.4798.
substance under examination with 50 ml of methanol on a
water bath for 15 minutes, cool and filter. Reflux the residue Optical rotation (2.4.22). +3.5º to +6.0º.
further with methanol till the last extract turns colorless, cool Peroxide value (2.3.35). Not more than 5.0.
and filter. Combine all the filtrates and concentrate to 100.0 ml.
Acid value (2.3.23). Not more than 2.0.
Reference solution. A 0.2 per cent w/v solution of bacoside A
RS in methanol, prepared by heating gently on a water bath. Acetyl value (2.3.22). Not less than 143.
Chromatographic system Hydroxyl value (2.3.27). Not less than 150.
– a stainless steel column 25 cm x 4.6 mm packed with Saponification value (2.3.37). 176 to 187.
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a gradient mixtures of acetonitrile and a Iodine value (2.3.28). 82 to 90.
buffer solution prepared by dissolving 0.5 g phosphoric Foreign fatty substances. A mixture of 2 ml of the substance
acid in 800 ml of water, adjust pH to 2.8 with dilute under examination and 8 ml of ethanol (95 per cent) is clear.
phosphoric acid and dilute to 1000 ml with water,
– flow rate. 1.5 ml per minute, B. Shake 10.0 ml with 20.0 ml of light petroleum (60º to 80º)
– spectrophotometer set at 205 nm, and allow to separate; the volume of the lower layer is not less
– a 20 µl loop injector. than 16.0 ml.
Time Buffer (pH 2.8) Acetonitrile Storage. Store protected from light and moisture at a
(min) ( per cent v/v) ( per cent v/v) temperature not exceeding 15º.
0 70 30 Labelling. The label states (1) the name and quantity of any
25 60 40 added antioxidant; (2) whether the contents are suitable for
35 40 60 use in the manufacture of parenteral preparations.
36 70 30
45 70 30

1394
IP 2007 CLOVE OIL

Clove Oil Weight per ml (2.4.29). 1.038 g to 1.060 g.


Refractive index (2.4.27). 1.527 to 1.535, determined at 20º.
Clove oil is the oil distilled from the dried flower buds of
Syzygium aromaticum (Linn.) Merrill and Perry [Eugenia Heavy metals (2.3.13). 0.5 g complies with the limit test for
caryophyllus (Spreng.) Bull. and Harr. heavy metals, Method B (40 ppm).
Description. A clear, colourless or pale yellow liquid when Phenol. Shake 1 ml with 20 ml of hot water; the mixture shows
freshly distilled, becoming darker and thicker by ageing or not more than a scarcely perceptible acid reaction with blue
exposure to air; odour as of clove. litmus paper. Cool the mixture, pass the aqueous layer through
a wetted filter and treat the clear filtrate with 1 drop of ferric
Clove Oil contains not less than 85.0 per cent w/w and not
chloride test solution. The mixture has only a transient greyish-
more than 95.0 per cent w/w of phenolic substances, chiefly
green colour but not a blue or violet colour.
eugenol, C10H12O2.
Alkali-soluble matter. Place 80 ml of a 5 per cent w/v solution
Identification of potassium hydroxide in a 150-ml flask with a long neck
which is graduated in tenths of a ml and is of such a diameter
Determine by thin-layer chromatography (2.4.17), coating the that not less than 15 cm in length has a capacity of 10 ml.
plate with silica gel GF254. Clean the flask with sulphuric acid and rinse well with water
Mobile phase. Toluene before use. Add 10 ml of the oil and shake thoroughly at
5 minute intervals for 30 minutes at ambient temperature. Raise
Test solution. Dissolve 20 µl of the substance under the undissolved portion of the oil into the graduated part of
examination in 2 ml of toluene. the neck of the flask by the gradual addition of more of the
Reference solution. Dissolve 20 µl of eugenol RS in 2 ml of potassium hydroxide solution; allow to stand for not less
toluene. than 24 hours and read off the volume of the undissolved
portion of the oil which measures between 1.0 and 1.5 ml.
Apply to the plate 20 µl of the test solution and 10 µl of the
reference solution as bands 20 mm by 3 mm. Use an unlined Assay. Determine by gas chromatography (2.4.14).
tank, develop the chromatogram immediately after pouring Test solution (a). A 0.2 per cent w/v solution of the oil under
the mobile phase into the tank and allow the mobile phase to examination in ethanol (95 per cent).
rise 10 cm. Dry the plate, allow to stand for 5 minutes and Test solution (b). A 0.2 per cent w/v solution of the oil under
again allow the mobile phase to rise 10 cm under the same examination and 0.15 w/v of 1-decanol (internal standard) in
conditions. Following the second development, dry the plate ethanol (95 per cent).
in air, examine in ultraviolet light at 254 nm and mark the
quenching bands. In the chromatogram obtained with the test Reference solution. A solution containing 0.2 per cent w/v
solution there is a quenching band in the middle of the plate solution of eugenol RS and 0.15 per cent w/v of the internal
corresponding to the quenching band due to eugenol in the standard in ethanol (95 per cent).
chromatogram obtained with the reference solution. A weak Chromatographic system
quenching band may also be seen in the chromatogram – a glass column 1.5 m x 4 mm, packed with 3 per cent w/w
obtained with the test solution with an Rf value slightly lower of dimethyl silicone fluid on acid-washed diatomaceous
than that of the band corresponding to eugenol support (120 mesh),
(acetyleugenol). Spray the plate with about 10 ml of – temperature:
anisaldehyde solution, heat at 100º to 105º for 10 minutes and column.110º for 18 minutes, then increased to 170º at a
examine in daylight. In the chromatogram obtained with the rate of 12º per minute and maintained at this temperature
test and reference solutions the bands corresponding to for 2 minutes,
eugenol are strongly coloured brownish-violet and any band inlet port. 220º,
corresponding to acetyleugenol in the chromatogram obtained detector. 300º,
with the test solution is faintly violet-blue. Other coloured – flow rate 40 ml per minute of the carrier gas.
bands may be visible in the chromatogram obtained with the
Calculate the eugenol content in the oil under examination
test solution, in particular a faint red band in the lower part of
using the ratios of the area of the peak corresponding to
the chromatogram and a reddish-violet band in the upper part
eugenol to the area of the peak due to the internal standard in
(caryophyllene).
the chromatogram obtained with test solutions (b) and the
reference solution.
Tests
Storage. Store protected from light in well-filled containers at
Optical rotation (2.4.22). 0º to –1.50º. a temperature not exceeding 30º.

1395
COLEUS IP 2007

Coleus 10 minutes and examine the plate in day light. The


chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 15.0 per cent.
Water-soluble extractive (2.6.3). Not less than 18.0 per cent
by method I.
Ash (2.3.19). Not more than 15.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
Coleus consists of the whole or cut dried roots of Coleus determined on 5 g by drying in an oven at 1050.
forskohlii Briq. (Fam. Lamiaceae).
Microbial contamination (2.2.9). Complies with the microbial
Coleus contains not less than 0.4 per cent w/w of forskolin, contamination tests.
calculated on the dried basis.
Assay. Determine by liquid chromatography (2.4.14).
Description. The roots are light brown in color, generally long
and radially spread. They have an aromatic characteristic odor Test solution. Weigh 3 g of coarsely powdered substance
and the taste is slightly pungent. under examination, add 50 ml of methanol and reflux on a
water bath for 15 minutes, cool and filter. Reflux the residue
Identification two times with 75 ml of methanol, cool and filter, Concentrate
the filterate to 100.0 ml.
A. Macroscopic — Roots are brown, longitudinally wrinkled,
fracture short, cut surface yellowish white. Reference solution. A 0.1 per cent w/v solution of forskolin
RS in methanol.
B. Microscopic — The outermost layer consists of rectangular
cork cells, cork cambium, rectangular parenchymatous region Chromatographic system
containing sclereids and calcium oxalate crystals. Vascular – a stainless steel column 25 cm x 4.6 mm packed with
cambium is present in the form of a continuous ring. The octadecylsilane bonded to porous silica (5 µm),
tracheids and tracheidal fibres have bordered pits. – mobile phase: filtered and degassed mixture of 45
volumes of acetonitrile and 55 volumes of water,
C. Determine by thin-layer chromatography (2.4.17), coating
– flow rate. 1.8 ml per minute,
the plate with silica gel.
– spectrophotometer set at 220 nm,
Mobile phase. A mixture of 75 volumes of benzene, and – a 20 µl loop injector.
25 volumes of ethyl acetate.
Inject the reference solution. The relative standard deviation
Test solution. To 5 g of the coarsely powdered substance for the replicate injections is not more than 2.0 per cent.
under examination, add 50 ml of methanol and reflux for
15 minutes, cool and filter. Reflux the residue further for two Inject the test solution and reference solution.
times with 50 ml of methanol, cool and filter. Combine all the Calculate the content of forskolin.
filtrates and concentrate under vacuum to 100 ml.
Storage. Store protected from moisture and against attack by
Reference solution. To 1 g of coleus RS add 50 ml of methanol insects and rodents.
and reflux for 15 minutes, cool and filter. Reflux the residue
further for two times with 50 ml of methanol, cool and filter.
Combine all the filtrates and concentrate under vacuum to Eucalyptus Oil
20 ml.
Apply to the plate 20 µl of each solution as bands 10 mm by Nilgiri Oil
2 mm. Allow the mobile phase to rise 8 cms. Dry the plate in air, Eucalyptus Oil is the essential oil obtained by steam distillation
spray with vanilin sulphuric acid reagent. Heat at 100º for 5- and rectification from the fresh leaves or the fresh terminal

1396
IP 2007 GARCINIA

branches of various species of eucalyptus like Eucalyptus liquid of the first determination with the addition of 0.5 ml of
globulus Labill., E. fruticetoruµm F. von Muell., and E. smithii 0.5 M potassium hydroxide in ethanol (60 per cent). Not
(R. T. Baker) (Fam. Myrtaceae). more than 2.0 ml of 0.5 M potassium hydroxide in ethanol
Eucalyptus Oil contains not less than 60 per cent w/w of (60 per cent) is required in the second determination.
cineole, C10H18O. Phellandrene. Mix 1 ml with 2 ml of glacial acetic acid and
5 ml of light petroleum (40º to 60º), add 2 ml of a saturated
Description. A colourless or pale yellow liquid; odour, aromatic
solution of sodium nitrite and shake gently; no crystalline
and camphoraceous; taste, pungent and camphoraceous,
precipitate is produced in the upper layer within 1 hour.
followed by a sensation of cold.
Assay (2.3.24) Determine the content of cineole in the oil.
Identification
Storage. Store in well-filled, tightly-closed containers at a
Determine by thin-layer chromatography (2.4.17), coating the temperature not exceeding 30º.
plate with silica gel G.
Mobile phase. A mixture of 90 volumes of toluene and
10 volumes of ethyl acetate. Garcinia
Test solution. Dissolve 1 g of the substance under examination
in 100 ml of toluene. Vilayati Imli
Reference solution. A 1 per cent w/v solution of cineole RS in
toluene.
Apply to the plate 2 µl of each solution. After development,
dry the plate in air, spray with anisaldehyde solution, using
about 10 ml for a 200 mm x 200 mm plate, heat at 105º for
10 minutes and examine in daylight and in ultraviolet light at
365 nm. In the chromatogram obtained with the reference
solution a dark brown spot due to cineole is visible in daylight
in the middle part; when examined in ultraviolet light at
365 nm, the spot shows a brown fluorescence. The principal
spot in the chromatogram obtained with the test solution
corresponds to that of cineole; no carmine-brown spot appears
in daylight in the upper third of the chromatogram and when
examined in ultraviolet light at 365 nm no spot showing a
greenish brown fluorescence appears in the upper third
Garcinia is the dried deseeded fruit of Garcinia cambogia
(citronellal). Other spots may be visible in the upper and lower
Desr. (Fam. Guttiferae).
thirds of the chromatogram.
Garcinia contains not less than 12.0 per cent of total
Tests hydroxycitric acid and hydroxycitric acid lactone, calculated
on the dried basis.
Optical rotation (2.4.22). 0º to +10º.
Description. Dark brown to blackish brown fruits. Taste, acidic.
Refractive index (2.4.27). 1.457 to 1.469.
Weight per ml (2.4.29). 0.897 g to 0.924 g. Identification
Aldehydes. Place 10 ml in a glass-stoppered tube (150 mm x A. Macroscopic — Dark brown to blackish brown fruits,
25 mm) add 5 ml of toluene and 4 ml of ethanolic ovoid, longitudinally grooved.
hydroxylamine solution, shake vigorously and titrate
immediately with 0.5 M potassium hydroxide in ethanol B. Microscopic — Mesocarp very wide composed of
(60 per cent) until the red colour changes to yellow. Continue parenchymatous cells of various sizes and shapes. Compound
the shaking and neutralising until the pure yellow colour of starch grains and prismatic crystals of calcium oxalate traverse
the indicator is permanent in the lower layer after shaking throughout the parenchymatous cells of the mesocarp.
vigorously for 2 minutes and allowing separation to take place; C. In the Assay, the principal peak in the chromatogram
the reaction is complete in about 15 minutes. Repeat the obtained with test solution has a retention time similar to that
operation using a further 10 ml of the substance under of the peak due to hydroxycitric acid in the chromatogram
examination, and as the standard for the end-point, the titrated obtained with reference solution.

1397
GOKHRU IP 2007

Tests not more than 1.5 and the relative standard deviation for the
replicate injections is not more than 2.0 per cent.
Citric Acid. Not more than 2.0 per cent.
Inject the test solution and reference solution (a).
Determine by liquid chromatography (2.4.14).
Calculate the sum of the contents of hydroxycitric acid and
Test solution, reference solutions (a), (b) and chromatographic hydroxycitric acid lactone.
system as described under Assay.
Storage. Store protected from heat, moisture and against attack
Inject test solution and reference solution (b). by insects and rodents.
Calculate the content of citric acid.
Foreign organic matter (2.6.1). Not more than 5.0 per cent. Gokhru
Water-soluble extractive (2.6.3). Not less than 40.0 per cent Tribulus
by Method I.
Ash (2.3.19). Not more than 8.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 1.5 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh about 5 g of the coarsely powdered
substance under examination and transfer to a 250-ml beaker.
Gokhru consists of dried fruits of Tribulus terrestris L.
Add 5 ml of dilute phosphoric acid and 100 ml of water,
concentrate to half the volume by heating, cool and filter. Gokhru contains not less than 0.5 per cent of diosgenin,
Extract the residue further with water till the last extract turns calculated on the dried basis.
colorless, cool and filter. Combine all the filtrates and Description. Pedicellate and globose fruits, having wedge-
concentrate under vacuum to 250.0 ml. shaped cocci, covered with short and stiff spines. Possesses
Reference solution (a). A 0.8 per cent w/v solution of faintly aromatic smell and acrid taste.
hydroxycitric acid calcium salt RS in dilute phosphoric acid.
Identification
Reference solution (b). A 0.1 per cent w/v solution of citric
acid in dilute phosphoric acid. A. Macroscopic — Fruit is pedicellate, having wedge-shaped
cocci, covered with spines. Surface of schizocarp is rough.
Chromatographic system
B. Microscopic — Pericarp is differentiated into epicarp,
– a stainless steel column 25 cm x 4.6 mm packed with
mesocarp and endocarp. Epicarp is surrounded by non-
octadecylsilane bonded to porous silica (5 µm),
glandular trichomes.
– mobile phase: a buffer solution prepared by dissolving
1.36 g of potassium dihydrogen phosphate in 900 ml of C. Determine by thin-layer chromatography (2.4.17), coating
water, adjusting the pH to 2.5 with dilute phosphoric the plate with silica gel G.
acid and diluting to 1000.0 ml with water, Mobile phase. A mixture of 8 volumes of toluene and 2 volumes
– flow rate. 1 ml per minute, of ethyl acetate.
– spectrophotometer set at 215 nm,
Test solution. Reflux 5 g of coarsely powdered substance under
– a 20 µl loop injector.
examination with 50 ml of methanol for 15 minutes, cool and
Inject the reference solution (a). The test is not valid unless filter. Reflux the residue further with 50 ml of methanol, cool
the relative retention times are about 0.8 for hydroxycitric and filter. Combine both the filtrates and concentrate under
acid lactone, 1.0 for hydroxycitric acid and 2.0 for citric vacuum to dryness. Extract the dried residue with 10 ml of
acid, the resolution factor between hydroxycitric acid lactone methanol at 50° for 10 minutes, filter the solution and use
and hydroxycitric acid is not less than 1.8, the tailing factor is filtrates for analysis.

1398
IP 2007 GUAR GUM

Reference solution. Reflux 2.5 g of gokhru RS with 50 ml of Storage. Store protected from heat, moisture and against attack
methanol for 15 minutes, cool and filter. Reflux the residue by insects and rodents.
further with 50 ml of methanol, cool and filter. Combine both
the filtrates and concentrate under vacuum to dryness. Extract
the dried residue with 5 ml of methanol at 50° for 10 minutes,
filter the solution and use the filtrates for analysis. Guar Gum
Apply to the plate 20 µl of each solution as bands 10 mm by Guar Gum is a gum obtained from the ground endosperms of
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air, the seeds of Cyamopsis tetragonolobus (Linn.) Taub or other
spray with anisaldehyde sulphuric acid. The chromatographic species of Cyamopsis (Fam. Leguminosae). It consists mainly
profile of the test solution is similar to that of the reference of a high molecular weight hydrocolloidal polysaccharide,
solution. composed of galactan and mannan units combined through
glycosidic linkages.
Tests
Description. An almost white to pale yellowish white powder;
Foreign organic matter (2.6.1). Not more than 2 per cent. odour, characteristic.
Ethanol-soluble extractive (2.6.2). Not less than 3 per cent.
Identification
Water-soluble extractive (2.6.3). Not less than 15 per cent by
method I. A. When mounted in lactophenol and examined under a
microscope, irregular, angular particles of various sizes and
Ash (2.3.19). Not more than 11 per cent.
shapes are seen.
Acid-insoluble ash (2.3.19). Not more than 1 per cent. B. To 0.1 g add 1 ml of 0.2 M iodine; the mixture does not
Heavy metals (2.3.13). 1.0 g complies with the limit test for acquire an olive-green colour.
heavy metals, Method B (20 ppm). C. Dissolve 0.1 g in 20 ml of water by shaking and add 0.5 ml
Loss on drying (2.4.19). Not more than 5 per cent, determined of hydrogen peroxide solution (20 vol) and 0.5 ml of a 1 per
on 5 g by drying in an oven at 105º. cent w/v solution of benzidine in ethanol (90 per cent), shake
and allow to stand; no blue colour is produced (distinction
Microbial contamination (2.2.9). Complies with the microbial
from acacia).
contamination tests.
D. Mount a small quantity in ruthenium red solution and
Assay. Determine by liquid chromatography (2.4.14).
examine under a microscope; the particles do not acquire a
Test solution. Reflux 5.0 g of the substance under examination pink colour (distinction from sterculia gum and agar).
with 50 ml of sulphuric acid (10 per cent) for 4 hours. Cool
E. To 2 ml of a 0.5 per cent w/v solution add 2 ml of a 20 per
and transfer to separating funnel. Extract with 50 ml of ethyl
cent w/v solution of lead acetate; a flocculent precipitate is
acetate. Repeat the extraction 3 times. Pass the ethyl acetate
produced (distinction from acacia, ghatti gum and sterculia).
layer through sodium sulphate and evaporate. Dissolve the
residue with 50 ml of methanol. Tests
Reference solution. A 0.1 per cent w/v solution of diosgenin
Acidity or alkalinity. A 0.5 per cent w/v solution is neutral to
RS in methanol.
litmus paper.
Chromatographic system
Tannin. To 5 ml of a 0.5 per cent w/v solution add 0.1 ml of
– a stainless steel column 25 cm x 4.6 mm packed with
ferric chloride test solution; no bluish black colour is produced.
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: 80 volumes of acetonitrile and 20 volumes Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium
of methanol, carbonate, add 10 ml of bromine solution and mix thoroughly.
– flow rate. 1 ml per minute, Evaporate to dryness on a water-bath, gently ignite and
– spectrophotometer set at 210 nm, dissolve the cooled residue in a mixture of 16 ml of brominated
– a 20 µl loop injector. hydrochloric acid and 45 ml of water. Remove the excess of
bromine with 2 ml of stannous chloride solution AsT. The
Inject the reference solution. The relative standard deviation
resulting solution complies with the limit test for arsenic
for the replicate injections is not more than 2.0 per cent.
(3 ppm).
Inject the test solution.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Calculate the content of diosgenin. heavy metals, Method B (20 ppm).

1399
GUDMAR IP 2007

Protein. Not more than 5.0 per cent, determined by the Identification
following method. Carry out the determination of nitrogen
(2.3.30), using about 3.5 g, accurately weighed, and multiplying A. Macroscopic — Leaves, simple, petiolate about 2 to 6 cm
the percentage of nitrogen determined by 6.25 to obtain the long and 1 to 4 cm broad, yellowish brown on adaxial and dark
percentage of protein. green on abaxial side.

Acid-insoluble matter. Not more than 3.0 per cent, determined B. Microscopic — Upper and lower epidermis covered with
by the following method. Weigh accurately about 1.5 g and cuticle having uni to tri cellular covering trichomes which are
disperse in 150 ml of water and 1.5 ml of sulphuric acid. Warm slightly curved at the bulbous base. Below the epidermis is
on a water-bath for 6 hours, replacing the water lost by single layer of palisade cells followed by 2-3 layered spongy
evaporation. Add about 0.5 g of a suitable filter-aid, accurately parenchyma. Starch gains are simple and present in spongy
weighed, and filter through a suitable ashless filter paper. Wash parenchyma. Midrib region shows 2-7 layers of
the residue several times with hot water, dry the filter and its collenchymatous cells. Stomata are of paracytic type, mostly
contents at 105º for 3 hours. Cool in a desiccator, weigh and on lower the surface. There is a fan shaped vascular bundle in
subtract the weight of the filter aid. the centre. Each vascular bundle is collateral, closed and
surrounded by parenchymatous sheath. Rosette crystal of
Microbial contamination (2.2.9). Total bacterial count: Not calcium oxalate present in the spongy parenchyma.
more than 5000 per g. 1 g is free from Escherichia coli and 10 g
is free from Salmonellae. C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G.
Ash (2.3.19). Not more than 2.0 per cent, determined on 1.0 g.
Mobile phase. A mixture of 5 volumes of chloroforms, 1 volume
Loss on drying (2.4.19). Not more than 13.0 per cent, of methanol and 1 volume of ethyl acetate.
determined on 0.5 g by drying in an oven at 105º.
Test solution. Reflux 5 g of the coarsely powdered substance
under examination with 50 ml of ethanol (50 per cent v/v) for
15 minutes, cool and filter. Reflux the residue further with 2 ×
50 ml of ethanol (50 per cent v/v), cool and filter. Combine all
Gudmar the filtrates and concentrate under vacuum to 25 ml. Take 5 ml
Gymnema of resulting solution, add 5 ml ethanol and 2 ml of potassium
hydroxide and reflux for 1 hour. Cool and add 1.8 ml of 12 M
hydrochloric acid and heat on water bath. Cool and adjust
the pH to 7.5-8.5 with 11 per cent potassium hydroxide. Dilute
the solution with ethanol (50 per cent v/v) to 100 ml and filter.
Reference Solution. Reflux 1 g of gudmar RS with 50 ml of
ethanol (50 per cent v/v) for 15 minutes, cool and filter. Reflux
the residue further with 2 × 50 ml of ethanol (50 per cent v/v),
cool and filter. Combine all the filtrates and concentrate under
vacuum to 10 ml. Take 5 ml of resulting solution, add 5 ml
ethanol and 2 ml of potassium hydroxide and reflux for 1 hour.
Cool and add 1.8 ml of 12 M hydrochloric acid and heat on
water bath. Cool and adjust the pH to 7.5-8.5 with 11 per cent
potassium hydroxide. Dilute the solution with ethanol
(50 per cent v/v) to 50 ml and filter.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with anisaldehyde sulphuric acid solution. Heat at
Gudmar consists of the dried mature leaves of Gymnema 100° for 10 minutes and examine the plate in day light. The
sylvestre R.Br. (Fam. Asclepiadaceae). chromatographic profile of the test solution is similar to that
Gudmar contains not less than 1.0 per cent of gymnemic acids of the reference solution.
(calculated as gymnemagenin), calculated on dried basis.
Tests
Description. Greenish-yellow in colour, surface pubescent
on both sides and characteristic odour with extremely bitter Foreign organic matter (2.6.1). Not more than 2.0 per cent.
and acrid taste. Ethanol-soluble extractive (2.6.2). Not less than 5.0 per cent.

1400
IP 2007 GUDUCHI

Water-soluble extractive (2.6.3). Not less than 20.0 per cent Guduchi
by method I.
Tinospora, Giloe, Amrita
Ash (2.3.19). Not more than 15.0 per cent .
Acid-insoluble ash (2.3.19). Not more than 6.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 14.0 per cent,
determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux 5 g of the coarsely powdered substance
under examination with 50 ml of ethanol (50 per cent v/v) for
15 minutes, cool and filter. Reflux the residue further with 2 ×
50 ml of ethanol (50 per cent v/v), cool and filter. Combine all
the filtrates and concentrate under vacuum to 25 ml. Take 5 ml
of resulting solution, add 5 ml ethanol and 2 ml of potassium Guduchi consists of the dried, mature pieces of stem of
hydroxide and reflux for 1 hour. Cool and add 1.8 ml of 12 M Tinospora cordifolia (Willd.) Miers (Fam. Menispermaceae).
hydrochloric acid and heat on water bath. Cool and adjust Guduchi contains not less than 0.02 per cent w/w of
the pH to 7.5-8.5 with 11 per cent potassium hydroxide. Dilute cordifolioside A, calculated on the dried basis.
the solution with ethanol (50 per cent v/v) to 100 ml and filter.
Description. A greyish-black in colour, fibrous fracture and
Reference solution. A 0.01 per cent w/v solution of no distinct odour with bitter taste.
gymnemagenin RS in methanol.
Chromatographic system Identification
– a stainless steel column 25 cm × 4.6 mm packed with A. Macroscopic — Stem-pieces glabrous, cylindrical, solid,
octadecylsilane bonded to porous silica (5 µm), lenticillate, 5-15 mm in diameter having light brown surface
– mobile phase (A). 80 per cent v/v acetonitrile, marked with warty protuberances due to circular lenticels.
(B). 0.1 per cent w/v dihydogen potassium Transversely smoothened surface shows a radial structure
phosphate, with conspicuous medullary rays traversing porous tissues.
– flow rate. 1.5 ml per minute,
– spectrophotometer set at 210 nm, B. Microscopic — Transverse section of stem shows
– a 20 µl loop injector. outermost layer of cork which is differentiated in to outer zone
of thick walled, compressed cells and inner zone of thin walled,
Time Mobile phase A Mobile phase B
tangential cells. Cork broken at some places due to lenticels.
(min) (per cent v/v) (per cent v/v)
Cortex consists of 3-5 rows of irregularly arranged tangential,
0 25 75 chlorenchymatous cells with numerous intercellular spaces.
20 50 50 Inner cortex filled with plenty of starch gains. Vascular zone
30 25 75 consists of 10-12 wedge-shaped strips of xylem externally
surrounded by semi-circular strips of phloem. Cambium
Inject the reference solution. The test is not valid unless the composed of one to two layers of tangentially elongated cells.
relative standard deviation for the replicate injections is not Primary phloem appears crushed and obliterated; secondary
more than 2.0 per cent. phloem groups are massive. Pith composed of large, thin walled
Inject the test solution and reference solution. cells mostly containing starch gains.

Calculate the content of gymnemagenin. C. Determine by thin-layer chromatography (2.4.17), coating


the plate with silica gel G.
Storage. Store protected from heat, moisture and against attack
by insects and rodents. Mobile phase. A mixture of 85 volumes of chloroform and
15 volumes of methanol.
Test solution. Reflux 2 g of the coarsely powdered substance

1401
GUGGUL RESIN IP 2007

under examination with 25 ml of methanol for 15 minutes, cool Time Water Acetonitrile
and filter. Reflux the residue further with 2 × 25 ml of methanol, (min) (per cent v/v) (per cent v/v)
cool and filter. Combine all the filtrates and concentrate under 0 80 20
vacuum to 5 ml.
25 20 80
Reference solution. Reflux 2 g of the guduchi RS with 25 ml of 30 80 20
methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 × 25 ml of methanol, cool and filter. Combine all Inject the reference solution. The test is not valid unless the
the filtrates and concentrate under vacuum to 5 ml. relative standard deviation for the replicate injections is not
more than 2.0 per cent.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air, Inject the test solution and reference solution.
spray with a anisaldehyde solution. Heat at 110° for 10 minutes
Calculate the content of cordifolioside A.
and examine the plate in day light. The chromatographic profile
of the test solution is similar to that of the reference solution. Storage. Store protected from heat, moisture and against attack
by insects and rodents.
Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 1.5 per cent. Guggul Resin
Water-soluble extractive (2.6.3). Not less than 9.0 per cent by Guggul, Commiphora
method I.
Ash (2.3.19). Not more than 10.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 10.0 per cent,
determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux 2 g of the coarsely powdered substance
under examination with 50 ml of methanol on a water-bath for
15 minutes, cool and filter. Reflux the residue further with Guggul Resin is the oleoresin exudation from Commiphora
methanol till the extract turns colourless, cool and filter. wightii (Arnott) Bhandari (Commiphora mukul (Arn.)
Combine all the filtrates and concentrate to a volume slightly Bhandari, Balsamodendron mukul Hook. ex Stocks) (Fam.
less than 25 ml. Dilute with methanol to 25.0 ml. Burseraceae).
Guggul Resin contains not less than 1.0 per cent and not more
Reference solution. A 0.004 per cent w/v solution of
than 1.5 per cent of gugulsterones (Z and E).
cordifolioside A RS in methanol.
Description. Light to dark-brown conglomerates of tears,
Chromatographic system
rounded or irregular, slightly sticky to touch; odour, faintly
– a stainless steel column 25 cm × 4.6 mm packed with
balsamic.
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a gradient mixtures of water and Identification
acetonitrile,
– flow rate. 1 ml per minute, A. Prepare a 0.005 per cent w/v solution in ethanol (95 per
– spectrophotometer set at 210 nm, cent) of the residue obtained in the test for Ethanol-soluble
– a 20 µl loop injector. extractive.
When examined in the range 230 nm to 360 nm (2.4.7), the
resulting solution shows absorption maxima at about 245 nm
and 327 nm.

1402
IP 2007 GUGULIPID

B. Determine by thin-layer chromatography (2.4.17), coating Chromatographic system


the plate with silica gel G. – a stainless steel column 25 cm x 4.6 mm, packed with
Mobile phase. A mixture of 3 volumes of light petroleum (60º octadecylsilyl silica gel (5 µm),
to 80º) and 1 volume of ethyl acetate. – mobile phase: a filtered and degassed mixture of 45
volumes of acetonitrile and 55 volumes of water,
Test solution. Dissolve 0.5 g of the residue obtained in the – flow rate. 2 ml per minute,
test for Ethanol-soluble extractive in 100 ml of ethanol (95 per – spectrophotometer set at 242 nm,
cent). – a 20 µl loop injector.
Reference solution. A 0.5 per cent w/v solution of the residue Inject the reference solution. The test is not valid unless the
obtained similarly from guggul resin RS in ethanol (95 per relative retention times are about 0.69 for gugulsterone E and
cent). 1.0 for gugulsterone Z and the relative standard deviation for
Apply to the plate 20 µl of each solution. After development, replicate injections is not more than 2.0 per cent.
dry the plate in air until the odour of the solvent is no longer Inject the test solution and the reference solution. Calculate
detectable and spray with a 50 per cent w/v solution of the contents of gugulsterones (Z and E).
sulphuric acid. The principal spots in the chromatogram
obtained with the test solution correspond to the spots in the Storage. Store protected from light at a temperature not
chromatogram obtained with the reference solution. exceeding 30º.

Tests
Ethyl acetate-soluble extractive. Not less than 25.0 per cent, Gugulipid
determined by the following method. Crush the substance
under examination to a coarse powder. Shake 5.0 g of the Gugulipid is the ethyl acetate extractive of Guggul Resin.
powder with 25 ml of light petroleum (60º to 80º) for 1 hour Gugulipid contains not less than 4.0 per cent and not more
and separate the liquid by filtration. Repeat the extraction than 6.0 per cent of gugulsterones (Z and E).
twice and dry the defatted material over phosphorus pentoxide
at room temperature at a pressure not exceeding 2.75 kPa for Description. A brown, viscous liquid.
8 hours. Crush the dried material and extract with four quantities,
Identification
each of 25 ml, of ethyl acetate by shaking each time for 1 hour
followed by filtration through a sintered-glass funnel (porosity A. When examined in the range 230 nm to 360 nm (2.4.7), a
No. 3) and combining the filtrates. Evaporate the combined 0.005 per cent w/v solution in chloroform shows absorption
filtrates, dry over phosphorus pentoxide at room temperature maxima at about 245 nm and 327 nm; absorbance at about
at a pressure not exceeding 2.75 kPa for 12 hours and weigh. 245 nm, about 0.87 and at about 327 nm, about 0.52.
Ethanol-soluble extractive (2.3.46). Not less than 35.0 per cent, B. Determine by thin-layer chromatography (2.4.17), coating
determined by the following method. Crush the substance the plate with silica gel G.
under examination to a coarse powder. Macerate 5.0 g of the
Mobile phase. A mixture of 3 volumes of light petroleum
powder with 100 ml of ethanol (95 per cent) in a closed flask
(60º to 80º) and 1 volume of ethyl acetate.
for 24 hours, shaking frequently during the first 6 hours and
allowing to stand for 18 hours. Filter rapidly taking care to Test solution. Dissolve 0.25 g of the substance under
avoid loss of ethanol, evaporate 25 ml of the filtrate to dryness, examination in 100 ml of ethanol (95 per cent).
dry at 105º and weigh. Reference solution. A 0.25 per cent w/v solution of
Sulphated ash (2.3.18). Not more than 10.0 per cent, determined gugulipid RS in ethanol (95 per cent).
on 0.5 g. Apply to the plate 5 µl of each solution. After development,
Assay. Determine by liquid chromatography (2.3.14). dry the plate in air until the odour of the solvent is no longer
detectable and spray with a 50 per cent w/v solution of
Test solution. Weigh accurately about 3.0 g of the substance
sulphuric acid. The principal spots in the chromatogram
under examination, add 50 ml of acetonitrile, reflux on a water-
obtained with the test solution correspond to those in the
bath for 30 minutes, cool and filter. Reflux the residue further
chromatogram obtained with the reference solution.
with three portions, each of 30 ml, of acetonitrile, cool and
filter. Combine the filtrates and concentrate to 100.0 ml. Tests
Reference solution. A 0.02 per cent w/v solution of
Assay. Determine by liquid chromatography (2.4.14).
gugulsterones (Z and E) RS in acetonitrile.

1403
GUGULIPID TABLETS IP 2007

Test solution. Weigh accurately about 0.75 g of the substance Apply to the plate 5 µl of each solution. After development,
under examination, add 50 ml of acetonitrile and warm on a dry the plate in air until the odour of the solvent is no longer
boiling water-bath for 10 minutes. Cool and add sufficient detectable and spray with a 50 per cent w/v solution of
acetonitrile to produce 100.0 ml. sulphuric acid. The principal spots in the chromatogram
Reference solution. A solution containing 0.02 per cent w/v of obtained with the test solution correspond to those in the
gugulsterones (Z and E) in acetonitrile. chromatogram obtained with the reference solution.

Chromatographic system Tests


– a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilyl silica gel (5 µm), Disintegration (2.5.1). 60 minutes.
– mobile phase: a filtered and degassed mixture of Other tests. Comply with the tests stated under Tablets.
45 volumes of acetonitrile and 55 volumes of water,
Assay. Determine by liquid chromatography (2.4.13).
– flow rate. 2 ml per minute,
– spectrophotometer set at 242 nm, Test solution. Weigh and powder 20 tablets. Weigh accurately
– a 20 µl loop injector. a quantity of the powder containing about 10 mg of
gugulsterones (Z and E) and extract with five quantities, each
Inject the reference solution. The test is not valid unless the
of 20 ml, of acetonitrile, with the aid of heat. Combine the
relative retention times are about 0.69 for gugulsterone E and
extracts and concentrate to 50.0 ml. Filter the solution through
1.0 for gugulsterone Z and the relative standard deviation for
a membrane filter disc with an average pore diameter not greater
replicate injections is not more than 2.0 per cent.
than 1.0 µm and use the filtrate.
Inject alternately the test solution and the reference solution.
Reference solution. A solution containing 0.02 per cent w/v of
Calculate the contents of gugulsterones (Z and E).
gugulsterones (Z and E) in acetonitrile.
Storage. Store protected from light at a temperature not
Chromatographic system
exceeding 30º.
– a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilyl silica gel (5 µm),
– mobile phase: a filtered and degassed mixture of
Gugulipid Tablets 45 volumes of acetonitrile and 55 volumes of water,
– flow rate. 2 ml per minute,
Gugulipid Tablets contain not less than 90.0 per cent and not – spectrophotometer set at 242 nm,
more than 110.0 per cent of the stated amount of gugulsterones – a 20 µl loop injector.
(Z and E). The tablets may be coated.
Inject the reference solution. The test is not valid unless the
Identification relative retention times are about 0.69 for gugulsterone E and
1.0 for gugulsterone Z and the relative standard deviation for
Extract a quantity of the powdered tablets containing about replicate injections is not more than 2.0 per cent.
20 mg of gugulsterones (Z and E) with two quantities, each of
15 ml, of ethyl acetate, combine the extracts, filter and Inject the test solution and the reference solution. Calculate
evaporate to dryness. The residue complies with the following the contents of gugulsterones (Z and E) in the tablets.
tests. Storage. Store protected from light at a temperature not
A. When examined in the range 230 nm to 360 nm (2.4.7), exceeding 30º.
a 0.005 per cent w/v solution in chloroform shows absorption Labelling. The label states the strength in terms of the
maxima at about 245 nm and 327 nm; absorbance at about equivalent amount of gugulsterones (Z and E).
245 nm, about 0.87 and at about 327 nm, about 0.52.
B. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G.
Mobile phase. A mixture of 3 volumes of light petroleum
(60º to 80º) and 1 volume of ethyl acetate.
Test solution. A 0.25 per cent w/v solution of the substance
under examination in ethanol (95 per cent).
Reference solution. A 0.25 per cent w/v solution of gugulipid
RS in ethanol (95 per cent).

1404
IP 2007 HARIDRA

Haridra Tests
Haldi; Turmeric; Curcuma Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 12 per cent by
Method I.
Ash (2.3.19). Not more than 10 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Water (2.3.43). Not more than 12.0 per cent, determined on
0.2 g.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux about 1 g of the coarsely powdered
Haridra consists of the dried rhizomes of Curcuma longa Linn. substance under examination with 50 ml of methanol on a
(Fam. Zingiberaceae). water bath for 15 minutes cool and filter. Reflux the residue
Haridra contains not less than 1.5 per cent of curcumin, further with 5 x 25 ml of methanol, cool and filter. Combine all
calculated on the dried basis. the filtrates and concentrate to 100.0 ml.
Description. Externally yellowish to yellowish brown with Reference solution. A 0.01 per cent w/v solution of curcumin
root scars and annulations. Odour, aromatic; taste, warmly RS in methanol.
aromatic and bitter. Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with
Identification silicagel consisting of porous spherical particles with
A. Macroscopic — Rhizome oblong, conical or cylindrical to chemically bonded nitrile group,
elongate, finger-like; internally orange yellow. Texture hard – mobile phase: 35 volumes of tetrahydrofuran and
and heavy; fracture short. 65 volumes of a buffer solution prepared by dissolving
10 g of citric acid in 1000 ml of water, adjusting the pH
B. Microscopic — Ground tissue of parenchyma cells; cells to 3.0 with dilute ammonia solution,
filled with gelatinized starch grains and yellow pigment. – flow rate. 1.2 ml per minute,
Fibrovascular bundles and oil cells scattered through out – spectrophotometer set at 430 nm,
ground tissue. – a 20 µl loop injector.
C. Determine by thin-layer chromatography (2.4.17), coating Inject the reference solution. The test is not valid unless the
the1 plate with silica gel GF 254. relative standard deviation for the replicate injections is not
Mobile phase. A mixture of 94 volumes of chloroform, more than 2.0 per cent.
5 volumes of ethanol and 1 volume glacial acetic acid. Inject the test solution and reference solution.
Test solution. Extract 1 g of the coarsely powdered substance Calculate the content of curcumin.
under examination with 5 ml methanol for 10 minutes with
slight warming. Filter and use the filtrate. Storage. Store protected from moisture.

Reference solution. Reflux 1 g of coarsely powdered haridra


RS with 5 ml methanol for 15 minutes, cool and filter.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cms above the line of
application. Dry the plate in air and examine in daylight and
ultra-violet light 365 nm. The chromatographic profile of the
test solution is similar to that of the reference solution.

1405
HARITAKI IP 2007

Haritaki Reference solution. To 0.1 g of the haritaki RS, add 50-75 ml


of methanol and reflux for 15 minutes, cool and filter. Reflux
Harad; Chebulic myrobalan; Terminalia the residue further for two times with 75 ml of methanol, cool
and filter. Combine all the filtrates and concentrate under
vacuum to 10 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cms above the line of
application. Dry the plate in air, spray with solution of 10 per
cent w/v ferric chloride solution in water. Examine the plate
in day light. The chromatographic profile of the test solution
is similar to that of the reference solution.
D. In the Assay, the chromatogram obtained with the test
solution corresponds to the chromatogram obtained with
reference solution (a).

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Haritaki consists of pericarp of the dried fruit of Terminalia
Ethanol-soluble extractive (2.6.2). Not less than 35.0 per cent.
chebula Retz. (Fam. Combretaceae).
Water-soluble extractive (2.6.3). Not less than 50 per cent by
Haritaki contains not less than 5 per cent and not more than
Method I.
12.5 per cnet of chebulinic acid, calculated on the dried basis.
Ash (2.3.19). Not more than 6.0 per cent.
Description. It has a shine on its external part and has
longitudinal ridges. The colour varies from yellowish brown Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
to light black. It has a astringent taste and is also slightly Heavy metals (2.3.13). 1.0 g complies with the limit test for
bitter heavy metals, Method B (20 ppm)
Identification Loss on drying (2.4.19). Not more than 12 per cent, determined
on 5 g by drying in an oven at 105º.
Test C may be omitted if tests A, B and D are carried out and
test D may be omitted if tests A, B and C are carried out. Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
A. Macroscopic — The fruit is 2 to 3 cm in length and 1 to
2 cm in diameter with hard stony appearance. Externally it is Assay. Determine by liquid chromatography (2.4.14).
shining and is adorned with longitudinal ridges. Color of the Test solution. Weigh 0.5 g of coarsely powdered sample, add
fruit rind varies from yellowish brown, uniform brown to light 50 ml of water, sonicate for 3 minutes and heat on a boiling
black. Internally the fruit is light yellow. water bath for 15 minutes, cool and dilute to 100.0 ml with
B. Microscopic — Epicarp has thick walls covered with cuticle. water and filter. Dilute 10.0 ml of the solution to 25.0 ml with
The mesocarp has many stone cells of various sizes and water.
shapes which forms a reticulum. Large quantity of tannin is Reference solution (a). Weigh 0.5 g of haritaki RS, add 50 ml
present in the mesocarp. Simple starch granules are present in of water, sonicate for 3 minutes and heat on a boiling water
plenty. bath for 15 minutes, cool and dilute to 100.0 ml with water and
C. Determine by thin-layer chromatography (2.4.17), coating filter. Dilute 10.0 ml of the solution to 25.0 ml with water.
the plate with silica gel 60. Reference solution (b). A 0.01 per cent w/v solution of
Mobile phase. A mixture of 35 volumes of toulene, 50 volumes chebulagic acid RS in water.
of acetone, 15 volumes of glacial acetic acid and 5 volumes Reference solution (c). A 0.01 per cent w/v solution of
of formic acid. chebulinic acid RS in water.
Test solution. To 1 g of the coarsely powdered substance
Chromatographic system
being examined, add 50-75 ml of methanol and reflux for
– a stainless steel column 25 cm x 4.6 mm packed with
15 minutes, cool and filter. Reflux the residue further for two
octadecylsilane bonded to porous silica (5 µm).
times with 75 ml of methanol, cool and filter. Combined all the
– mobile phase: filtered and degassed gradient mixtures
filtrates and concentrate under vacuum to 100 ml.

1406
IP 2007 ISPAGHULA HUSK

of acetonitrile and a buffer solution pH 2.5 prepared by Acid value (2.3.23). Not more than 2.0.
dissolving 0.136 g of potassium di-hydrogen Hydroxyl value (2.3.27). 154 to 162.
orthophosphate in 500 ml of water, add 0.5 ml of
orthophosphoric acid and make upto 1000 ml with Iodine value (2.3.28). Not more than 5.0.
water, Peroxide value (2.3.35). Not more than 5.0.
– flow rate. 1.5 ml per min.,
– spectrophotometer set at 270 nm, Saponification value (2.3.37). 176 to 182.
– a 20 µl loop injector. Storage. Store at a temperature not exceeding 30º. Avoid
Time Buffer solution Acetonitrile exposure to excessive heat.
(in min.) (per cent v/v) (per cent v/v)
0 95 5
18 65 35 Ispaghula Husk
25 45 55 Isapgol Husk; Plantago
28 45 55
35 95 5
Inject the reference solution (b) and (c). The relative standard
deviation for the replicate injections is not more than 2.0 per
cent.
Inject the test solution and reference solution (a).
Calculate the content of Chebulagic acid and Chebulinic
acid.
Storage. Store protected from heat, moisture and against attack
by insects and rodents.

Hydrogenated Castor Oil


Castor Wax; Opalwax
Hydrogenated Castor Oil is refined, bleached, hydrogenated
and deodorised castor oil. It consists mainly of the triglyceride Ispaghula Husk consists of the epidermis and collapsed
of hydroxystearic acid. adjacent layers removed from the dried ripe seeds of Plantago
ovata Forssk.
Description. A white to yellow powder of uniform consistency
and texture. It may have a hard, waxy consistency. Description. Pale buff, brittle flakes, more or less lanceolate,
up to 2 mm long and 1 mm wide at the centre, much broken into
Tests smaller fragments; many of the flakes having a small, brownish,
oval spot, about 0.8 to 1.0 mm long, in the centre; the material
Melting range (2.4.21). 85 º to 88º, determined by Method II. swells rapidly in water, forming a stiff mucilage.
Free fatty acids. Weigh accurately about 20 g, melt on a water-
bath, add 75 ml of hot ethanol (95 per cent), previously Identification
neutralised to phenolphthalein solution with 0.1 M sodium When mounted in cresol and examined under a microscope,
hydroxide, swirl, add 1 ml of phenolphthalein solution and the particles are found to be transparent and angular, the edges
titrate with 0.1 M sodium hydroxide, swirling vigorously until straight or curved and sometimes rolled. They are composed
the solution remains faintly pink after being shaken for of polygonal prismatic cells with four to six straight or slightly
60 seconds; not more than 11.0 ml of 0.1 M sodium hydroxide curved walls; the cells vary in size in different parts of the
is required. seed coat, from about 25 mm to 60 mm at the summit of the
Heavy metals (2.3.13). 2.0 g complies with the limit test for seed, that is, near and over the brown spot, to 25 mm to
heavy metals, Method B (10 ppm). 100 mm for the remainder of the epidermis except at the edges
of the seed, where the cells are smaller, about 45 mm to 70 mm.
Acetyl value (2.3.22). Not less than 143.
When mounted in ethanol (95 per cent) and irrigated with

1407
KALMEGH IP 2007

water, the mucilage in the outer part of the epidermal cells Identification
swells rapidly and goes into solution, while the two inner
layers of mucilage are more resistant and swell to form rounded A. Macroscopic — Mixture of crisp, dark green-coloured
papillae. When mounted in 0.005 M iodine, occasional simple broken leaves and quadrangular stems; leaves brittle. Stem
and two- to four-compound starch granules, about 2 mm to fracture short, fibrous.
10 mm, can be seen in some of the cells. Occasional fragments B. Microscopic — Stems quadrangular with collenchyma
of thick-walled, reddish brown endosperm, cells with pitted strands at angles and on side; small acicular crystals of calcium
walls and elongated fragments of grey embryo may be present. oxalate present in pith and cortex. Trichomes 1-3 celled,
Swelling power. Transfer 1 g to a 100-ml stoppered cylinder glandular hair disc-shaped and multicellular.
containing 90 ml of water, shake well for 30 seconds and allow C. Determine by thin-layer chromatography (2.4.17), coating
to stand 24 hours, shaking gently on three occasions during the plate with silica gel G.
this period. Add sufficient water to produce 100 ml, mix gently
for 30 seconds, avoiding the entrapment of air, allow to stand Mobile phase. A mixture of 7 volumes of chloroform and
for 5 hours and measure the volume of mucilage. Repeat the 1 volume of methanol.
determination three times. The average of four determinations Test solution. Reflux 1 g of coarsely powdered substance under
is not less than 40 ml. examination with 50 ml methanol for 15 minutes, cool and
Ash (2.3.19). Not more than 4.5 per cent, determined on 1 g. filter. Reflux the residue further with 2 × 50 ml of methanol,
cool and filter. Combine all the filtrates and concentrate to
Acid-insoluble ash (2.3.19). Not more than 0.45 per cent. 10 ml.
Loss on drying (2.4.19). Not more than 12.0 per cent, Reference solution. Reflux 0.5 g of kalmegh RS with 50 ml
determined on 0.5 g by drying in an oven at 105º for 5 hours. methanol for 15 minutes, cool and filter. Reflux the residue
Storage. Store protected from moisture and from attack by further with 2 × 50 ml of methanol, cool and filter. Combine all
insects and rodents. the filtrates and concentrate to 5 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cms. Dry the plate in air
and spray with methanolic sulphuric acid (20 per cent). Heat
Kalmegh at 120º for 5-10 minutes and examine the plate in day light. The
Andrographis chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent.
Water-soluble extractive (2.6.3). Not less than 12.0 per cent
by Method I.
Ash (2.3.19). Not more than 15 per cent.
Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 1050.

Kalmegh consists of the dried aerial parts, mainly stems and Microbial contamination (2.2.9). Complies with the microbial
leaves, of Andrographis paniculata Nees. (Fam. contamination tests.
Acanthaceae). Assay. Determine by liquid chromatography (2.4.14).
Kalmegh contains not less than 1.0 per cent of Test solution. Reflux about 2.5 g of the coarsely powdered
andrographolide, calculated on the dried basis. substance under examination with 50 ml of methanol on a
Description.Taste, intensely bitter. water bath for 15 minutes, cool and filter. Reflux the residue

1408
IP 2007 KUNDURU

further with methanol till the last extract turns colorless, cool aromatic.
and filter. Combine all the filtrates and concentrate to 50.0 ml. B. Determine by thin-layer chromatography (2.4.17), coating
Reference solution. A 0.01 per cent w/v solution of the plate with silica gel GF 254.
andrographolide RS in methanol.
Mobile phase. A mixture of 7 volumes of hexane and 3 volumes
Chromatographic system of ethyl acetate.
– a stainless steel column 25 cm x 4.6 mm packed with
Test solution. Reflux 1 g of coarsely powdered substance under
octadecylsilane bonded to porous silica (5 µm),
examination with 50 ml methanol on a boiling water-bath for
– mobile phase: 65 volumes of methanol and 35 volumes
30 minutes, cool and filter. Evaporate the filtrate to dryness
of water,
and dissolve the residue in 5 ml of methanol.
– flow rate. 1 ml per minute,
– spectrophotometer set at 223 nm, Reference solution. Reflux 1 g of coarsely powdered kunduru
– a 20 µl loop injector. RS with 50 ml methanol on a boiling water-bath for 30 minutes,
cool and filter. Evaporate the filtrate to dryness and dissolve
Inject the reference solution. The test is not valid unless the the residue in 5 ml of methanol.
relative standard deviation for the replicate injections is not
more than 2.0 per cent. Apply to the plate 10µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cms above the line of
Inject the test solution and reference solution.
application. Dry the plate in air and spray with methanolic
Calculate the content of andrographolide. sulphuric acid (10 per cent) and heat at 110º for 5-10 minutes
and examine the plate in day light. The chromatographic profile
Storage. Store protected from heat, moisture and against attack
of the test solution is similar to that of the reference solution.
by insects and rodents.
Tests
Kunduru Foreign organic matter (2.6.1). Not more than 2.0 per cent.

Sallaki Gum; Gum of Boswellia Serrata Ethanol-soluble extractive (2.6.2). Not less than 35 per cent.
Ash (2.3.19). Not more than 10.0 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Water (2.3.43). Not more than 12.0 per cent, determined on
0.2 g.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux about 2 g of the coarsely powdered
substance under examination with 50 ml of methanol on a
water bath for 15 minutes, cool and filter. Reflux the residue
further with methanol till the last extract turns colorless, cool
Kunduru is the gum-resin from Boswellia serrata Roxb. (Fam.
and filter. Combine all the filtrates and concentrate to 100.0 ml.
Burseraceae).
Reference solution (a). A 0.01 per cent w/v solution of
Kunduru contains not less than 1.0 per cent of total 11-keto-
11-keto-β-boswellic acid RS in methanol.
β-boswellic acid and acetyl-11-keto-β-boswellic acid,
calculated on the dried basis. Reference solution (b). A 0.05 per cent w/v solution of
Description. Translucent, brittle, whitish yellow substance, acetyl-11-keto-β-boswellic acid RS in methanol.
in roundish, club-shaped, pear-shaped, or irregular tears. Chromatographic system
– a stainless steel column 25 cm x 4.6 mm packed with
Identification octadecylsilane bonded to porous silica (5 µm),
A. Macroscopic — Fracture dull. Slightly sticky to touch; – mobile phase: 90 volumes of methanol and 10 volumes
odour, balsamic; taste slightly mucilaginous, bitter and of a mixture containing 5 ml of acetonitrile and 95 ml of

1409
KUTKI IP 2007

water, adjusting the pH to 2.8 with dilute C. Determine by thin-layer chromatography (2.4.17), coating
orthophosphoric acid, the plate with silica gel G.
– flow rate. 1.5 ml per minute, Mobile phase. A mixture of 7.5 volumes of ethyl acetate,
– spectrophotometer set at 247 nm, 2.2 volumes of methanol and 0.1 volume glacial acetic acid.
– a 20 µl loop injector.
Test solution. Reflux 5 g of coarsely powdered substance under
Inject the reference solution (a) and (b). The test is not valid
examination with 50 ml of methanol for 15 minutes, cool and
unless the relative retention times are about 0.65 for 11-keto-
filter. Reflux the residue further with 50 ml of methanol, cool
β-boswellic acid and 1.0 for acetyl-11-keto-β-boswellic acid
and filter. Combine both the filtrates and concentrate under
and the relative standard deviation for the replicate injections
vacuum to dryness. Extract the dried residue with 10 ml of
is not more than 2.0 per cent.
methanol at 50° for 10 minutes, filter the solution and use the
Inject the test solution. filtrate for analysis.
Calculate the sum of the contents of 11-keto-β-boswellic acid Reference solution. Reflux 5 g of kutki RS with 50 ml of
and acetyl-11-keto-β-boswellic acid. methanol for 15 minutes, cool and filter. Reflux the residue
Storage. Store protected from heat, moisture and against attack further with 50 ml of methanol, cool and filter. Combine both
by insects and rodents. the filtrates and concentrate under vacuum to dryness. Extract
the dried residue with 10 ml of methanol at 50° for 10 minutes,
filter the solution and use the filtrate for analysis.
Kutki Apply to the plate 20 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
Picrorhiza spray with anisaldehyde sulphuric acid. The chromatographic
profile of the test solution is similar to that of the reference
solution.

Tests
Foreign organic matter (2.6.1). Not more than 2 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 15 per cent.
Water-soluble extractive (2.6.3). Not less than 25 per cent by
method I.
Ash (2.3.19). Not more than 6 per cent.
Acid-insoluble ash (2.3.19). Not more than 1 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Kutki consists of dried roots of Picrorhiza kurroa Royle ex Loss on drying (2.4.19). Not more than 5 per cent, determined
Benth.(Fam. Scrophulariaceae) on 5 g by drying in an oven at 105º.
Kutki contains not less than 5 per cent of kutkin, calculated Microbial contamination (2.2.9). Complies with the microbial
on the dried basis. contamination tests.
Description. Rhizomes are sub cylindrical, straight or slightly Assay. Determine by liquid chromatography (2.4.14).
curved, externally greyish with wrinkled surfaces, circular scars Test solution. Dissolve 100 mg of the substance under
of roots and bud scales, with cork exposed at places. examination in 25.0 ml of methanol, filter.
Identification Reference solution. A 0.1 per cent w/v solution of kutkin RS
in methanol.
A. Macroscopic — 3-6 cm long and about 1 cm thick, sub-
cylindrical, straight, grayish brown, wrinkled roots. Odour is Chromatographic system
pleasant and tastes bitter. – a stainless steel column 15 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm),
B. Microscopic — There is well-developed periderm. About – mobile phase: 83 volumes of 1 per cent v/v
7-10 layers of cork cells are seen. Cells of phelloderm loosely orthophosphoric acid in water and 17 volumes of
arranged. acetonitrile,

1410
IP 2007 LASUNA

– flow rate. 1 ml per minute, B. Microscopic — The protective leaf contains an epidermis
– spectrophotometer set at 280 nm, enclosing a mesophyll free from chlorophyll. The outer
– a 20 µl loop injector. epidermis consists of lignified sclereid cells of thick, pitted
Inject the reference solution. The relative standard deviation walls, elongated, covered with thin cuticle. The cortical cells
for the replicate injections is not more than 2.0 per cent. are thick walled, non lignified, tending to collapse on maturity,
isodiametric and contain purple pigments. The vascular
Inject the test solution and reference solution. bundles consist of lignified spiral and annular vessels. The
Calculate the content of kutkin. storage leaves show an outer epidermis of thin, delicate cells
of variable shape. Stomata are present on the outer epidermis
Storage. Store protected from heat, moisture and against attack
only at extreme tip near the base of the foliage leaves.The
by insects and rodents.
mesophyll consists of swollen storage parenchyma cells filled
with fine granular reserve material.
Lasuna C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel F254.
Garlic, Allium sativum bulb
Mobile phase. A mixture of 3 volumes of butyl alcohol,
1 volume of n-propyl alcohol, 1 volume of glacial acetic acid
and 1 volume of water.
Test solution. Reflux 1 g of substance under examination with
20 ml of methanol (50 per cent) for 10 minutes, cool and filter.
Reflux the residue with another 20 ml of methanol (50 per
cent), cool and filter. Combine all the filtrates and concentrate
under vacuum to 5 ml.
Reference solution. Reflux 1 g of lasuna RS with 20 ml of
methanol (50 per cent) for 10 minutes, cool and filter. Reflux
the residue with another 20 ml of methanol (50 per cent), cool
and filter. Combine all the filtrates and concentrate under
vacuum to 5 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
Lasuna consists of the fresh or dried compound bulbs of 2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
Allium sativum Linn. (Fam. Liliaceae). spray with 0.2 per cent w/v solution of ninhydrin in mixture of
95 volumes of butyl alcohol and 5 volumes of 2 M acetic
Lasuna contains not less than 0.2 per cent of alliin, calculated acid. Heat at 100º to 105º for about 10 minutes and examine the
on the dried basis. plate in day light. The chromatographic profile of the test
Description. Bulbs made up of cloves and is wrapped in a solution is similar to that of the reference solution.
white papery sheath with pungent taste and odour
D. Transfer about 10 g of garlic bulbs that have been cut into
Identification small pieces to a beaker, add 10 ml of 1 M sodium hydroxide
and 10 ml of water, heat the beaker in a boiling water for
A. Macroscopic — Each bulb has several cloves which are 10 minutes, cool and filter. To 2 ml of this filtrate add few drops
arranged in concentric rings and enclosed in a shining white of freshly prepared solution of sodium nitroprusside.
or pinkish papery envelope. The cloves are attached to a flat, Appearance of a red or orange red colour indicates the presence
circular hard disc with numerous thin wiry roots from its of sulphur containing compounds in the sample.
underside and short, cylindrical out growth from the upper
surface. Each clove is ovoid and further covered by papery Tests
sheath with a tail like structure at one and opposite to its
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
attachment. The cloves in the outer ring are loose and white in
colour where as cloves in the inner ring are adherent and pale Ash (2.3.19). Not more than 5 per cent.
pinkish in colour. Each clove covered with a white scale leaf Acid-insoluble ash (2.3.19). Not more than 1.0 per cent.
and a pinkish white epidermis, easily separated from the solid
portion. In the middle of the bulb a hollow, cylindrical, linear Heavy metals (2.3.13). 1.0 g complies with the limit test for
remnant of the scape is seen. heavy metals, Method B (20 ppm).

1411
MALT EXTRACT IP 2007

Loss on drying (2.4.19). Not more than 65.0 per cent for fresh Lipase. To 95 ml of water add 6.5 ml of triacetin and 0.2 ml of
bulbs, determined on 5 g by drying in an oven at 105º. a 0.1 per cent w/v solution of bromocresol purple, neutralise
Microbial contamination (2.2.9). Complies with the microbial with 0.5 M sodium hydroxide and add sufficient water to
contamination tests. produce 110 ml. Place 50 ml of this solution in each of two
large test-tubes (20 cm × 3 cm) A and B kept in a water-bath at
Assay. Determine by liquid chromatography (2.4.14). 30º ± 1º. Insert in each tube a rubber stopper having two
Test solution. Weigh accurately about 2 g of the freshly peeled holes, one for the tip of a burette and the other for a short
substance under examination. Add 15 ml of hot water and glass tube through which passes a thread operating a glass
grind in a porcelain mortor and filter. Grind the residue further stirring coil. Stir the contents of the tube until they attain the
with 2 × 15 ml of hot water and filter. Combine all the filtrates temperature of the water-bath. Prepare a solution of 5.0 g of
and make up to volume 50 ml with distilled water. the substance under examination in 10 ml of water. To tube A
add 1 ml of this solution; to tube B add 1 ml of this solution
Reference solution. A 0.004 per cent w/v solution of alliin RS after previous boiling. Adjust and maintain the pH of the two
in water. tubes to between 6.2 and 6.4 by the dropwise addition of 0.05
Chromatographic system M sodium hydroxide, stirring frequently. After 6 hours, the
– a stainless steel column 25 cm × 4.6 mm packed with difference between the amounts of 0.05 M sodium hydroxide
octadecylsilane bonded to porous silica (5 µm), added to the tubes is not more than 1.0 ml.
– mobile phase: 0.1 per cent v/v phosphoric acid prepared Assay. Weigh accurately about 5.0 g into a 200-ml long-necked
by diluting 1ml of phosphoric acid to 1000 ml with water, flask and carry out the determination of nitrogen, Method A
– flow rate. 0.5 ml per minute, (2.3.30), using 0.05 M sulphuric acid instead of 0.1 M
– spectrophotometer set at 210 nm, sulphuric acid.
– a 20 µl loop injector.
1 ml of 0.05 M sulphuric acid is equivalent to 0.001401 g of N
Inject the reference solution. The test is not valid unless the and multiply the result by 6.25 to obtain the protein content.
relative standard deviation for the replicate injections is not
more than 2.0 per cent. Storage. Store protected from moisture.
Inject the test and reference solution.
Calculate the content of alliin.
Mandukaparni
Storage. Store protected from heat, moisture and against attack
by insects and rodents. Centella, Gotu Kola

Malt Extract
Malt Extract is a product obtained by extracting malted grains
of cereals (barley, cholam or wheat) with water at a suitable
temperature and evaporation of the strained liquid until a
viscous product is obtained. It may be mixed with 10 per cent
by weight of Glycerin.
Malt Extract contains nitrogen equivalent to not less than
4.0 per cent w/w of protein.
Description. A sweet, viscous, light brown liquid; odour,
pleasant and characteristic.

Tests
Mandukaparni consists of the dried aerial parts of Centella
Refractive index (2.4.27). 1.489 to 1.498, determined at 20º.
asiatica (Linn.) Urban. (Fam. Umbelliferae).
Arsenic (2.3.10). Dissolve 10.0 g in 10 ml of water, add 10 ml
Mandukaparni contains not less than 0.5 per cent of
of brominated hydrochloric acid, allow to stand for 5 minutes
asiaticoside, calculated on the dried basis.
and remove the excess of bromine with a few drops of stannous
chloride solution AsT. The resulting solution complies with Description. A green to greenish yellow in colour, taste,
the limit test for arsenic (1 ppm). slightly bitter and sweet.

1412
IP 2007 MANJISTHA

Identification water bath for 15 minutes, cool and filter. Reflux the residue
further with methanol till the last extract turns colorless, cool
A. Macroscopic — A slender trailing herb with rooted nodes and filter. Combine all the filtrates and concentrate to 100.0 ml.
and internodes. Leaves with elongated petioles and sheathing
leaf bases; lamina reniform with crenate margin. Reference solution. A 0.1 per cent w/v solution of
asiaticoside RS in methanol.
B. Microscopic — Animocytic stomata on both surfaces,
more on lower surface; petiole epidermis has calcium oxalate Chromatographic system
prisms; vascular bundles seven arranged in ‘U’ shape, – a stainless steel column 25 cm x 4.6 mm packed with
parenchyma cells contain simple and compound starch octylsilane bonded to porous silica (10 µm),
granules. – mobile phase: 25 volumes of acetonitrile and 75 volumes
of water,
C. Determine by thin-layer chromatography (2.4.17), coating – flow rate. 1.5 ml per minute,
the plate with silica gel G.. – spectrophotometer set at 210 nm,
Mobile phase. A mixture of 60 volumes of chloroform, – a 20 µl loop injector.
32 volumes of glacial acetic acid, 12 volumes of methanol Inject the reference solution. The test is not valid unless the
and 8 volumes of water. relative standard deviation for the replicate injections is not
Test solution. Reflux 2 g of coarsely powdered substance under more than 2.0 per cent.
examination with 50 ml methanol for 15 minutes, cool and Inject the test solution and reference solution.
filter. Reflux the residue further with 2 × 50 ml of methanol,
cool and filter. Combine all the filtrates and concentrate to Calculate the content of asiaticoside.
10 ml under reduced pressure. Storage. Store protected from heat, moisture and against attack
Reference solution. Reflux 1 g of mandukaparni RS with by insects and rodents.
50 ml methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 × 50 ml of methanol, cool and filter. Combine all
the filtrates and concentrate to 5 ml. Manjistha
Apply to the plate 10 µl of each solution as bands 10 mm by Indian Madder; Rubia Cordifolia
2 mm. Allow the mobile phase to rise 8 cms above the line of
application. Dry the plate in air, spray with anisaldehyde
solution. Heat at 100º for 5-10 minutes and examine the plate in
day light. The chromatographic profile of the test solution is
similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent.
Water-soluble extractive (2.6.3). Not less than 15.0 per cent
by Method I.
Ash (2.3.19). Not more than 24 per cent.
Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Manjistha consists of dried stem of Rubia cordifolia
Heavy metals (2.3.13). 1.0 g complies with the limit test for Linn.sensu Hook.f. (Fam. Rubiaceae).
heavy metals, Method B (20 ppm).
Manjistha contains not less than 0.02 per cent of rubiadin,
Loss on drying (2.4.19). Not more than 12.0 per cent, calculated on the dried basis.
determined on 5 g by drying in an oven at 1050.
Description. A cylindrical, slightly flattened, wiry pieces of
Microbial contamination (2.2.9). Complies with the microbial brown to purple coloured root with mild bitter taste.
contamination tests.
Assay. Determine by liquid chromatography (2.4.14). Identification
Test solution. Reflux about 3 g of the coarsely powdered A. Microscopic — Exfoliating cork, consisting of 4-12 or more
substance under examination with 50 ml of methanol on a layered radially arranged, thin walled cells.

1413
MARICHA IP 2007

B. Macroscopic — Stem, slender, cylindrical, wiry and about octadecylsilane bonded to porous silica (5 µm),
0.5 cm thick. It is brown to purple coloured and with – mobile phase: filtered and degassed gradient mixtures
longitudinal cracks. of acetonitrile and a buffer solution pH 2.5 prepared by
C. Determine by thin-layer chromatography (2.4.17), coating dissolving 0.136 g of potassium di-hydrogen
the plate with silica gel G.. orthophosphate in 900 ml of water, add 0.5 ml of
orthophosphoric acid and dilute to 1000 ml with water,
Mobile phase. A mixture of 7 volumes of toluene, 25 volumes – flow rate. 1.5 ml per minute,
of ethyl acetate and 0.5 volumes glacial acetic acid. – spectrophotometer set at 278 nm,
Test solution. To 4 g of coarsely powdered substance under – a 20 µl loop injector.
examination, add 100 ml of methanol, reflux for 15 minutes, Time Buffer solution Acetonitrile
cool and filter. Reflux the residue further for two times with 50 (in min.) (per cent v/v) ( per cent v/v)
ml methanol, cool and filter. Combine all the filtrates and 0 65 35
concentrate under vacuum to 100 ml. 5 50 50
Reference solution. To 1 g of coarsely powdered manjistha, 15 20 80
add 100 ml of methanol, reflux for 15 minutes, cool and filter. 30 20 80
Reflux the residue further for two times with 50 ml methanol, 35 50 50
cool and filter. Combine all the filtrates and concentrate under 40 65 35
vacuum to 25 ml.
Inject the reference solution. The relative standard deviation
Apply to the plate 20 µl of each solution as bands 10 mm by for the replicate injections is not more than 2.0 per cent.
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air Inject the test solution and reference solution.
and examine in the 254 nm, 366 nm and spray the plate with
anisaldehyde sulphuric acid. The chromatographic profile of Calculate the content of rubiadin.
the test solution is similar to that of the reference solution. Storage. Store protected from heat, moisture and against attack
by insects and rodents.
Tests
Foreign organic matter (2.6.1). Not more than 2 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 4 per cent.
Maricha
Water-soluble extractive (2.6.3). Not less than 20 per cent by Black pepper; pepper; Piper nigrum
method I.
Ash (2.3.19). Not more than 10 per cent.
Acid-insoluble ash (2.3.19). Not more than 0.5 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 5 per cent, determined
on 5 g by drying in an oven at 105º.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh 4 g of coarsely powdered substance
under examination, add 100 ml of methanol, reflux on a water-
bath 15 minutes, cool and filter. Reflux the residue 2 x 50 ml Maricha consists of the unripe fruits of Piper nigrum Linn.
methanol, cool and filter. Combine all the filtrates and (Fam. Piperaceae).
concentrate under vacuum to 100.0 ml.
Maricha contains not less than 2.5 per cent w/w of piperine,
Reference solution. A 0.002 per cent w/v solution of rubiadin calculated on the dried basis.
RS in methanol.
Description. Fruits are globular or oblong. They have a blackish
Chromatographic system brown cover, with raised reticulated wrinkles. The odor is
– a stainless steel column 25 cm x 4.6 mm packed with aromatic and the taste is strong and pungent.

1414
IP 2007 MENTHA OIL

Identification Test solution. Weigh 2 g of coarsely powdered substance


under examination, add 50 ml of methanol, sonicate for
A. Macroscopic — The fruits are globular or oblong, 4-6 mm 3 minutes and heat on a boiling water bath for 15 minutes, cool
in diameter. The outer cover is blackish brown, with raised
reticulated wrinkles. One seeded, seeds are white and hollow. and dilute to 100.0 ml with methanol and filter. Dilute further if
necessary.
B. Microscopic — The fruit has a well differentiated pericarp,
testa and perisperm. Isolated, tangentially elongated oil cells Reference solution. A 0.01 per cent w/v solution of
are in the outer region of the mesocarp. Endocarp has beaker piperine RS in methanol.
shaped stone cells and numerous polyhedral masses of starch Chromatographic system
grains. Testa has a single layer of yellow colored cells. – a stainless steel column 25 cm x 4.6 mm packed with
C. Determine by thin-layer chromatography (2.4.17), coating octadecylsilane bonded to porous silica (5 µm),
the plate with silica gel GF254. – mobile phase. filtered and degassed gradient mixtures
of acetonitrile and a buffer solution pH 2.5 prepared by
Mobile phase. A mixture of 60 volumes of benzene, 30 volumes dissolving 0.136 g of potassium di-hydrogen
of ethyl acetate and 10 volumes of diethyl ether. orthophosphate in 500 ml of water, add 0.5 ml of
Test solution. To 2 g of the coarsely powdered substance orthophosphoric acid and dilute to 1000 ml with water,
under examination, add 50 ml of methanol and reflux for – flow rate. 1.5 ml per minute,
15 minutes, cool and filter. Reflux the residue further for two – spectrophotometer set at 345 nm,
times with 75 ml of methanol, cool and filter. Combine all the – a 20 µl loop injector.
filtrates and concentrate under vacuum to 50 ml. Time Buffer solution. Acetonitrile
(in min.) (per cent v/v) (per centv/v)
Reference solution. To 2 g of the maricha RS, add 50 ml of
methanol and reflux for 15 minutes, cool and filter. Reflux the 0 95 5
residue further for two times with 75 ml of methanol, cool and 18 55 45
filter. Combine all the filtrates and concentrate under vacuum 25 20 80
to 50 ml.
Inject the reference solution. The relative standard deviation
Apply to the plate 10 µl of each solution as bands 10 mm by for the replicate injections is not more than 2.0 per cent.
2 mm. Allow the mobile phase to rise 8 cms. Dry the plate in air
and examine in ultraviolet light at 254 nm. The chromatogram Inject the test solution and reference solution. The relative
obtained with test solution corresponds to the band in the retention time of piperine is 1.
chromatogram obtained with reference solution. Spray with Calculate the content of piperine.
vanilin sulphuric acid reagent. Heat at 100º for 5-10 minutes
Storage. Store protected from moisture and against attack by
and examine the plate in day light. The chromatographic profile
insects and rodents.
of the test solution is similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent. Mentha Oil
Ethanol-soluble extractive (2.6.2). Not less than 6.0 per cent. Mentha
Water-soluble extractive (2.6.3). Not less than 6.0 per cent by Mentha Oil is the volatile oil distilled with steam from various
method I. species of Mentha (Fam. Labiatae) and rectified if necessary.
Ash (2.3.19). Not more than 7.0 per cent. Mentha Oil contains not less than 50.0 per cent w/w of total
menthol, C10H20O.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Description. A colourless or yellowish, clear liquid; odour,
Heavy metals (2.3.13). 1.0 g complies with the limit test for
characteristic and pleasant.
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent, Tests
determined on 5 g by drying in an oven at 1050.
Acidity or alkalinity. A solution of 1 ml in 3.5 ml of ethanol
Microbial contamination (2.2.9). Complies with the microbial (70 per cent) is neutral to litmus.
contamination tests.
Weight per ml (2.4.29). 0.892 g to 0.910 g.
Assay. Determine by liquid chromatography (2.4.14).
Specific optical rotation (2.4.22). –18.0º to –33.0º.

1415
OPIUM IP 2007

Assay. Place about 10.0 g in an acetylation flask, add 10 ml of Opium contains not less than 10.0 per cent of morphine,
acetic anhydride and 1 g of anhydrous sodium acetate, attach C17H 19NO3, and not less than 2.0 per cent of codeine,
a reflux condenser and boil for 2 hours. Cool, add 30 ml of C18H21NO3, both calculated on the dried basis.
water and warm on a water-bath for 15 minutes with occasional Description. Masses of various sizes which tend to be soft
shaking. Transfer the contents of the flask to a separating and shiny and, after drying, hard and brittle; usually in
funnel, reject the water layer and wash the remaining oil with somewhat irregularly shaped masses (natural opium) or
water until the last washing no longer shows acid reaction. moulded into masses of more uniform size and shape
Dry the resulting oil by shaking with 2 g of anhydrous sodium (manipulated opium); colour, blackish brown; odour, strong
sulphate, allow to stand for 30 minutes and filter through a and characteristic.
dry filter paper. Weigh accurately about 1.5 g of the dry
acetylated oil, add 3 ml of ethanol (95 per cent) and 0.1 ml of Identification
phenolphthalein solution and dropwise, 0.5 M ethanolic
potassium hydroxide until the solution acquires a faint pink Strip off any covering, cut the substance under examination
colour. Add a further 20.0 ml of the alkali, attach a reflux into thin slices, if necessary, dry at about 60º for 48 hours and
condenser and boil for 1 hour on a water-bath. Cool, add 1 ml reduce to a powder.
of phenolphthalein solution and titrate the excess of alkali A. When examined under a microscope, a suspension in a
with 0.5 M hydrochloric acid. Repeat the operation with the 2 per cent w/v solution of potassium hydroxide appears as
same quantities of the same reagents in the same manner granules of latex agglomerated in irregular masses and light
without the oil and calculate the amount of total menthol from brown elongated filaments. Some fragments of vessels and
the following expression. rather elongated, refringent crystals are also visible, as well as
a smaller number of round pollen grains and fragments of
elongated fibres. Hairs of various lengths with sharp points
and a few grains of starch introduced during the handling of
the latex may be present. Fragments of epicarp consisting of
where, S is the amount, in g, of the acetylated sample taken, a
polygonal cells with thick walls defining a stellate lumen may
is the amount, in ml, of 0.5 M hydrochloric acid consumed in
also be present.
the blank test, and b is the amount, in ml, of 0.5 M
hydrochloric acid consumed in saponification of the B. Determine by thin-layer chromatography (2.4.17), coating
acetylated oil. the plate with silica gel G.
Storage. Store protected from light and moisture. Mobile phase. A freshly prepared mixture of 20 volumes of
acetone, 20 volumes of toluene, 3 volumes of ethanol (95 per
cent) and 1 volume of strong ammonia solution.
Test solution. Triturate 0.1 g of the powdered substance with
Opium 5 ml of ethanol (70 per cent), add 3 ml of ethanol (70 per
Raw Opium; Papaver cent), transfer to a 25-ml conical flask and heat in a water-bath
at 50º to 60º for 30 minutes, with stirring. Cool, filter, wash the
filter with ethanol (70 per cent) and dilute the filtrate to 10 ml
with the same solvent.
Reference solution. Dissolve 2 mg of papaverine
hydrochloride RS, 12 mg of codeine phosphate RS, 12 mg of
noscapine hydrochloride RS, and 25 mg of morphine
hydrochloride RS in ethanol (70 per cent) and dilute to 25 ml
with the same solvent.
Apply to the plate 20 µl of each solution as 20 mm bands.
After development, dry the plate at 100º to 105º for 15 minutes,
allow to cool and spray with potassium iodobismuthate
solution and then with a 0.4 per cent w/v solution of sulphuric
acid. The chromatogram obtained with the reference solution
shows in the lower part an orange-red or red band (morphine),
above it a similarly coloured band (codeine) and in the upper
Opium is the air-dried latex obtained by incision from the unripe part an orange-red or red band (papaverine) and above it a
capsules of Papaver somniferum Linn. similarly coloured band (noscapine). The chromatogram

1416
IP 2007 OPIUM POWDER

obtained with the test solution shows orange-red or red bands octylsilane bonded to porous silica (5 µm), fitted with a
corresponding to those in the chromatogram obtained with guard column 4 cm x 4.6 mm packed with octylsilane
the reference solution. The chromatogram obtained with the bonded to porous silica (5 µm),
test solution may also show a dark red band (thebaine) situated – mobile phase: 1.0 g of sodium heptanesulphonate in
between those due to codeine and to papaverine. 420 ml of water, adjusting to pH 3.2 with phosphoric
C. To 1 g of the powdered substance add 5 ml of water, shake acid that has been diluted to contain 0.49 per cent w/v
for 5 minutes, filter and add to the filtrate 0.25 ml of ferric solution of H3PO4 (about 5 ml) and adding 180 ml of
chloride solution; a red colour develops which does not acetonitrile,
disappear on the addition of 0.5 ml of 2 M hydrochloric acid. – flow rate. 1.5 ml per minute,
– spectrophotometer set at 280 nm.
Tests Inject suitable volumes of each solution. The assay is not
valid unless the resolution between the peaks corresponding
Thebaine. Not more than 3 per cent, calculated on the dried
to morphine and codeine is at least 2.5. If necessary, adjust
basis.
the volume of acetonitrile in the mobile phase. Inject the
Determine by liquid chromatography (2.4.14). reference solution six times. The assay is not valid unless the
Test solution. Use the test solution prepared in the assay. relative standard deviation of the peak area for morphine is
not more than 1.0 per cent.
Reference solution. Dissolve 25 mg of thebaine in sufficient
mobile phase to produce 25.0 ml and dilute 10.0 ml of this Inject the test solution and the reference solution.
solution to 100.0 ml with the mobile phase.
Calculate the percentage content of each alkaloid from the
Chromatographic system as described in the Assay. expression
The test is not valid unless the capacity factor for thebaine is
at least 3.0 and the number of theoretical plates is at least
3000. Calculate the percentage content of thebaine from the
expression given in the assay. where, w1 = weight, in g, of the alkaloid used to prepare the
reference solution,
w 1 × A 2 × 625Ash (2.3.19). Not more than 6.0 per cent, determined on 0.5 g.
× 100
Loss w2 = weight, in g, of the substance under
w 2 × A1 × 5(100 − h)on drying (2.4.19). Not more than 15.0 per cent,
determined on 0.25 g cut into thin slices, by drying in an oven examination used to prepare the test solution,
at 105º for 4 hours. A 1 = area of the peak corresponding to the alkaloid
in the chromatogram obtained with the
Assay. Determine by liquid chromatography (2.4.14).
reference solution,
Test solution. Suspend about 1.0 g, accurately weighed A 2 = area of the peak corresponding to the alkaloid
substance under examination, cut into thin slices, in 50 ml of in the chromatogram obtained with the test
ethanol (50 per cent), mix with the aid of ultrasound for solution,
1 hour, allow to cool, dilute to 100.0 ml with the same solvent h = percentage loss on drying.
and allow to stand. To 10.0 ml of the supernatant liquid add
5 ml of ammonia buffer pH 9.5, dilute to 25.0 ml with water, For calculation, 1 mg of morphine hydrochloride may be taken
mix, transfer 20.0 ml of the solution to a column (about 15 cm × as equivalent to 0.759 mg of morphine and 1 mg of codeine
30 mm) containing 15 g of kieselguhr for column phosphate may be taken as equivalent to 0.943 mg of codeine.
chromatography and allow to stand for 15 minutes. Elute with Storage. Store protected from light and moisture.
two quantities, each of 40 ml, of a mixture of 85 volumes of
dichloromethane and 15 volumes of 2-propanol, evaporate
the eluate to dryness at 40º at a pressure of 2 kPa, transfer the
residue to a 25-ml volumetric flask with the aid of the mobile Opium Powder
phase and dilute to volume with the same solvent. Opium Powder is Opium dried at a temperature not exceeding
Reference solution. Weigh accurately 0.1 g of morphine 70º, reduced to a fine or moderately fine powder and adjusted
hydrochloride and 25 mg of codeine, dissolve in sufficient of by the addition of Lactose, suitably coloured with Caramel, or
the mobile phase to produce 25.0 ml and dilute 10.0 ml of this other suitable diluent to contain about 10 per cent of morphine
solution to 100.0 ml with the same solvent. and 2 per cent of codeine.
Chromatographic system Opium Powder contains not less than 9.5 per cent and not
– a stainless steel column 25 cm x 4.6 mm, packed with more than 10.5 per cent of morphine, C17H19NO3, and not less

1417
OPIUM POWDER IP 2007

than 1.9 per cent and not more than 2.1 per cent of codeine, similarly coloured band (noscapine). The chromatogram
C18H21NO3, both calculated on the dried basis. obtained with the test solution shows orange-red or red bands
Description. A light brown powder consisting of yellowish corresponding to those in the chromatogram obtained with
brown or brownish red particles; odour, characteristic. the reference solution. The chromatogram obtained with the
test solution may also show a dark red band (thebaine) situated
Identification between those due to codeine and to papaverine.
C. To 1 g of the powdered substance add 5 ml of water, shake
A. When examined under a microscope, the residue obtained
for 5 minutes, filter and add to the filtrate 0.25 ml of ferric
after extraction with water, appears as granules of latex
chloride solution; a red colour develops which does not
agglomerated in irregular masses and light brown elongated
disappear on the addition of 0.5 ml of 2 M hydrochloric acid.
filaments. Some fragments of vessels and rather elongated,
refringent crystals are also visible, as well as a smaller number Tests
of round pollen grains and fragments of elongated fibres. Hairs
of various lengths with sharp points and a few grains of starch Thebaine. Not more than 3 per cent, calculated on the dried
introduced during the handling of the latex may be present. basis.
Fragments of epicarp consisting of polygonal cells with thick Determine by liquid chromatography (2.4.14).
walls defining a stellate lumen may also be present and if
powdered cocoa husk is present, the following: brown colour Test solution. Use the test solution prepared in the Assay.
of the fragments; narrow spiral vessels about 10 to 20 mm Reference solution. Dissolve 25 mg of thebaine in sufficient
wide, in groups of from one to six, traversing a spongy mobile phase to produce 25.0 ml and dilute 10.0 ml of this
parenchyma of thin-walled cells about 40 to 60 mm in either solution to 100.0 ml with the mobile phase.
direction and united by arm-like projections enclosing almost
circular intercellular spaces; fragments of the Chromatographic condition as described in the Assay.
sclerenchymatous layer, consisting of thick-walled lignified The test is not valid unless the capacity factor for thebaine is
brown, rectangular to polyhedral cells in a single layer, at least 3.0 and the number of theoretical plates is at least
individual cells about 5 to 10 µm wide and 10 to 30 µm long; 3000. Calculate the percentage content of thebaine from the
fragments of mucilage staining in ruthenium red solution. expression given in the Assay.
B. Determine by thin-layer chromatography (2.4.17), coating Ash (2.3.19). Not more than 6.0 per cent, determined on 0.5 g.
the plate with silica gel G..
Loss on drying (2.4.19). Not more than 15.0 per cent,
Mobile phase. A mixture of 20 volumes of acetone, 20 volumes determined on 0.25 g by drying in an oven at 105º for 4 hours.
of toluene, 3 volumes of ethanol (95 per cent) and 1 volume
Assay. Determine by liquid chromatography (2.4.14).
of strong ammonia solution.
Test solution. Suspend about 1.0 g, accurately weighed, of
Test solution. Triturate 0.10 g of the powdered substance with
the substance under examination, cut into thin slices, in 50 ml
5 ml of ethanol (70 per cent), add 3 ml of ethanol (70 per
of ethanol (50 per cent), mix with the aid of ultrasound for
cent), transfer to a 25-ml conical flask and heat in a water-bath
1 hour, allow to cool, dilute to 100.0 ml with the same solvent
at 50º to 60º for 30 minutes, with stirring. Cool, filter, wash the
and allow to stand. To 10.0 ml of the supernatant liquid add
filter with ethanol (70 per cent) and dilute the filtrate to 10 ml
5 ml of ammonia buffer pH 9.5, dilute to 25.0 ml with water,
with the same solvent.
mix, transfer 20.0 ml of the solution to a column (about 15 cm ×
Reference solution. Dissolve 2 mg of papaverine 30 mm) containing 15 g of kieselguhr for column
hydrochloride RS, 12 mg of codeine phosphate RS, 12 mg of chromatography and allow to stand for 15 minutes. Elute with
noscapine hydrochloride RS, and 25 mg of morphine two quantities, each of 40 ml, of a mixture of 85 volumes of
hydrochloride RS in ethanol (70 per cent) and dilute to 25 ml dichloromethane and 15 volumes of 2-propanol, evaporate
with the same solvent. the eluate to dryness at 40º at a pressure of 2 kPa, transfer the
Apply to the plate 20 µl of each solution as 20 mm bands. residue to a 25-ml volumetric flask with the aid of the mobile
After development, dry the plate at 100º to 105º for 15 minutes, phase and dilute to volume with the same solvent.
allow to cool and spray with potassium iodobismuthate Reference solution. Weigh accurately 0.1 g of morphine
solution and then with a 0.4 per cent w/v solution of sulphuric hydrochloride RS and 25 mg of codeine phosphate RS,
acid. The chromatogram obtained with the reference solution dissolve in sufficient of the mobile phase to produce 25.0 ml
shows in the lower part an orange-red or red band (morphine), and dilute 10.0 ml of this solution to 100.0 ml with the same
above it a similarly coloured band (codeine) and in the upper solvent.
part an orange-red or red band (papaverine) and above it a

1418
IP 2007 PEPPERMINT OIL

Chromatographic system Description. A white to light brown, amorphous or slightly


– a stainless steel column 25 cm x 4.6 mm, packed with granular powder; odour, characteristic.
octylsilane bonded to porous silica (5 µm), fitted with a
guard column 4 cm x 4.6 mm packed with octylsilane Tests
bonded to porous silica (5 µm),
Microbial contamination (2.2.9). 1.0 g is free from Escherichia
– mobile phase: 1.0 g of sodium heptanesulphonate in
coli and 10.0 g is free from salmonellae.
420 ml of water, adjusting to pH 3.2 with phosphoric
acid that has been diluted to contain 0.49 per cent w/v Loss on drying (2.4.19). Not more than 7.0 per cent, determined
solution of H3PO4 (about 5 ml) and adding 180 ml of on 1 g by drying in an oven at 60o at a pressure not exceeding
acetonitrile, 0.7 kPa for 4 hours.
– flow rate. 1.5 ml per minute, Assay. Weigh accurately about 0.5 g, triturate with 10 ml of
– spectrophotometer set at 280 nm. cysteine hydrochloride solution and dilute to 100.0 ml with
water. To 30 ml of water in each of two flasks add 15.0 ml of
Inject suitable volume of reference solution. The test is not casein solution and maintain at 60o by heating on a water-
valid unless the resolution between the peaks corresponding bath. To the first flask add 5.0 ml of the solution of the
to morphine and codeine is at least 2.5. If necessary, adjust substance under examination, and to the second flask add
the volume of acetonitrile in the mobile phase. The relative 5.0 ml of the same solution, previously boiled for 2 minutes
standard deviation of the peak area for morphine is not more and cooled. Maintain the solutions at 60o for 30 minutes, cool
than 1.0 per cent. rapidly to room temperature and add to each flask 0.75 ml of
Inject the test solution and the reference solution. phenolphthalein solution and 10 ml of formaldehyde solution,
previously neutralised to phenolphthalein solution. Titrate
Calculate the percentage content of each alkaloid from the
both solutions with 0.1 M sodium hydroxide to the same
expression
definite pink colour; the difference between the two titrations
w 1 × A 2 × 625 × 100 is not less than 4.5 ml.
w 2 × A1 × 5(100 − h) Storage. Store protected from light and moisture.
Labelling. The label states the name of any added substance.
where, w1 = weight, in g, of the alkaloid used to prepare the
reference solution,
w2 = weight, in g, of the substance under
examination used to prepare the test solution, Peppermint Oil
A 1 = area of the peak corresponding to the alkaloid
Peppermint Oil is obtained by steam distillation from the aerial
in the chromatogram obtained with the
parts of the flowering plant of Mentha piperita L. and M.
reference solution,
arvensis var. piperascens.
A 2 = area of the peak corresponding to the alkaloid
in the chromatogram obtained with the test Peppermint Oil contains not less than 4.5 per cent w/w and
solution,h not more than 10.0 per cent w/w of esters, calculated as menthyl
h = percentage loss on drying. acetate, C12H22O2, not less than 44.0 per cent w/w of free
alcohols, calculated as menthol, C10H20O, and not less than
For calculation, 1 mg of morphine hydrochloride may be taken 15.0 per cent w/w and not more than 32.0 per cent w/w of
as equivalent to 0.759 mg of morphine and 1 mg of codeine ketones, calculated as menthone, C10H18O.
phosphate may be taken as equivalent to 0.943 mg of codeine.
Description. A colourless, pale yellow or pale greenish yellow
Storage. Store protected from light and moisture. liquid; odour, characteristic; taste, characteristic followed by
sensation of cold.

Identification
Papain Determine by thin-layer chromatography (2.4.17), coating the
Papain is an enzyme or a mixture of enzymes obtained from the plate with silica gel G F254.
juice of the unripe fruit of Carica papaya Linn. (Fam. Mobile phase. A mixture of 95 volumes of toluene and
Caricaceae). It may contain a suitable diluent such as Lactose. 5 volumes of ethyl acetate.
Papain contains not less than the minimum protease activity Test solution. Dissolve 1 g of the oil under examination in
determined under the conditions of the Assay. 100 ml of toluene.

1419
PEPPERMINT OIL IP 2007

Reference solution. Dissolve 50 mg of (–)-menthol RS, 20 µl or a few pieces of porous pot and heat under a reflux condenser
of cineole RS, 10 mg of thymol RS and 10 µl of menthyl acetate on a water-bath for 30 minutes. Add 1 ml of phenolphthalein
RS in sufficient toluene to produce 10 ml. solution and immediately titrate with 0.5 M hydrochloric acid.
Apply to the plate 20 µl of test solution and 10 µl of reference Repeat the operation without the substance under examination.
solution as bands 20 mm by 3 mm. After development, dry the The difference between the titrations represents the volume
plate in air until the odour of solvent is no longer detectable of alkali required to saponify the esters.
and examine in ultraviolet light at 254 nm. In the chromatogram 1 ml of 0.5 M ethanolic potassium hydroxide is equivalent to
obtained with test solution there are no quenching bands at 0.09915 g of esters, calculated as menthyl acetate, C12H22O2
Rf values slightly lower than that of the faint band due to
For free alcohols — To 1 g in a dry, 150-ml acetylation flask,
thymol in the chromatogram obtained with reference solution
add 3 ml of a mixture of 3 volumes of pyridine and 1 volume of
(carvone and pulegone). Spray the plate with anisaldehyde
acetic anhydride. Determine the weight of the acetylation
solution and examine in daylight after heating at 105o for
mixture to the nearest mg, keeping the flask closed while
5 minutes. The chromatogram obtained with reference solution
weighing. Boil under a reflux condenser in a water-bath for
shows, in order of increasing Rf value, an intense blue to
3 hours, maintaining the water level 2 to 3 cm above the level
violet band (menthol) in the lower third, a violet-blue to brown
of the liquid in the flask throughout. Remove the flask from
band (cineole), a pink band (thymol) and a violet-blue band
the water-bath and add 50 ml of water through the condenser,
(menthyl acetate). The chromatogram obtained with test
remove the condenser and wash the walls of the flask with
solution shows an intense band corresponding to menthol, a
10 ml of water. Allow to stand for 15 minutes and titrate with
band corresponding to cineole, a violet-blue band
0.5 M sodium hydroxide using 1 ml of phenolphthalein
corresponding to menthyl acetate in the middle of the
solution as indicator. Repeat the operation without the
chromatogram and at a slightly lower Rf value a greenish band
substance under examination. The difference between the
(menthone). The chromatogram obtained with test solution
titrations represents the volume of sodium hydroxide required.
does not show intense greyish green or faint bluish grey bands
at Rf values between those of cineole and thymol in the 1 ml of 0.5 M sodium hydroxide is equivalent to 0.07815 g of
chromatogram obtained with reference solution (carvone, free alcohols, calculated as menthol, C10H20O. If the quantities
pulegone, isomenthone). The chromatogram obtained with of acetic anhydride in pyridine used in the two determinations
test solution shows an intense reddish violet band near the differ by more than 5 mg, adjust the volume of alkali used in
solvent front (hydrocarbons) and a brownish yellow band the second titration by multiplying with a/b where a is the
(menthofuran) at a slightly lower Rf value; other less intensely weight, in g, of acetic anhydride in pyridine used in the first
coloured bands may also be seen. determination and b is the weight, in g, of acetic anhydride in
pyridine used in the second test.
Tests For ketones — To 2 g add 25 ml of a 5.0 per cent w/v solution
Acidity. To 2 g add 0.25 ml of phenolphthalein solution; not of hydroxylamine hydrochloride in ethanol (95 per cent),
more than 0.1 ml of 0.5 M ethanolic potassium hydroxide is heat on a water-bath for 1 hour, allow to cool, add about 1 mg
required to change the colour of the solution. of methyl orange and titrate with 0.5 M ethanolic potassium
hydroxide until an orange-yellow colour is obtained. Repeat
Optical rotation (2.4.22). –10o to –30o. the heating for further periods of 1 hour until, after cooling,
Refractive index (2.4.27). 1.460 to 1.467. not more than 0.1 ml of 0.5 M ethanolic potassium hydroxide
is required to neutralise the solution.
Weight per ml (2.4.29). 0.900 g to 0.916 g.
1 ml of 0.5 M ethanolic potassium hydroxide is equivalent to
Dimethyl sulphide. Distil 25 ml, collect the first 1 ml of the
0.07710 g of ketones, calculated as menthone, C10H18O.
distillate and carefully superimpose it on 5 ml of a 6.5 per cent
w/v solution of mercuric chloride; no white film is produced Storage. Store protected from light and moisture, in well-filled
within 1 minute at the interface of the two liquids. containers.
Fixed oils and resinified volatile oils. Allow 0.05 ml to fall on
a filter paper. The oil evaporates completely within 24 hours
without leaving a translucent or greasy mark.
Assay. For esters — To 2 g in a borosilicate glass flask add
2 ml of ethanol (90 per cent) and 0.25 ml of phenolphthalein
solution, neutralise with 0.5 M ethanolic potassium
hydroxide, add an additional 25.0 ml and a little pumice powder

1420
IP 2007 PIPPALI LARGE

Pippali, Large Reference solution. Reflux 0.4 g of the coarsely powdered


pippali, Big RS with 50-75 ml of methanol for 15 minutes, cool
Long pepper; Catkins (Big) and filter. Reflux the residue further for two times with 75 ml of
methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 10 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultra violet light at 254 nm. The chromatographic
profile of the test solution corresponds to that in the
chromatogram obtained with reference solution. Spray with
solution of vanillin sulphuric acid reagent. Heat at 100º for
5-10 minutes and examine the plate in day light. The
chromatographic profile of the test solution is similar to that
of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 8 per cent.
Pippali, Large consists of the fruiting spikes of Piper longum
Water-soluble extractive (2.6.3). Not less than 10 per cent by
Linn. (Syn. P. sarmentosum Wall., P. latifolium Hunter, Chavica
Method I.
roxburghii Miq., C. sarmentosa Miq.) (Fam. Piperaceae)
Ash (2.3.19). Not more than 8 per cent.
Pippali, Large contains not less than 1 per cent w/w of piperine,
calculated on the dried basis. Acid-insoluble ash (2.3.19). Not more than 3 per cent.
Description. The spikes are blacking green to green in colour, Heavy metals (2.3.13). 1.0 g complies with the limit test for
cylindrical, erect and blunt. It has pungent taste and the odour heavy metals, Method B (20 ppm).
is aromatic and characteristic. The spikes are 2 to 4 cm long Loss on drying (2.4.19). Not more than 12.0 per cent,
and 0.4-0.7 cm in diameter. determined on 5 g by drying in an oven at 1050.
Identification Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
A. Macroscopic — The spikes are blackish green to green in
colour, surface rough. The spikes bear bracts and numerous Assay. Determine by liquid chromatography (2.4.14).
small fruits sunk in solid spike. Test solution. Weigh 2 g of coarsely powdered substance
B. Microscopic — Epidermis is a single layer of tangentially under examination, add 50 ml of methanol, sonicate for
elongated cells. Cells are packed with abundant starch grains 3 minutes and heat on a boiling water bath for 15 minutes, cool
which are small. Crystals are present in some of the cells, they and dilute to 100.0 ml with methanol and filter. Dilute further if
appear unequal in size. The epicarp is a single layer of thick necessary.
walled cells having greenish content. Endocarp is wavy in Reference solution. A 0.01 per cent w/v solution of
outline. Mostly, endocarp and seed coat are fused together to piperine RS in methanol.
form a deep zone with hyaline content in outer layer and orange
Chromatographic system
red (brown) inner region.
– a stainless steel column 25 cm × 4.6 mm packed with
C Determine by thin-layer chromatography (2.4.17), coating octadecylsilane bonded to porous silica (5 µm),
the plate with silica gel GF254. – mobile phase: a gradient mixture of acetonitrile and a
Mobile phase. A mixture of 60 volumes of benzene, 30 volumes buffer solution prepared by dissolving 0.136 g of
of ethyl acetate and 10 volumes of diethyl ether. potassium di-hydrogen orthophosphate in 900 ml of
water, adjust the pH to 2.5 with orthophosphoric acid
Test solution. Reflux 2 g of the coarsely powdered substance and dilute to 1000 ml with water,
under examination with 50-75 ml of methanol for 15 minutes, – flow rate. 1.5 ml per minute,
cool and filter. Reflux the residue further for two times with – spectrophotometer set at 270 nm,
75 ml of methanol, cool and filter. Combine all the filtrates and – a 20 µl loop injector.
concentrate under vacuum to 50 ml.

1421
PIPPALI SMALL IP 2007

Time Buffer solution Acetonitrile which are small. Crystals are present in some of the cells, they
(min) (per cent v/v) (per cent v/v) appear unequal in size. The epicarp is a single layer of thick
0 95 5 walled cells having greenish content. Endocarp is wavy in
outline. Mostly, endocarp and seed coat are fused together to
18 55 45
form a deep zone with hyaline content in outer layer and orange
25 20 80 red (brown) inner region.
30 95 5 C Determine by thin-layer chromatography (2.4.17), coating
Inject the reference solution. The relative standard deviation the plate with silica gel GF254.
for the replicate injections is not more than 2.0 per cent.
Mobile phase. A mixture of 60 volumes of benzene, 30 volumes
Inject the test solution and the reference solution. of ethyl acetate and 10 volumes of diethyl ether.
Calculate the content of piperine. Test solution. Reflux 2 g of the coarsely powdered substance
Storage. Store protected from heat, moisture and against attach under examination with 50-75 ml of methanol for 15 minutes,
by insects and rodents. cool and filter. Reflux the residue further for two times with
75 ml of methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 50 ml.
Reference solution. Reflux 0.4 g of the coarsely powdered
Pippali, Small pippali, small RS with 50-75 ml of methanol for 15 minutes,
Small pepper; Catkins (small) cool and filter. Reflux the residue further for two times with
75 ml of methanol, cool and filter. Combine all the filtrates and
concentrate under vacuum to 10 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
and examine in ultra violet light at 254 nm. The chromatographic
profile of the test solution corresponds to that in the
chromatogram obtained with reference solution. Spray with
vanillin sulphuric acid reagent. Heat at 100º for 5-10 minutes
and examine the plate in day light. The chromatographic profile
of the test solution is similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 8 per cent.
Water-soluble extractive (2.6.3). Not less than 10 per cent by
Pippali, small consists of the fruiting spike of Piper longum Method I.
Linn. (Fam. Piperaceae)
Ash (2.3.19). Not more than 8 per cent.
Pippali, small contains not less than 0.4 per cent w/w of piperine,
Acid-insoluble ash (2.3.19). Not more than 3 per cent.
calculated on the dried basis.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Description. The spikes are greenish black to black in colour,
heavy metals, Method B (20 ppm).
cylindrical, erect and blunt. It has pungent taste and the odour
is aromatic and characteristic. The spikes are 1.0 to 1.9 cm Loss on drying (2.4.19). Not more than 12.0 per cent,
long and 0.2 to 0.3 cm in diameter. determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
Identification contamination tests.
A. Macroscopic — The spikes are greenish black to black in Assay. Determine by liquid chromatography (2.4.14).
colour, surface rough. The spikes bear bracts and numerous
small fruits sunk in solid spike. Test solution. Weigh 2 g of coarsely powdered substance
under examination, add 50 ml of methanol, sonicate for
B. Microscopic — Epidermis is a single layer of tangentially 3 minutes and heat on a boiling water bath for 15 minutes, cool
elongated cells. Cells are packed with abundant starch grains and dilute to 100.0 ml with methanol and filter. Dilute further if

1422
IP 2007 PUNARNAVA

necessary. Punarnava contains not less than 0.005 per cent of boeravinone
Reference solution. A 0.01 per cent w/v solution of B, calculated on the dried basis.
piperine RS in methanol. Description. A greyish-brown in colour, short and fibrous
Chromatographic system fracture and no distinct odour with bitter taste.
– a stainless steel column 25 cm x 4.6 mm packed with
Identification
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a gradient mixtures of acetonitrile and a A. Macroscopic — Stout, tapering, somewhat knotty and
buffer solution prepared by dissolving 0.136 g of twisted roots upto 30 cm or more long and 0.5-1.5 cm thick
potassium di-hydrogen orthophosphate in 900 ml of often crowned with stem bases, greyish-brown, surface is
water, adjust the pH to 2.5 with orthophosphoric acid rough due to minute, irregular, longitudinal striations and root
and dilute to 1000 ml with water, scars.
– flow rate. 1.5 ml per minute,
B. Microscopic — Transverse section of root shows
– spectrophotometer set at 270 nm,
outermost layer of cork which consists of thin walled
– a 20 µl loop injector.
tangentially elongated cells with brownish walls followed by
Time Buffer solution Acetonitrile thin walled 1-2 layered cork cambium. Cortex many layered
(min) (per cent v/v) (per cent v/v) and composed of thin walled cells. Secondary cortex 2-4
0 95 5 layered and parenchymtous. Concentric bands of xylem tissues
18 55 45 alternating with parenchymatous tissues. Vessels are radial
with reticulate thickening. Simple and compound starch grains
25 20 80
with centric hilum and raphide crystals of calcium oxalate in
30 95 5 cortex region. Fibres are aseptate and spindle shaped with
Inject the reference solution. The relative standard deviation pointed ends.
for the replicate injections is not more than 2.0 per cent. C. Determine by thin-layer chromatography (2.4.17), coating
Inject the test solution and the reference solution. The relative the plate with silica gel G.
retention time of piperine is 1. Mobile phase. A mixture of 35 volumes of chloroform,
Calculate the content of piperine. 6 volumes of methanol and 1 volume of glacial acetic acid.

Storage. Store protected from heat, moisture and against attach Test solution. Reflux 2 g of the coarsely powdered substance
by insects and rodents. under examination with 25 ml of methanol for 15 minutes, cool
and filter. Reflux the residue further with 2 × 25 ml of methanol,
cool and filter. Combine all the filtrates and concentrate under
vacuum to 5 ml.
Punarnava Reference solution. Reflux 2 g of the punarnava RS with 25 ml
Boerhaavia; Hogweed of methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 × 25 ml of methanol, cool and filter. Combine all
the filtrates and concentrate under vacuum to 5 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with a anisaldehyde solution. Heat at 110° for 10 minutes
and examine the plate in day light. The chromatographic profile
of the test solution is similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 0.5 per cent.
Water-soluble extractive (2.6.3). Not less than 9.0 per cent by
method I.

Punarnava consists of the dried root of Boerhaavia diffusa Ash (2.3.19). Not more than 10.0 per cent.
Linn. (syn. B. reperis Linn) (Fam. Nyctaginaceae). Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.

1423
SERPGANDHA IP 2007

Heavy metals (2.3.13). 1.0 g complies with the limit test for Identification
heavy metals, Method B (20 ppm).
A. Macroscopic — Roots are sub cylindrical to tapering,
Loss on drying (2.4.19). Not more than 10.0 per cent, tortuous or curved, rarely branched. Occurs as segments
determined on 5 g by drying in an oven at 1050. usually from 5 to 15 cms in length and 3 to 20 mm in diameter.
Microbial contamination (2.2.9). Complies with the microbial Externally grayish yellow to brown, wood pale yellow. Roots
contamination tests. tough with longitudinal marking & slightly wrinkled surface.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux 2 g of the coarsely powdered substance
under examination with 50 ml of methanol on a water-bath for
15 minutes, cool and filter. Reflux the residue further with
methanol till the extract turns colourless, cool and filter.
Combine all the filtrates and concentrate to a volume slightly
less than 25 ml. Dilute to 25.0 ml with methanol.
Reference solution. A 0.002 per cent w/v solution of
boeravinone B RS in methanol.
Chromatographic system
– a stainless steel column 25 cm × 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a gradient mixtures of water and
acetonitrile,
– flow rate. 1 ml per minute,
– spectrophotometer set at 272 nm, When scraped, bark separates readily from the wood. Fracture
– a 20 µl loop injector. is short and irregular.
Time Water Acetonitrile B. Microscopic — In transverse section, cork cells in 2 to 8
(min) (per cent v/v) ( per cent v/v) alternating bands of radically narrow and broader cells, thin,
0 90 10 lignified up to 75 µm in tangential width, broader cells up to
25 10 90 about 90 µm in radial length, phelloderm, tangentially elongated
to isodimetric parenchyma cells containing starch and short
35 90 10 latex cells with brown resinous matter; secondary cortex
Inject the reference solution. The test is not valid unless the consists of parenchyma cells, heavily packed with starch
relative standard deviation for the replicate injections is not grains secondary phloem contains phloem parenchyma and
more than 2.0 per cent. sieve elements, parenchyma contains starch and angular
crystals of calcium oxalate 3 to 20 µm in length. Xylem is
Inject the test solution and reference solution.
about 4/5 of the diameter of the root, wood is transversed by
Calculate the content of boeravinone B. medullay rays 1 to 5 cells in width. Xylem consists of vessels,
Storage. Store protected from heat, moisture and against attack tracheids, wood parenchyma & wood fibers. Xylem vessels
by insects and rodents. are elongated up to 350 µm in length and 50 µm in width and
contains simple or bordered pits; tracheids lignified, pitted;
wood parenchyma with moderately thick, lignified and pitted
walls containing starch; wood fibres highly thickened with
Serpgandha pointed ends, stone cells absent.
Rauwolfia Root C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel G..
Serpgandha consists of the dried roots of Rauwolfia
Serpentina Bentham ex Kurz (Fam Apocynaceae). Mobile phase. A mixture of 70 volumes of chloroform and
30 volumes of acetone.
Serpgandha contains not less than 0.15 per cent of reserpine
and ajmalicine, calculated on the dried basis. Test solution. To 250 mg of the coarsely powdered substance
under examination, add 5 ml methanol, shake for 10 minutes,
Description. Taste bitter, odour indistinct. and filter. Wash the residue with 5 ml of methanol and add the
washing to the filtrate.

1424
IP 2007 SENNA PODS

Reference solution. To 250 mg of sepgandha RS, add 5 ml – a 10 µl loop injector.


methanol, shake for 10 minutes, and filter. Wash the residue Inject the reference solution (a). The test is not valid unless
with 5 ml of methanol and add the washing to the filtrate. the relative standard deviation for the replicate injections is
Apply to the plate 10 µl of each solution as bands 10 mm by not more than 2.0 per cent. Inject reference solution (b) for
2 mm. Allow the mobile phase to rise 12 cm. Dry the plate in air, peak identification.
spray with anisaldehyde solution and heat at 100° for 5 minutes
Inject the test solution and reference solution (a).
and examine the plate in day light. The chromatogram obtained
with test solution shows two pinkish-violet bands Calculate the content of reserpine and ajmalicine.
corresponding to the bands in the chromatogram obtained Storage. Store protected from heat, moisture and against attack
with reference solution. A dark brown band may also appear by insects and rodents.
at the line of application in the chromatogram obtained with
test solution.

Tests Senna Pods


Foreign organic matter (2.6.1). Not more than 2.0 per cent Senna Fruit; Pods of cassia
Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent.
Water-soluble extractive (2.6.3). Not less than 5.0 per cent.
Ash (2.3.19). Not more than 8.0 per cent.
Acid-insoluble ash (2.3.19): Not more than 2.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°.
Microbial contamination limits (2.2.9). Complies with the
microbial contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Reflux about 2 g of the coarsely powdered
substance under examination with 50 ml of Senna pods consist of the dried compound Pods of Cassia
ethanol (95 per cent), on a water bath for 15 minutes, cool angustifolia or Cassia senna Vhal. (Family: Leguminosae).
and filter. Reflux the residue further with ethanol (95 per Senna Pods contains not less than 1 per cent of sennosides A
cent), till the last extract turns colorless, cool and filter. Combine and B, claculated on the dried basis.
all the filterates and concentrate to 100.0 ml.
Description. Pale yellowish green coloured pods with slight
Reference solution (a). A solution containing a 0.002 per cent odour. Leaflets with mucilaginous and faint odour.
w/v reserpine RS and 0.002 per cent w/v ajmalicine RS in
ethanol (95 per cent). Identification
Reference solution (b). A 0.002 per cent w/v reserpine RS in A. Macroscopic — Flattened reniform pods, brownish yellow
ethanol (95 per cent) for peak identification. at the edges, dark brown in the central area about 40 to 50 mm
Chromatographic system long and about at least 20 mm wide. At one end is a stylar
– a stainless steel column 25 cm × 4.6 mm packed with point and at the other a short stalk. The pods contains 5 to 7
octadecylsilane bonded to porous silica (5µm), flattened and obovate seeds, green to pale brown, with a
– mobile phase: a mixture of 35 volumes of acetonitrile continuous network of prominent ridges on the tests and in
and 65 volumes of a buffer solution prepared by complate wavy transverse ridges on the testa. Leaflets, 2.5 to
dissolving 6.80 g of potassium dihydrogen phosphate 8 cm long and 5-15 mm wide at centre, pale yellowish green,
in 1000 ml water, adjust the pH to 3.0 with dilute elongated lanceolate, slightly asymmetric at base; margins
orthophosphoric acid, entire, flat, apex acute with a sharp spine; both surface smooth
– flow rate. 1 ml per minute, with sparce trichomes; odour, faint but distinctive; taste,
– spectrophotometer set at 268 nm, mucilaginous and disagreeable but not distinctly bitter.

1425
SENNA LEAF IP 2007

B. Microscopic — The pods present an epicarp with strongly Assay. Determine by liquid chromatography (2.4.14).
cuticularised isodiametric cells, occasional anomocytic or
Test solution. Weigh accurately 1.0 g of the coarsely powdered
paracytic stomata, and very few conical, unicellular and warty
substance in a round bottom flask, add about 10 ml of 1 per
trichomes. Hypodermis with collenchymatous cells, mesocarp
cent v/v acetic acid and 25 ml of methanol and reflux on a
with parenchymatous tissue, a layer of prism s of calcium
water bath for about 30 minutes. Cool to room temperature;
oxalate and containing vascular bundles incompletely
make up the volume up to 50 ml with methanol and filter.
surrounded by fibres with a crystals sheth of calcium oxalate
prisms, endocarp consisting of thick-walled and inter lacing Reference solution. A 0.004 per cent w/v solution of
fibres. The seeds present a sub epidermal layers of palisade sennosides RS in methanol.
cells with thick outer walls, endosperm composed of Chromatographic system:
polyhedral cells with mucilaginous wall. – a stainless steel column 25 cm x 4.6 mm packed with
Reduce to moderately fine powder examine microscopically octadecylsilane bonded to porous silica (5 µm),
using chloral hydrate solution. The powder consists of epicarp – mobile phase. a mixtures of 82 volumes of 1 per cent
with polygonal cells and a small number of warty trichomes v/v acetic acid in water and 18 volumes of acetonitrile.
and occasional anomocytic stomata, fibres in two crossed – flow rate. 1 ml per minute,
layers, accomparied by a crystal sheath of calcium oxalate – spectrophotometer set at 350 nm,
prism, characteristic palisade cells in the seeds and stratified – a 20 µl loop injector.
cells in the endosperm, cluster and prisms of calcium oxalate Inject the reference solution. The test is not valid unless the
C. In the Assay, the chromatogram obtained with test solution relative standard deviation for the replicate injections is not
corresponds to the chromatogram obtained with reference more than 2.0 per cent.
solution. Inject the test solution and reference solution.
D. Determine by thin-layer chromatography (2.4.17), coating Calculate the content of sennoside A and B.
the plate with silica gel F254.
Storage. Store protected from light and moisture.
Mobile phase. A mixture of 40 volumes of n- propyl alcohol,
40 volumes of ethyl acetate, 29 volumes of water and 1 volume
of glacial acetic acid. Senna Leaf
Test solution. Take 1 g of the dried leaves powder substance
under examination. Add 25 ml of methanol, reflux for 10 minutes,
Cassia leaf
cool and filter. Reflux the residue with another 20 ml of
methanol, cool and filter. Combine all the filtrates and
concentrate to 10 ml.
Reference solution. A solution containing 0.01 per cent w/v of
sennoside A and B RS in methanol.
Apply to the plate 10 µl of each solution as bands 10 mm by
6 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with 20 per cent v/v of nitric acid solution. Heat at 1000
to 1050 for about 10 minutes and immediately examine the
plate. The chromatographic profile of the test solution is similar
to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 1.0 per cent. Senna leaf consists of the dried compound leaves of Cassia
angustifolia or Cassia senna Vhal. (Family: Leguminosae).
Ash (2.3.19). Not more than 14.0 per cent.
Description. Pale yellowish green coloured leaflets with
Acid-insoluble ash (2.3.19). Not more than 2.5 per cent.
mucilaginous and faint odour.
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 105°. Identification
Microbial contamination (2.2.9). Complies with the microbial A. Macroscopic — Leaflets,2.5 to 8 cm long and 5-15 mm
contamination tests. wide at centre, pale yellowish green, elongated lanceolate,

1426
IP 2007 SHATAVARI

slightly asymmetric at base; margins entire, flat, apex acute Reference solution. A 0.004 per cent w/v solution of
with a sharp spine; both surface smooth with sparce trichomes; sennosides RS in methanol.
odour, faint but distinctive; taste, mucilaginous and Chromatographic system
disagreeable but not distinctly bitter. – a stainless steel column 25 cm x 4.6 mm packed with
B. Microscopic — Transverse section shows outer single octadecylsilane bonded to porous silica (5 µm),
layered mucilaginous epidermal cells. Unicellular hairs – mobile phase: a mixtures of 82 volumes of 1 per cent
presents. Stomata paracytic, numerous on both surface. v/v acetic acid in water and 18 volumes of acetonitrile,
Mesophyll consists of upper and lower palisade layers with – flow rate. 1 ml per minute,
spongy layer in between , primatic crystals of calcium oxalate – spectrophotometer set at 350 nm,
present on larger veins. – a 20 µl loop injector.
C. In the Assay, the chromatogram obtained with test solution Inject the reference solution. The test is not valid unless the
corresponds to the chromatogram obtained with reference relative standard deviation for the replicate injections is not
solution. more than 2.0 per cent.

D. Determine by thin-layer chromatography (2.4.17), coating Inject the test solution and reference solution.
the plate with silica gel F254. Calculate the content of sennoside A and B.
Mobile phase. A mixture of 40 volumes of n- propyl alcohol, Storage. Store protected from light and moisture.
40 volumes of ethyl acetate, 29 volumes of water and 1 volume
of glacial acetic acid.
Test solution. Take 1 g of the dried leaves powder substance Shatavari
under examination. Add 25 ml of methanol, reflux for 10 minutes,
cool and filter. Reflux the residue with another 20 ml of Asparagus racemosus root
methanol, cool and filter. Combine all the filtrates and
concentrate to 10 ml.
Reference solution. A 0.001 per cent w/v solution of
Sennoside A and B in methanol.
Apply to the plate 10 µl of each solution as bands 10 mm by
6 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with 20 per cent v/v of nitric acid solution. Heat at 1000
to 105° for about 10 minutes and immediately examine the
plate. The chromatographic profile of the test solution is similar
to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 1.0 per cent.
Ash (2.3.19). Not more than 14.0 per cent. Shatavari consists of the tuberous roots of Asparagus
Acid-insoluble ash (2.3.19). Not more than 2.5 per cent. racemosus willd. (Fam .Liliaceae).

Loss on drying (2.4.19). Not more than 12.0 per cent, Shatavari contains not less than 0.1 per cent of shatavarin IV,
determined on 5 g by drying in an oven at 105°. calculated on the dried basis.

Microbial contamination (2.2.9). Complies with the microbial Description. The tuberous root bits are dirty white in color,
contamination tests. longitudinally wrinkled with yellow hard central core. It is
starchy and slightly bitter followed by sweet taste.
Assay. Determine by liquid chromatography (2.4.14).
Test solution. Weigh accurately 1.0 g of the coarsely powdered
Identification
substance in a round bottom flask, add about 10 ml of 1 per A. Macroscopic — The tuberous roots are borne in a compact
cent v/v acetic acid and 25 ml of methanol and reflux on a bunch and are fleshy, and spindle shaped. They are marketed
water bath for about 30 minutes. Cool to room temperature; in pieces 5-15 cm in length and 2 cm in thickness. They are
make up the volume up to 50.0 ml with methanol and filter. silvery white or ash-colored externally and white internally,

1427
SHATAVARI IP 2007

more or less smooth when fresh, developing longitudinal Method I


wrinkles when dry.
Determine by thin-layer chromatography (2.4.17), coating the
B. Microscopic — The inner parenchymatous zone of cortex plate with silica gel GF 254.
is composed of 18-24 layers in upper and 42-47 layers in the
middle tuberous portion of the roots. The cells are thin - walled Mobile phase. A mixture of 6.5 volumes of chloroform and
cellulosic, with circular to oral outlines and distinct inter cellular 3.5 volumes of methanol.
spaces. In some roots 3-4 layers of cortex immediately adjacent Test solution. Reflux 4 g of the coarsely powdered substance
to the endodermis are modified into a sheath of stone cells under examination with 50 ml of methanol on a water-bath for
round the endodermis. The number of vascular bundles is 30- 30 minutes, cool and filter. Reflux the residue further with
35 in the upper levels and 35-45 in the middle tuberous portions methanol till the last extract turns colourless, cool and filter.
of the roots. Combine all the filtrates and concentrate to 50 ml.
C. Determine by thin-layer chromatography (2.4.17), coating Reference solution. A 0.008 per cent w/v solution of shatavarin
the plate with silica gel F254. IV RS in methanol.
Mobile phase. A mixture of 13 volumes of chloroform, Apply to the plate 5 µl of each solution as bands 10 mm by
10 volumes of methanol and 2 volumes of water. 2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air
Test solution. Reflux 1 g of the coarsely powdered substance and spray with 10 per cent v/v sulphuric acid in methanol.
under examination with 30 ml of methanol for 30 minutes, cool Heat the plate at 100º for 5 minutes, scan the plate in absorbance
and filter. Reflux the residue further with 2 × 30 ml of methanol, mode at 500 nm. Record the chromatograms and measure the
cool and filter. Combine all the filtrates and concentrate under responses for the analyte peak.
vacuum to 10.0 ml. Calculate the content of shatavarin IV.
Reference solution. Reflux 1 g of shatavari RS with 30 ml of
Method II
methanol for 30 minutes, cool and filter. Reflux the residue
further with 2 × 30 ml of methanol, cool and filter. Combine all Determine by liquid chromatography (2.4.14).
the filtrates and concentrate under vacuum to 10.0 ml. Test solution. Reflux 5 g of the coarsely powdered substance
Apply to the plate 5 µl of each solution as bands 10 mm by under examination with 50 ml of methanol on a water-bath for
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air, 30 minutes, cool and filter. Reflux the residue further with
spray with vanillin sulphuric acid reagent prepared by mixing methanol till the last extract turns colourless, cool and filter.
1 per cent w/v vanillin in ethanol (95 per cent) and methanolic Combine all the filtrates and concentrate to 50.0 ml.
sulphuric acid (5 per cent) before spraying. Heat at 120º for Reference solution. A 0.01 per cent w/v solution of
10 minutes and examine the plate in day light. The shatavarin IV RS in methanol. Dilute suitably to prepare
chromatographic profile of the test solution is similar to that 0.0075- 0.075 per cent w/v solution.
of the reference solution.
Chromatographic system
Tests – a stainless steel column 25 cm x 4.6 mm packed with
octadecylsilane bonded to porous silica (5 µm),
Foreign organic matter (2.6.1). Not more than 2.0 per cent. – mobile phase: a mixture of 60 volumes of acetonitrile
Ethanol-soluble extractive (2.6.2). Not less than 15.0 per cent. and 40 volumes of water,
Water- soluble extractive (2.6.3). Not less than 20.0 per cent – flow rate. 1 ml per minute,
by Method I. – use evaporative light scattering detector,
– temperature
Ash (2.3.19). Not more than 15.0 per cent. evaporator 110º,
Acid-insoluble ash (2.3.19). Not more than 3.0 per cent. nebulizer 90º,
Heavy metals (2.3.13). 1.0 g complies with the limit test for nebulizer gas nitrogen and gas flow 1 SLM,
heavy metals, Method B (20 ppm). – a 20 µl loop injector.

Loss on drying (2.4.19). Not more than 15.0 per cent, Inject the reference solution. The test is not valid unless the
determined on 5 g by drying in an oven at 105º. regression coefficient is not more than 0.9.
Microbial contamination (2.2.9). Complies with the microbial Inject the test solution and reference solution.
contamination tests. Calculate the content of shatavarin IV.
Assay. Perform the Assay by following method I or by method Storage. Store protected from heat, moisture and against attack
II. The result of the method II will be official in case of dispute. by insects and rodents.

1428
IP 2007 SHATI

Shati and filter. Reflux the residue further with 2 × 25 ml of methanol,


cool and filter. Combine all the filtrates and concentrate under
Hedychium vacuum to 25 ml.
Reference solution. Reflux 0.5 g of the shati RS with 25 ml of
methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 × 25 ml of methanol, cool and filter. Combine all
the filtrates and concentrate under vacuum to 12.5 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with a anisaldehyde solution. Heat at 110° for 10 minutes
and examine the plate in day light. The chromatographic profile
of the test solution is similar to that of the reference solution.

Tests
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 0.7 per cent.
Shati consists of the dried rhizomes of Hedychium spicatum Water-soluble extractive (2.6.3). Not less than 12 per cent by
Buch.-Ham.ex Smith (Fam. Zingiberaceae). method I.
Shati contains not less than 0.80 per cent of p-methoxy Ash (2.3.19). Not more than 8 per cent.
cinnamic acid ethyl ester, calculated on the dried basis. Acid-insoluble ash (2.3.19). Not more than 3.0 per cent.
Description. A reddish-brown outer surface and white in side, Heavy metals (2.3.13). 1.0 g complies with the limit test for
short and uneven fracture and camphoraceous odour with heavy metals, Method B (20 ppm).
aromatic and pungent taste.
Loss on drying (2.4.19). Not more than 14.0 per cent,
Identification determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
A. Macroscopic — Tuberous rhizome having reddish brown,
contamination tests.
rough outer surface with round root scars or rootlets which
remain attached at some places. Transversely sliced pieces of Assay. Determine by liquid chromatography (2.4.14).
dried rhizome are spherical, flat, 1 cm in thickness and 2-3 cm Test solution. Reflux 2 g of the coarsely powdered substance
in diameter having white and starchy surface. under examination with 50 ml of methanol on a water-bath for
B. Microscopic — Transverse section of rhizome shows 15 minutes, cool and filter. Reflux the residue further with
outermost layer of cork having 2-6 layers of isodiametic, methanol till the extract turns colourless, cool and filter.
nonlignified and suberised cells which are radially arranged. Combine all the filtrates and concentrate to a volume slightly
In old rhizome, the cork cells are exfoliated or crushed. Cortex less than 100 ml. Dilute to 100.0 ml with methanol.
is a broad zone with 20-25 layers of thin walled parenchymatous Reference solution. A 0.01 per cent w/v solution of
cells. Cortex region is filled with abundant starch grains and p-methoxy cinnamic acid ethyl ester RS in methanol.
numerous oleo-resin cells. Vascular bundles are closed and
collateral and scattered throughout the ground tissues. Chromatographic system
Cambium has 4-5 rows of tangentially elongated cells. It forms – a stainless steel column 25 cm × 4.6 mm packed with
a complete ring between the xylem and phloem groups. Starch octadecylsilane bonded to porous silica (5 µm),
grains tissue are simple, circular or oval in shape, found in – mobile phase: 40 volumes of water and 60 volumes of
ground cells. acetonitrile,
– flow rate. 1 ml per minute,
C. Determine by thin-layer chromatography (2.4.17), coating – spectrophotometer set at 310 nm,
the plate with silica gel G. – a 20 µl loop injector.
Mobile phase. A mixture of 80 volumes of n-haxane and Inject the reference solution. The test is not valid unless the
20 volumes of acetone. relative standard deviation for the replicate injections is not
Test solution. Reflux 1 g of the coarsely powdered substance more than 2.0 per cent.
under examination with 25 ml of methanol for 15 minutes, cool Inject the test solution and reference solution.

1429
SHELLAC IP 2007

Calculate the content of p-methoxy cinnamic acid ethyl ester. Arsenic ( 2.3.10). Heat gently 5.0 g with 2 ml of nitric acid and
Storage. Store protected from heat, moisture and against attack 0.5 ml of sulphuric acid in a long-necked flask, until the first
by insects and rodents. reaction has subsided, cool, add carefully and in small portions,
15 ml of nitric acid and 6 ml of sulphuric acid taking care to
avoid excessive foaming, and continue heating, adding further
small portions of nitric acid, if necessary, until white fumes
Shellac are evolved and the solution becomes colourless or almost
colourless. Cool, add carefully 10 ml of water, evaporate until
Lac
white fumes are evolved, and repeat the addition of water and
Shellac consists of a resinous substance prepared from a evaporation until all the nitric acid has been removed, cool,
secretion that encrusts the bodies of a scale insect, Laccifer dilute to 50 ml with water, and add 10 ml of stannated
lacca Kerr (Fam. Ciccidae). hydrochloric acid AsT. The resulting solution complies with
Description. Lemon-yellow to brownish orange thin scales or the limit test for arsenic (2 ppm). Use 0.5 ml of arsenic standard
hard, brittle masses; odourless or with a faint odour. solution (10 ppm As) for the standard stain.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
Identification heavy metals, Method B (20 ppm).
To 50 mg add a few drops of a mixture of 1 g of ammonium Sulphated ash (2.3.18). Not more than 1.0 per cent, determined
molybdate and 3 ml of sulphuric acid; a green colour is on 0.5 g.
produced and it becomes lilac on standing for 5 minutes.
Storage. Store protected from moisture.
Tests
Acid value (2.3.23). 50 to 70, determined by the following
method. Weigh accurately about 2.0 g and dissolve with the Starch
aid of gentle heat, in 50 ml of ethanol (95 per cent) previously Starch consists of polysaccharide granules obtained from the
neutralised to ethanolic thymol blue solution. Titrate with caryopsis of maize or corn, Zea mays Linn., or of rice, Oryza
0.1 M ethanolic potassium hydroxide using ethanolic thymol sativa Linn., or of wheat, Triticum aestivum Linn., or from the
blue solution as an external indicator. Calculate the acid value tuber of potato, Solanum tuberosum Linn., or from the rhizomes
from the expression of tapioca, Manihot utilissima Pohl.
5.61 × a /w Description. A very fine, white or slightly yellowish powder
where, a = number of ml of 0.1 M ethanolic potassium or irregular white masses which are readily reducible to powder,
hydroxide and creaks when pressed between the fingers; odourless and
w = weight, in g, of the sample. tasteless. The presence of granules showing cracks or edge
irregularities is exceptional in starches other than wheat starch;
Ethanol-insoluble matter. Not more than 2.0 per cent, wheat starch may contain granules with cracks on the edges.
determined by the following method. Weigh accurately about
5.0 g in an extraction thimble, cover with ethanol (95 per Identification
cent) and allow to stand for 16 hours. Place in an apparatus
for the continuous extraction of drugs, extract with ethanol A. Corn or maize starch — Polyhedral granules, 2 to 23 µm in
(95 per cent) for 4 hours, dry the residue at 100º for 3 hours size, or rounded granules, 25 to 32 µm in diameter. The central
and weigh. hilum consists of a distinct cavity or two- to five-rayed cleft;
no concentric striations. Viewed between crossed nicol prisms,
Colophony. Dissolve 2.0 g by shaking with 10 ml of ethanol, a distinct black cross is seen intersecting at the hilum.
add slowly, with shaking 50 ml of light petroleum (40º to 60º),
wash with two successive portions, each of 50 ml, of water, Potato starch — Single granules, either irregular, ovoid or
filter the washed light petroleum solution, and evaporate to pear-shaped, 30 to 100 µm in size, or rounded, 10 to 35 µm in
dryness; to the residue add 2 ml of a mixture of 1 volume of size; compound granules consisting of groups of two to four
liquified phenol and 2 volumes of carbon tetrachloride and elements are rare. Eccentric hilum; clearly visible concentric
transfer to a cavity of a colour-reaction porcelain tile; fill an striations. Viewed between crossed nicol prisms, a distinct
adjacent cavity with a mixture of 1 volume of bromine and black cross is seen intersecting at the hilum.
4 volumes of carbon tetrachloride, and cover both cavities Rice starch — Polyhedral granules, 2 to 5 µm in size, either
with an inverted watch-glass; no purple or deep indigo-blue isolated or aggregated in ovoid masses, 10 to 20 µm in size.
colour is produced in the liquid containing the residue. Central hilum poorly visible; no concentric striations. Viewed

1430
IP 2007 SUNTHI

between crossed nicol prisms, a distinct black cross is seen Storage. Store protected from light and moisture.
intersecting at the hilum. Labelling. The label states the type of starch.
Tapioca starch — Principally simple granules, sub-spherical,
muller-shaped or rounded polyhedral; smaller granules 5 to
10 µm, larger granules 20 to 35 µm in diameter; hilum, central,
punctate, linear or triradiate; striations, faint, concentric; Sunthi
compound granules, few, of two to three unequal components. Saunth; Ginger
Wheat starch — Large discoid or, more rarely, reniform
granules, 10 to 45 µm in size; profile, elliptical and fusiform, slit
along the main axis. Small rounded or polyhedral granules,
2 to 10 µm in size. Granules of intermediate size very rarely
occur. Hilum and striations invisible or barely visible. Viewed
between crossed nicol prisms, a distinct black cross is seen
intersecting at the hilum.
B. Heat to boiling for 1 minute a suspension of 1 g in 50 ml of
water and cool; a thin and cloudy mucilage is produced with
all starches except potato starch which gives a thick and more
transparent mucilage.
C. To 10 ml of the mucilage obtained in test B add 0.05 ml of
0.01 M iodine; a dark blue colour is produced, which
disappears on heating and reappears on cooling.

Tests Sunthi is the whole or cut scraped or unscraped, dried rhizomes


of Zingiber officinale Rosc. (Fam. Zingiberaceae).
Acidity. Add 10.0 g to 100 ml of ethanol (70 per cent)
previously neutralised to phenolphthalein solution, shake Sunthi contains not less than 0.8 per cent of total gingerols,
for 1 hour, filter and titrate 50 ml of the filtrate with 0.1 M calculated on the dried basis.
sodium hydroxide. Not more than 2.0 ml is required to change Description. Odour, agreeable and aromatic; taste, agreeable
the colour of the solution. and pungent.
Iron (2.3.14). Dissolve the residue obtained in the test for
sulphated ash in 4 ml of hydrochloric acid with the aid of Identification
gentle heat, dilute with water to 50 ml and mix; 25 ml of the A. Macroscopic — Rhizome laterally compressed, bearing
resulting solution complies with the limit test for iron (40 ppm). short, flattened, oblique branches; outer surface buff-coloured,
Fluorescence. No fluorescence should be visible under longitudinally striate; inner surface pale yellow, starchy and
screened ultra-violet light. fibrous. Fracture short with projecting fibers.
Oxidising substances. To 5.0 g add 10 ml of water and 1 ml of B. Microscopic — Fibro-vascular bundles and oleoresin cells
acetic acid and stir until a homogeneous suspension is with yellow pigment scattered in ground tissue. Starch grains
obtained. Add 0.5 ml of a freshly prepared saturated solution abundant in parenchyma cells, mostly simple, sack shaped,
of potassium iodide, mix and allow to stand for 5 minutes; no spherical; hilum eccentric.
distinct brown or blue colour is observed. C. Determine by thin-layer chromatography (2.4.17), coating
Microbial Contamination (2.2.9). 1 g is free from Escherichia the plate with silica gel G..
coli and salmonellae. Mobile phase. A mixture of 30 volumes of hexane and
Sulphated ash (2.3.18). Not more than 0.6 per cent (for all 70 volumes of diethyl ether.
starches except rice starch) and not more than 0.8 per cent (for Test solution. Reflux 1 g of the coarsely powdered substance
rice starch), determined on 2.0 g. under examination with 25 ml of methanol for 15 minutes, cool
Loss on drying (2.4.19). Not more than 15.0 per cent (for all and filter. Wash the residue with 10 ml of methanol. Combine
starches except potato starch) and not more than 20.0 per cent all the filtrates and concentrate to 10 ml.
(for potato starch), determined on 0.2 g by drying in an oven Reference solution. Reflux 0.5 g of coarsely powdered
at 105º. sunthi RS with 5 ml methanol for 15 minutes, cool and filter.

1431
SUNTHI IP 2007

Apply to the plate 10 µl of each solution as bands 10 mm by Tolu Balsam


2 mm. Allow the mobile phase to rise 8 cms. Dry the plate in air,
spray with 1 per cent w/v vanillin in ethanol (95 per cent) Balsam of Tolu
followed by 10 per cent w/v ethanolic sulphuric acid. Heat at Tolu Balsam is a solid or semi-solid, balsam obtained by incision
100º for 5-10 minutes and examine the plate in day light. The from the trunk of Myroxylon balsamum (Linn.) Harms (Fam.
chromatographic profile of the test solution is similar to that Leguminosae).
of the reference solution.
Tolu Balsam contains not less than 35.0 per cent and not more
Tests than 50.0 per cent of total balsamic acids, calculated as
cinnamic acid, C9H8O2, on the dry, ethanol-soluble matter.
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Description. A soft, tenacious, brownish yellow or brown
Ethanol-soluble extractive (2.6.2). Not less than 2.0 per cent. mass, when first collected; subsequently, becoming harder
Water-soluble extractive (2.6.3). Not less than 10.0 per cent and finally brittle. Transparent in thin films; odour, aromatic
by Method I. and vanilla-like. Warmed and pressed between pieces of glass
Ash (2.3.19). Not more than 8.0 per cent. and examined with a lens, it exhibits crystals of cinnamic acid.

Acid-insoluble ash (2.3.19). Not more than 1.5 per cent. Identification
Heavy metals (2.3.13). 1.0 g complies with the limit test for A. To a solution in ethanol (90 per cent) add ferric chloride
heavy metals, Method B (20 ppm). test solution; a green colour is produced.
Water (2.3.43). Not more than 12.0 per cent, determined on B. To 1 g add to 5 ml of water, heat to boiling, filter, add 30 mg
0.2 g. of potassium permanganate and continue heating; the odour
Microbial contamination (2.2.9). Complies with the microbial of benzaldehyde is produced.
contamination tests.
Tests
Assay. Determine by liquid chromatography (2.4.14).
Acidity. A solution in ethanol (90 per cent) is acidic to litmus
Test solution. Reflux about 3 g of the coarsely powdered
solution.
substance under examination with 100 ml of methanol on a
water–bath for 15 minutes cool and filter. Reflux the residue Acid value (2.3.23). 97 to 160, calculated on the dry, ethanol-
further with methanol till the last extract turns colorless, cool soluble matter, determined by the following method. Dissolve
and filter. Combine all the filtrates and concentrate to 50.0 ml 5.0 g in 50 ml of boiling ethanol (90 per cent), add 3 ml of
phenolphthalein solution and titrate the hot solution with
Reference solution. A 0.1 per cent w/v solution of
1 M ethanolic potassium hydroxide. When the colour
6-gingerol RS in methanol.
becomes dark brown, attach to a reflux condenser, boil for a
Chromatographic system few minutes to break up the precipitate and complete the
– a stainless steel column 25 cm x 4.6 mm packed with titration.
octadecylsilane bonded to porous silica (5 µm),
Ethanol-insoluble matter. Not more than 5 per cent, determined
– mobile phase: 55 volumes of acetonitrile and 45 volumes
by the following method. Digest 2.5 g with 50 ml of ethanol
of water,
(90 per cent), filter through a sintered glass crucible, transfer
– flow rate. 1.3 ml per minute,
the residue to the filter crucible with the aid of more ethanol
– spectrophotometer set at 278 nm,
(90 per cent), wash with hot ethanol (90 per cent) until all
– a 20 µl loop injector.
soluble matter is removed and dry to constant weight at 100º.
Inject the reference solution. The test is not valid unless the Colophony. Add 5 g to 25 ml of carbon disulphide, warm gently
relative standard deviation for the replicate injections is not on a water-bath under a reflux condenser, filter, evaporate the
more than 2.0 per cent. solution to dryness, dissolve the residue in 6 ml of light
Inject the test solution and reference solution. petroleum (40º to 60º) and shake with 10 ml of a 0.5 per cent
Calculate the contents of total gingerols by summing the peak w/v solution of cupric acetate; the light petroleum layer is
areas of 6-gingerol with all other peaks, which elute after 6- not coloured green.
gingerol and have a peak area of at least 5 per cent of the peak Ester value (2.3.26). 47 to 95, calculated on the dry, ethanol-
area of 6-gingerol. soluble basis.
Storage. Store protected from heat, moisture and against attack Saponification value. (2.3.37). 170 to 230, calculated on the
by insects and rodents. dry, ethanol-soluble basis.

1432
IP 2007 TRAGACANTH

Loss on drying (2.4.19). Not more than 4 per cent, determined concentric transverse ridges; white, translucent, horny;
on 2.0 g by drying in a thin layer in an oven at 60º over fracture, short. May also be in the form of thicker, less brittle
phosphorus pentoxide at a pressure not exceeding 2.7 kPa. pieces, white to pale yellow and more opaque.
Assay. Weigh accurately about 2.0 g and boil with 25 ml of B. Microscopic — Reduce to powder. Examine under a
dilute ethanolic potassium hydroxide solution under a reflux microscope; the powder shows in the gummy mass numerous
condenser for 1 hour. Remove the ethanol and digest the stratified cellular membranes which turn violet on the addition
residue with 50 ml of hot water until diffused. Cool the liquid, of iodinated zinc chloride solution. The mass includes starch
add 150 ml of water and 1.5 g of magnesium sulphate granules, isolated or in small clusters, rounded or occasionally
dissolved in 50 ml of water. Mix thoroughly and set aside for deformed, diameter 4 to 10 µm, and up to 20 µm, with a central
10 minutes. Filter, wash the residue on the filter with 20 ml of hilum, visible in polarised light.
water, acidify the combined filtrate and washings with C. Moisten 0.5 g of the powdered material with 1 ml of ethanol
hydrochloric acid and extract with successive quantities of (95 per cent) and add gradually, while shaking, 50 ml of water
50, 40, 30, 30 and 30 ml of ether. Combine the ether extracts and until a homogeneous mucilage is obtained. To 5 ml of the
discard the aqueous portion. Extract with successive mucilage add 5 ml of water and 2 ml of barium hydroxide
quantities of 20, 20, 10, 10 and 10 ml of sodium bicarbonate solution. A slightly flocculent precipitate is formed which,
solution, washing each aqueous extract with the same 20 ml when heated for 10 minutes on a water-bath, gives an intense
of ether. Discard the ether layers, acidify the combined yellow colour.
aqueous extracts with hydrochloric acid and extract with
successive quantities of 30, 20, 20 and 10 ml of chloroform, D. Add 4 ml of a 0.5 per cent w/v dispersion in water to 0.5 ml
filtering each chloroform extract through a plug of cotton wool of hydrochloric acid and heat on a water-bath for 30 minutes.
on which a layer of anhydrous sodium sulphate is placed. To one half of the resulting liquid add 1.5 ml of sodium
Evaporate the chloroform on a water-bath until about 10 ml hydroxide solution and 3 ml of alkaline cupric-tartrate
remains and remove the remainder in a current of air stopping solution and heat on a water-bath; a reddish brown precipitate
immediately when the last trace of solvent is removed. Dissolve is formed. To the other half of the liquid, add a few drops of
the residue by warming with 10 ml of ethanol (95 per cent), barium chloride solution; no precipitate is formed (freedom
previously neutralised to phenol red solution, cool and titrate from agar).
with 0.1 M sodium hydroxide using phenol red solution as
Tests
indicator.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.01482 g of Acacia and other soluble gums. To 20 ml of a 2.5 per cent w/v
total balsamic acids, calculated as cinnamic acid, C9H8O2. suspension of the powdered material prepared with freshly
boiled water add 10 ml of lead acetate solution; a flocculent
Storage. Store protected from light and moisture. Avoid precipitate is formed. Filter and add to the filtrate 10 ml of lead
exposure to excessive heat. subacetate solution; a slight cloudiness may appear, but there
is no precipitate.
Karaya gum. Boil 1 g with 20 ml of water until a mucilage is
Tragacanth formed, add 5 ml of hydrochloric acid and again boil for
5 minutes; no pink or red colour develops.
Tragacanth is the air-hardened gummy exudate, flowing
naturally or obtained by incision, from the trunk and branches Sterculia. A. Shake 0.2 g of the powdered material with 10 ml
of Astragalus gummifer Labill. and certain other species of of ethanol (60 per cent) in a 10-ml stoppered cylinder; any
Astragalus. gel formed occupies not more than 1.5 ml.
Description. Pale yellow, thin, flattened ribbons or brittle B. Shake 1 g of the powdered material with 100 ml of water and
pieces; odourless and almost tasteless. On the addition of titrate with 0.01 M sodium hydroxide, using methyl red
about 10 times its weight of water, it forms a mucilaginous gel. solution as indicator. Not more than 5.0 ml is required to change
the colour of the solution.
It has the macroscopic and microscopic characteristics
described under Identification tests A and B. Foreign matter. Not more than 1.0 per cent, determined by the
following method. To 2.0 g of the powdered material in a 250-
Identification ml round-bottomed flask add 95 ml of methanol, swirl to
moisten the powder and add 60 ml of 7 M hydrochloric acid.
A. Macroscopic — Occurs as thin, flattened pieces, 30 mm Add a few glass beads and heat under a reflux condenser in a
long, 10 mm wide and up to 1 mm in thickness, more or less water-bath for 3 hours, shaking occasionally. Remove the glass
curved, marked on the surface by fine longitudinal striae and beads and filter the hot suspension under reduced pressure

1433
TULASI IP 2007

through a sintered-glass crucible (porosity No. 1). Rinse the 5-7 mm in length. It has both male and female parts. Calyx:
flask with a small quantity of water, passing the rinsings There are 5 sepals and it is greenish in colour. Corolla: There
through the filter. Wash the residue on the filter with about are 5 petals, bilabiate in shape and covered with scattered
40 ml of methanol and dry to constant weight at 110º. hairs. Petals whitish-purple.
Arsenic (2.3.10). Mix 3.3 g with 3 g of anhydrous sodium B. Microscopic — Transverse section of leaf shows a pot
carbonate, add 10 ml of bromine solution and mix thoroughly. shaped midrib. Upper epidermis consists of a layer of small,
Evaporate to dryness on a water-bath, gently ignite, and quadrangular transparent cells with thin walls and thin smooth
dissolve the cooled residue in 16 ml of brominated cuticle. On tangential view, these cells are polygonal with
hydrochloric acid AsT and 45 ml of water. Remove the excess straight or wavy walls. Lower epidermis consists of a layer of
of bromine with 2 ml of stannous chloride AsT. The resulting small, quadrangular transparent cells with thin walls and thin
solution complies with the limit test for arsenic (3 ppm). and thin smooth cuticle. Trichomes bent, consisting of 2-6 of
Heavy metals (2.3.13). 0.5 g complies with the limit test for 1 stalk cell and 2-4 cells with rounded heads. Palisade
heavy metals, Method B (40 ppm). parenchyma consists of layer of long cylindrical cells
containing chlorophyll; spongy parenchyma consists of
Ash (2.3.19). Not more than 4.0 per cent, determined on 1.0 g.
polygonal cells with thin, straight or slightly wavy side walls.
Microbial contamination (2.2.9). 1 g is free from Escherichia Vascular bundles collateral type. Stomata diacytic.
coli and 10 g is free from salmonellae.
C. Determine by thin-layer chromatography (2.4.17), coating
Storage. Store protected from moisture. the plate with silica gel G.
Mobile phase. A mixture of 97 volumes of toluene and
3 volumes of ethyl acetate.
Tulasi Test solution. Reflux 2 g of the coarsely powdered substance
Ocimum, Basil under examination with 25 ml of methanol for 15 minutes, cool
and filter. Reflux the residue further with 2 × 25 ml of methanol,
cool and filter. Combine all the filtrates and concentrate under
vacuum to 10 ml.
Reference solution. Reflux 1 g of tulasi RS with 25 ml of
methanol for 15 minutes, cool and filter. Reflux the residue
further with 2 × 25 ml of methanol, cool and filter. Combine all
the filtrates and concentrate under vacuum to 5 ml.
Apply to the plate 10 µl of each solution as bands 10 mm by
2 mm. Allow the mobile phase to rise 8 cm. Dry the plate in air,
spray with a anisaldehyde solution. Heat at 110° for 10 minutes
and examine the plate in day light. The chromatographic profile
of the test solution is similar to that of the reference solution.

Tests
Tulasi consists of leaves of Ocimum sanctum Linn (Fam.
Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Lamiaceae).
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent.
Tulasi contains not less than 0.40 per cent of eugenol,
calculated on the dried basis. Water-soluble extractive (2.6.3). Not less than 10.0 per cent
by method I.
Description. Greyish-black in colour having characteristic
odour with slightly pungent and aromatic taste. Ash (2.3.19). Not more than 15.0 per cent.

Identification Acid-insoluble ash (2.3.19). Not more than 5.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
A. Macroscopic — Leaves simple, elliptic, 2.7-7.5 cm long,
heavy metals, Method B (20 ppm).
1-3 cm wide, with acute top, cuneate, obtuse to rounded base,
margin entire, undulate or serrate, both surfaces thinly Loss on drying (2.4.19). Not more than 12.0 per cent,
pubescent and dotted; petiole 0.2-3.0 cm long. Flowers are determined on 5 g by drying in an oven at 1050.

1434
IP 2007 VASAKA

Microbial contamination (2.2.9). Complies with the microbial Vasaka contains not less than 0.6 per cent of vasicine,
contamination tests. calculated on the dried basis.
Assay. Determine by gas chromatography (2.4.13). Description.Taste, bitter.
Test solution. Reflux 0.5 g of the coarsely powdered substance Identification
under examination with 50 ml of methanol on a water-bath for
15 minutes, cool and filter. Reflux the residue further with A. Macroscopic — Leaf pieces membranous, brittle, greyish-
methanol till the extract turns colourless, cool and filter. brown, a few pieces green coloured. Floral bracts leaf-like.
Combine all the filtrates and concentrate to a volume slightly
B. Microscopic — Stomata diacytic, more on the lower
less than 100 ml. Dilute to 100.0 ml with methanol.
epidermis; glandular and non-glandular hair on both surfaces
Reference solution. A 0.004 per cent w/v solution of of leaf; elongated cystoliths present in palisade cells.
eugenol RS in methanol.
C. Determine by thin-layer chromatography (2.4.17), coating
Chromatographic system the plate with silica gel G..
– a capillary column 30 m x 0.25 x 0.25 mm coated with100
Mobile phase. A mixture of 8 volumes of ethyl acetate,
per cent dimethylpolysiloxane
2 volumes of methanol and 0.2 volume of strong ammonia
– temperature:
solution.
oven 60° to 260° @10° per minute, (Initially and finally
hold for 5 minutes respectively) Test solution. Reflux 1 g of coarsely powdered substance under
Injector 240°, examination with 50 ml methanol for 15 minutes, cool and
detector 280°, filter. Reflux the residue further with 2 × 50 ml of methanol,
– flow rate. 0.8 ml per minute, cool and filter. Combine all the filtrates and concentrate to
– split flow 20 ml per minute. 10 ml.
Inject 1 µl of the reference solution. The test is not valid Reference solution. Reflux 0.5 g of vasaka RS with 50 ml
unless the relative standard deviation for the replicate methanol for 15 minutes, cool and filter. Reflux the residue
injections is not more than 2.0 per cent. further with 2 × 50 ml of methanol, cool and filter. Combine all
the filtrates and concentrate to 5 ml.
Inject 1 µl of the test solution and reference solution.
Apply to the plate 10 µl of each solution as bands 10 mm by
Calculate the content of eugenol.
2 mm. Allow the mobile phase to rise 8 cms. Dry the plate in air
Storage. Store protected from heat, moisture and against attack and spray with dragendorf’s reagent. The chromatographic
by insects and rodents. profile of the test solution is similar to that of the reference
solution.
Vasaka Tests
Adulasa; Adhatoda Foreign organic matter (2.6.1). Not more than 2.0 per cent.
Ethanol-soluble extractive (2.6.2). Not less than 3.0 per cent.
Water-soluble extractive (2.6.3). Not less than 22 per cent by
Method I.
Ash (2.3.19). Not more than 21 per cent.
Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
Heavy metals (2.3.13). 1.0 g complies with the limit test for
heavy metals, Method B (20 ppm).
Loss on drying (2.4.19). Not more than 12.0 per cent,
determined on 5 g by drying in an oven at 1050.
Microbial contamination (2.2.9). Complies with the microbial
contamination tests.
Assay. Determine by liquid chromatography (2.4.14).
Vasaka consists of the dried mature leaves of Adhatoda vasica Test solution. Reflux about 2 g of the coarsely powdered
Nees. (Fam. Acanthaceae). substance under examination with 50 ml of methanol on a

1435
YASTI IP 2007

water bath for 15 minutes, cool and filter. Reflux the residue Identification
further with methanol till the last extract turns colorless, cool
and filter. Combine all the filtrates and concentrate to 100.0 ml. A. Macroscopic — Root with few branches, up to 1 m long
and 0.5 to 3 cm in diameter. Bark, brownish-grey to brown with
Reference solution. Dissolve 25 mg of vasicine hydrochloride longitudinal striations, bearing traces of lateral roots. Stolons,
RS in 50 ml of methanol. Dilute 5.0 ml of this solution to 50.0 ml cylindrical, 1 to 2 cm in diameter and up to several metres long,
with methanol. but may be cut into lengths of 10 to 15 cm; similar in external
Chromatographic system appearance to the root but with occasional small buds. Fracture
– a stainless steel column 25 cm x 4.6 mm packed with of the root and stolon, granular and fibrous. Cork layer, thin;
octadecylsilane bonded to porous silica (5 µm), secondary phloem region, wide, light yellow with radial
– mobile phase: a mixture of 3 volumes of the solution striations; xylem, compact, yellow, with radiate structure. The
prepared by dissolving 1 g of sodium hexane- stolon has a central pith which is absent from the root.
sulphonate in 1000 ml of water, 1 volume of acetonitrile
B. Microscopic — Cork and phelloderm are narrow. Phloem
and 20 volumes of glacial acetic acid,
consisting of bundles of thick-walled, yellow fibres with narrow
– flow rate. 1 ml per minute,
lumina surrounded by cells each containing a calcium oxalate
– spectrophotometer set at 300 nm,
prism, alternating in the external layers with areas of strongly
– a 20 µl loop injector.
hyaline keratenchyma; functional sieve tissue near the
Inject the reference solution. The test is not valid unless the cambium. Medullary rays parenchymatous, widening towards
relative standard deviation for the replicate injections is not the exterior, 3 to 12 cells wide. Xylem composed of radial rows
more than 2.0 per cent. of tracheids and vessels alternating with bundles of lignified
Inject the test solution and reference solution. fibres with crystal sheaths similar to those of the secondary
phloem; vessels 30 µm to 150 µm in diameter with thick walls
Calculate the content of vasicine. (5 µm to 10 µm) having reticulate thickenings or numerous
Storage. Store protected from heat, moisture and against attack bordered pits with slit-shaped openings associated with
by insects and rodents. lignified xylem parenchyma. Medullary rays, 2 to 5 cells wide.
Parenchymatous cells throughout containing simple, round,
oval or fusiform starch granules 2 µm to 20 µm, mostly 5 µm to
Yasti 12 µm, in diameter; parenchymatous pith present solely in the
stolon.
Liquorice root; Mulethi; Glycyrrhiza
C. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
Mobile phase. The upper layer, even if turbid, of a mixture of
60 volumes of ethyl acetate, 27 volumes of 1 M ammonia and
13 volumes of ethanol, shaken together and allowed to stand
for 5 minutes.
Test solution. Shake 1 g, in No. 180 powder, with 20 ml of
chloroform for 15 minutes, filter and reserve the extracted
powder for the preparation of reference solution (a). Evaporate
the filtrate to dryness and dissolve the residue in 2 ml of a
mixture of equal volumes of chloroform and methanol.
Reference solution (a). Add to the extracted powder 30 ml of
0.5 M sulphuric acid and heat under a reflux condenser for
1 hour, allow to cool and extract with two quantities, each of
20 ml, of chloroform; dry the combined chloroform extracts
with anhydrous sodium sulphate, filter, evaporate to dryness
Yasti consists of the dried, unpeeled roots and stolons of
and dissolve the residue in 2 ml of a mixture of equal volumes
Glycyrrhiza glabra Linn. (Fam. Leguminoseae).
of chloroform and methanol.
Yasti contains not less than 3.0 per cent of glycyrrhizinic acid.
Reference solution (b). Dissolve 10 mg of glycyrrhetinic acid
Description. Odour, characteristic and slightly aromatic; taste, in 2 ml of a mixture of equal volumes of chloroform and
very sweet and faintly astringent; the bark is not bitter. methanol.

1436
IP 2007 YASTI

Apply to the plate 10 µl of test solution, reference solution (a) filtrate to dryness on a water-bath, dry the residue at 105º and
and 20 µl of reference solution (b) as bands 20 mm by 3 mm. weigh.
After development, dry the plate in air for 5 minutes and Acid-insoluble ash (2.3.19). Not more than 2.0 per cent.
examine in ultraviolet light at 254 nm. The chromatogram
obtained with reference solution (b) exhibits a band Sulphated ash (2.3.18). Not more than 10.0 per cent.
corresponding to b-glycyrrhetinic acid with an Rf value of Assay. Determine by liquid chromatography (2.4.14).
about 0.1. The chromatogram obtained with reference solution Test solution. Weigh accurately about 1.0 g of the coarsely
(a) exhibits a corresponding band but this is not seen in the powdered substance under examination in a 250-ml conical
chromatogram obtained with the test solution. Spray the plate flask, add 100 ml of 0.1 M ammonia and mix with the aid of
with anisaldehyde solution, using about 10 ml for a plate, ultrasound for 30 minutes. Centrifuge a part of the supernatant
20 cm × 20 cm in size, heat at 105º for 10 minutes and examine liquid and dilute 1.0 ml to 5.0 ml with 0.1 M ammonia. Filter the
in daylight. The b-glycyrrhetinic acid bands become violet- solution through a membrane filter disc with an average pore
blue. One or two bands with an Rf value of about 0.6, visible in diameter not greater than 1.0 µm and use the filtrate.
daylight before spraying, become orange-yellow and several
Reference solution. A 0.005 per cent w/v solution of
other violet-blue bands appear in the chromatograms obtained
glycyrrhizinic acid RS in 0.1 M ammonia.
with the test solution and reference solution (a). The band
corresponding to b-glycyrrhetinic acid in the chromatogram Chromatographic system
obtained with reference solution (a) is at least equal in size to – a stainless steel column 25 cm x 4.6 mm, packed with
the band in the chromatogram obtained with reference solution octadecylsilane bonded to porous silica (5 µm),
(b). – mobile phase: a mixture of 6 volumes of glacial acetic
acid, 30 volumes of acetonitrile and 64 volumes of water,
D. Mix a small quantity, in powder, with 0.05 ml of sulphuric – flow rate. 1.5 ml per minute,
acid; the powder particles become orange-yellow and some – spectrophotometer set at 254 nm,
fragments change, more slowly, to pinkish red. – a 20 µl loop injector.
Tests Inject the reference solution. The test is not valid unless the
relative standard deviation for replicate injections is not more
Curcuma. When examined under a microscope, none of the than 2.0 per cent.
fragments of the powder in sulphuric acid (see Identification
test D) should immediately take on a carmine-red colour. Inject the test solution and the reference solution and measure
the responses for the analyte peak.
Water-soluble extractive (2.6.3). Not less than 20 per cent,
determined by the following method. Mix 2.5 g of the finely Calculate the content of glycyrrhizinic acid.
powdered drug with 50 ml of water and allow to stand for Storage. Store protected from light and moisture.
2 hours, shaking frequently. Filter, evaporate 10.0 g of the

1437
YASTI IP 2007

1438
IP 2007

1439
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

BLOOD AND BLOOD-RELATED PRODUCTS

Anti-A Blood Grouping Serum ........................................................................................


Anti-B Blood Grouping Serum ..........................................................................................
Anti-D (Rho) Immunoglobulin ..........................................................................................
Anti-D Immunoglobulin Human for Intravenous Use ..........................................................
Anti-Human Globulin Serum ................................................................................................
Blood Grouping Serums Anti-D, Anti-C, Anti-E, Anti-c, Anti-e ...........................................
Concentrated Human Red Blood Cells ...............................................................................
Cryoprecipitated Antihemophilic Factor ..............................................................................
Dried Human Antihaemophilic Fraction ...............................................................................
Fibrin Sealant Kit ..............................................................................................................
Human Albumin ................................................................................................................
Human Coagulation Factor IX ..........................................................................................
Human Coagulation Factor VII ........................................................................................
Human Coagulation Factor VIII (rDNA) ............................................................................
Human Normal Immunoglobulin .........................................................................................
Human Plasma Protein Fraction ..........................................................................................
Human Prothrombin Complex ............................................................................................
Normal Immunoglobulin for Intravenous Use ......................................................................
Plasma for Fractionation .........................................................................................
Platelet Concentrate ...........................................................................................................
Whole Human Blood ..........................................................................................................

1441
IP 2007 ANTI-A BLOOD GROUPING SERUM

Anti-A Blood Grouping Serum Storage. Store at a temperature between 2° and 8°.

Anti-A Blood Grouping Serum is a sterile, liquid or dried Labelling. Label states that the source material was not reactive
preparation containing the particular blood group antibodies for hepatitis B surface antigen, but that no known test method
derived from high-titered blood plasma or serum of human offers assurance that products derived from human blood will
subjects, with or without stimulation by the injection of Blood not transmit hepatitis in case of polyclonal. Label also states
Group Specific Substance A (or AB). It contains a suitable that it is for in vitro diagnostic use.
antimicrobial preservative. It is of two types polyclonal &
monoclonal. NOTE—The labeling is in black lettering imprinted on paper
that is white or is coloured completely or in part to match
Production the specified blue colour standard.
The monoclonal antibody technique devised by Kohler and
Milsten has proved useful in producing high titre and specific
antibodies. Laboratory animals, usually mice are immunized Anti-B Blood Grouping Serum
for the production of monoclonal antibodies. After suitable
immune response, mouse spleen cells containing antibody Anti-B Blood Grouping Serum is a sterile, liquid or dried
secreting lymphocytes are fused to neoplastic plasma cells of preparation containing the particular blood group antibodies
infinite reproductive capacity from a mouse (that is myeloma derived from high-titered blood plasma or serum of human
cells). The resulting hybridomas are screened for antibody subjects, with or without stimulation by the injection of Blood
with the required specificity and affinity. The antibody Group Specific Substance B (or AB). It contains a suitable
secreting clones may then be propagated in tissue culture or antimicrobial preservative.
by inoculation into mice in which case the antibodies are
collected as ascites. The clonal line produces a single antibody, Production
there is no need for absorption or to remove heterospecific
antibodies. All antibody molecules produced by a clone of The monoclonal antibody technique devised by Kohler and
hybridoma cells are identical in terms of antibody structure Milsten has proved useful in producing high titre and specific
and antigen specificity. Once one antibody secreting clone of antibodies. Laboratory animals, usually mice are immunized
cells has been established, antibody with same specificity for the production of monoclonal antibodies. After suitable
and reaction characteristics will be available indefinitely. immune response, mouse spleen cells containing antibody
secreting lymphocytes are fused to neoplastic plasma cells of
Identification infinite reproductive capacity from a mouse (that is myeloma
cells). The resulting hybridomas are screened for antibody
It agglutinates human red cells containing A-antigens, are with the required specificity and affinity. The antibody
blood groups A and AB (including subgroups A1, A2, A1B, secreting clones may then be propagated in tissue culture or
and A2B but not necessarily weaker subgroups). by inoculation into mice in which case the antibodies are
collected as ascites. The clonal line produces a single antibody,
Tests there is no need for absorption or to remove heterospecific
antibodies. All antibody molecules produced by a clone of
It meets the requirements to the test for potency, in parallel hybridoma cells are identical in terms of antibody structure
with, and not less than equivalent to, the Reference Blood and antigen specificity. Once one antibody secreting clone of
Grouping Serum Anti-A, in agglutinating red blood cells from cells has been established, antibody with same specificity
Group A1 and Group A2B donors. It complies with the tests and reaction characteristics will be available indefinitely.
for specificity with Group A1, A2B, B, and O cells and confirms
the absence of contaminating antibodies reactive with Mg, Identification
Wra antigens as well as other antigens having an incidence of
1 per cent or greater in the general population (see under It agglutinates human red cells containing B-antigens, i.e.,
Blood Grouping Serums Anti-D, Anti-C, Anti-E, Anti-c, blood groups B and AB (including subgroups A1B and A2B).
rAnti-e). It complies with the tests for avidity with Group A1
and A2B cells. All fresh or frozen red blood cell suspensions Tests
used for these tests are prepared under specified conditions
and meet specified criteria. Anti-A Blood Grouping Serum may It complies with the test for potency, in parallel with, and not
be artificially coloured blue. less than equivalent to, the Reference Blood Grouping Serum
Anti-B, in agglutinating red blood cells from Group B donors.
Expiration date. The expiration date for liquid serum is not It complies with the tests for specificity with Group A1, B, and
later than 1 year, and for dried serum not later than 5 years O cells and confirms the absence of contaminating antibodies
after date of issue from manufacturer’s cold storage (5o, 1 reactive with Mg, Wra antigens as well as other antigens having
year; or 00, 2 years), provided that the expiration date for dried an incidence of 1.0 per cent or greater in the general population
serum is not later than 1 year after constitution. (see under Blood Grouping Serums Anti-D, Anti-C, Anti-E,

1443
ANTI-D (Rho) IMMUNOGLOBULIN IP 2007

Anti-c, Anti-e). It complies with the test for avidity with Group Immunisation
B cells. All fresh or frozen red blood cell suspensions used for
Immunisation of the plasma donor is carried out under proper
these tests are prepared under specified conditions and meet
specified criteria. Anti-B Blood Grouping Serum may be medical supervision. Recommendations concerning donor
immunisation, including testing of erythrocyte donors, have
artificially coloured yellow.
been formulated by the World Health Organisation
Expiration date. The expiration date for liquid serum is not (Requirements for the collection, processing and quality
later than 1 year and for dried serum not later than 5 years after control of blood, blood components and plasma derivatives,
WHO Technical Report Series, No. 840, 1994 or subsequent
date of issue from manufacturer’s cold storage (5°, 1 year; or
revision).
0°, 2 years), provided that the expiration date for dried serum
is not later than 1 year after constitution. Pooled plasma
Storage. Store at a temperature between 2° and 8°. To limit the potential B19 virus burden in plasma pools used
for the manufacture of anti-D immunoglobulin, the plasma pool
Labelling. Label states that the source material was not is tested for B19 virus using validated nucleic acid
reactive for hepatitis B surface antigen, but that no known amplification techniques (2.8.1).
test method offers assurance that products derived from human B19 virus DNA. Maximum 104 IU per ml.
blood will not transmit hepatitis in case of polyclonal. Label
also states that it is for in vitro diagnostic use. A positive control with 104 IU of B19 virus DNA per ml and, to
test for inhibitors, an internal control prepared by addition of
NOTE—The labelling is in black lettering imprinted on paper a suitable marker to a sample of the plasma pool are included
that is white or is coloured completely or in part to match in the test. The test is invalid if the positive control is non-
the specified yellow color standard. reactive or if the result obtained with the internal control
indicates the presence of inhibitors.
B19 virus DNA for NAT testing reference preparation is
Anti-D (Rho) Immunoglobulin suitable for use as a positive control.
If Human Normal Immunoglobulin is added to the preparation,
Human Anti-D Immunoglobulin
the plasma pool from which it is derived complies with the
Human anti-D immunoglobulin is a liquid or freeze-dried above requirement for B19 virus DNA.
preparation containing immunoglobulins, mainly
immunoglobulin G. The preparation is intended for Tests
intramuscular administration. It contains specific antibodies
Potency. Determine the assay of human anti-D
against erythrocyte D-antigen and may also contain small
immunoglobulin by Method A (2.8.2). The estimated potency
quantities of other blood-group antibodies. Human normal
is not less than 90.0 per cent of the stated potency. The
immunoglobulin may be added.
confidence limits (P = 0.95) are not less than 80.0 per cent and
It complies with the monograph on Human Normal not more than 120.0 per cent of the estimated potency.
Immunoglobulin, except for the minimum number of donors
Method B or C (2.8.3) may be used for potency determination
and the minimum total protein content. For products prepared
if a satisfactory correlation with the results obtained by
by a method that eliminates immunoglobulins with specificities
Method A has been established for the particular product.
other than anti-D, where authorised, the test for antibodies to
hepatitis B surface antigen is not required. Storage. For the liquid preparation, store protected from light,
in a plastic container. For the freeze-dried preparation, store
Production protected from light, in a plastic container.
Human anti-D immunoglobulin is preferably obtained from Labelling. The label states (1) for liquid preparations, the
the plasma of donors with a sufficient titre of previously volume of the preparation in the container and the protein
acquired anti-D antibodies. Where necessary, in order to content expressed in grams per litre; (2) for freeze-dried
ensure an adequate supply of human anti-D immunoglobulin, preparations, the quantity of protein in the container; (3) the
it is obtained from plasma derived from donors immunised route of administration; (4) for freeze-dried preparations, the
with D-positive erythrocytes that are compatible in relevant name or composition and the volume of the reconstituting
blood group systems in order to avoid formation of undesirable liquid to be added; (5) where applicable, that the preparation
antibodies. is suitable for use in the prophylaxis of hepatitis A infection;
(6) where applicable, the anti-hepatitis A virus activity in
Erythrocyte donors International Units per ml; (7) where applicable, the name and
Erythrocyte donors comply with the requirements for donors amount of antimicrobial preservative in the preparation.
prescribed in the monograph on Human Plasma for The label also states the number of International Units per
Fractionation. container.

1444
IP 2007 ANTI-D IMMUNOGLOBULIN FOR INTRAVENOUS USE

Anti-D Immunoglobulin for immunisation, including testing of erythrocyte donors, have


been formulated by the World Health Organisation
Intravenous Use (Requirements for the collection, processing and quality
control of blood, blood components and plasma derivatives,
Anti-D Immunoglobulin Human for Intravenous Use WHO Technical Report Series, No. 840, 1994 or subsequent
Anti-D Immunoglobulin for intravenous administration is a revision).
liquid or freeze-dried preparation containing immunoglobulins,
mainly immunoglobulin G. It contains specific antibodies Pooled plasma
against erythrocyte D-antigen and may also contain small To limit the potential B19 virus burden in plasma pools used
quantities of other blood-group antibodies. Human normal for the manufacture of anti-D immunoglobulin, the plasma
immunoglobulin for intravenous use may be added. pool is tested for B19 virus using validated nucleic acid
It complies with the monograph on Human Normal amplification techniques (2.8.1).
Immunoglobulin for Intravenous Administration, except for
the minimum number of donors, the minimum total protein Identification
content, the limit for osmolality and the limit for prekallikrein
activator. For products prepared by a method that eliminates Examine by a suitable immunoelectrophoresis technique. Using
immunoglobulins with specificities other than anti-D: where antiserum to normal human serum, compare normal human
authorised, the test for antibodies to hepatitis B surface antigen serum and sample under examination in a dilution of 10 g per
is not required; a suitable test for Fc function is carried out. litre of protein. The main component of the sample corresponds
to the IgG component of normal human serum.
Production
Tests
Polyclonal (human) anti Rh (D) serum
B19 virus DNA . Maximum 104 IU per ml.
Human anti-D immunoglobulin is preferably obtained from
the plasma of donors with a sufficient titre of previously A positive control with 104 IU of B19 virus DNA per ml and, to
acquired anti-D antibodies. Where necessary, in order to test for inhibitors, an internal control prepared by addition of
ensure an adequate supply of human anti-D immunoglobulin, a suitable marker to a sample of the plasma pool are included
it is obtained from plasma derived from donors immunised in the test. The test is invalid if the positive control is non-
with D-positive erythrocytes that are compatible in relevant reactive or if the result obtained with the internal control
blood group systems in order to avoid formation of undesirable indicates the presence of inhibitors.
antibodies. B19 virus DNA for NAT testing RP is suitable for use as a
positive control.
Erythrocyte donors
If Human normal immunoglobulin for intravenous
Laboratory tests are carried out for each donation to detect administration is added to the preparation, the plasma pool
the following viral markers: from which it is derived complies with the above test for B19
virus DNA.
1. Antibodies against human immunodeficiency virus
1 (anti-HIV-1), Assay. Determine by method A of human anti-D
2. Antibodies against human immunodeficiency virus immunoglobulin (2.8.2). The estimated potency is not less
2 (anti-HIV-2), than 90.0 per cent of the stated potency. The confidence limits
3. Antibodies against hepatitis C virus (anti-HCV). (P = 0.95) are not less than 80.0 per cent and not more than
120.0 per cent of the estimated potency.
4. Hepatitis B surface antigen (HBsAg),
Method B or C (2.8.3) may be used for potency determination
if a satisfactory correlation with the results obtained by Method
Pending complete harmonisation of the laboratory tests to be A has been established for the particular product.
carried out, the competent authority may require that a test for
alanine aminotransferase (ALT) also be carried out. Storage. For the liquid preparation, store protected from light,
in a colourless glass container, at the temperature stated on
The test methods used are of suitable sensitivity and the label. For the freeze-dried preparation, store protected from
specificity and comply with the regulations in force. If a repeat- light, in colourless glass container, at a temperature not
reactive result is found in any of these tests, the donation is exceeding 25°.
not accepted.
Labelling. The label states (1) for liquid preparations, the
Immunisation volume of the preparation in the container and the protein
content expressed in gram per litre; (2) for freeze-dried
Immunisation of the plasma donor is carried out under proper preparations, the quantity of protein in the container; (3) the
medical supervision. Recommendations concerning donor amount of immunoglobulin in the container; (4) the route of

1445
ANTI- HUMAN GLOBULIN SERUM IP 2007

administration, (5) for freeze-dried preparations, the name or also states the specific antibody activities present; the
composition and the volume of the reconstituting liquid to be application for which the reagent is intended; a cautionary
added; (6) the distribution of subclasses of immunoglobulin statement that it does not contain antibodies to
G present in the preparation; (7) where applicable, the amount immunoglobulins or that it does not contain antibodies to
of albumin added as a stabilizer; (8) the maximum content of complement components, wherever and whichever is
immunoglobulin A.
applicable; and states that it is for in vitro diagnostic use.
The label states the number of International Units per container.
NOTE—The lettering on the label of the general-purpose
polyspecific reagent is black on a white background. The
label of all other Anti-Human Globulin Serum containers is
Anti-Human Globulin Serum in white lettering on a black background.

Anti-Human Globulin Serum is a sterile, liquid preparation of


serum produced by immunizing lower animals such as rabbits
or goats with human serum or plasma, or with selected human Blood Grouping Serums Anti-D, Anti-
plasma proteins. It is free from agglutinins and from hemolysins
to nonsensitized human red cells of all blood groups. It C, Anti-E, Anti-c, Anti-e
contains a suitable antimicrobial preservative. Anti-Rh Blood Grouping Serums
Three varieties of Anti-Human Globulin Serum are recognized: Blood Grouping Serums Anti-D, Anti-C, Anti-E, Anti-c, Anti-e
(1) a general-purpose polyspecific reagent which, as a
(Anti-Rh Group) are sterile, liquid or dried preparations derived
minimum, contains antibodies specific for immunoglobulin IgG,
from the blood plasma or serum of human subjects who have
and at least the C3d component of human complement (for developed specific Rh antibodies. They are free from
use in the direct antiglobulin test, it contains this Anti-C3d
agglutinins for the A or B antigens and from alloantibodies
and Anti-IgG activity) and which may be artificially coloured
other than those for which claims are made in the labelling.
green; (2) a reagent containing antibodies only against They contain a suitable antimicrobial preservative. Liquid
immunoglobulin IgG (not heavy chain specific) intended for
serums are not artificially coloured. Two varieties of Anti-Rh
use in the indirect antiglobulin test, and which may be
Blood Grouping Serums are recognized, i.e., (1) saline
artificially coloured green; and (3) reagents containing agglutinating “complete” antiserums, which specifically
antibodies specific for individual or selected components of
agglutinate human red blood cells suspended in saline and
human complement, such as Anti-C3, and Anti-C3b-C3d-C4,
(2) “blocking or incomplete” antiserums, which contain protein
or a single class of immunoglobulins, such as Anti-IgG (heavy or other macromolecular substances, usually require the cells
chain specific), used only to identify plasma components
to be suspended in serum or plasma, and generally are for
coated on the surface of red blood cells. Anti-Human Globulin
slide or rapid tube tests. The most commonly used of these
Serum containing Anti-IgG complies with the test for potency, blood grouping serums are listed in Table 1 each reacting with
in parallel with the Reference Anti-Human Globulin (Anti-
the antigen(s) designated by the corresponding letter(s) with
IgG) Serum (at a 1:4 dilution) when tested with red cells
the alternative nomenclature indicated parenthetically.
suspended in isotonic saline sensitized with decreasing
amounts of nonagglutinating Anti-D (Anti-Rho) serum, and Table - 1
with cells sensitized in the same manner with an ______________________________________________
immunoglobulin IgG Anti-Fya serum of similar potency. Anti- Serum Antigen(s) Reacting
Human Globulin Serum containing one or more Anti- ______________________________________________
complement components complies with the tests for potency
in giving a 2+ agglutination reaction (i.e., agglutinated cells Anti-D (Anti-Rho) D (Rho)
dislodged into many small clumps of equal size) by the low- Anti-C (Anti-rh′) C (rh′)
ionic sucrose or sucrose–trypsin procedures when tested as
recommended in the labelling. Anti-Human Globulin Serum Anti-E (Anti-rh′′) E (rh′′)
containing Anti-3Cd activity meets the requirements for Anti-CD (Anti-Rho′) D (Rho) and C (rh′)
stability, by potency testing of representative lots every 3
months during the dating period. Anti-DE (Anti-Rho′′) D (Rho) and E (rh′′)
Anti-CDE (Anti-Rho′′′) D (Rho), C (rh′), and E (rh′)
Expiration date. Its expiration date is not later than 1 year after Anti-c (Anti-hr′) c (hr′)
the date of issue from manufacturer’s cold storage (5°, 1 year;
Anti-e (Anti-hr′′) e (hr′′)
or 0°, 2 years).
______________________________________________
Storage. Store at a temperature between 2° and 8°. Each serum meets the requirements of the test for potency in
the case of serums for saline tube test in parallel with, and not
Labelling. Label states the animal source of the product. Label less than equivalent to, the Reference Blood Grouping Serum

1446
IP 2007 BLOOD GROUPING SERUM ANTI-D, ANTI-C, ANTI-E, ANTI-c, ANTI-e

for Anti-D, Anti-C, or Anti-E, whichever is applicable, or, in Anti-DE cDe, Ccde, cdEe, A1 cde, B cde, O cde
the case of Anti-c and Anti-e for saline tube test which have
Anti-CDE cDe, Ccde, cdEe, A1 cde, B cde, O cde, and
no reference preparations, the test for minimum agglutination
reactivity at a specified dilution; and in the case of serums for where recommended for detection of the G
antigen, rGr
slide or rapid tube test in parallel with, and not less than
equivalent to, the Reference Blood Grouping Serum for Anti- Anti-c Ccde, A1 CDe, B CDe, O CDe, and CDEe or
D, Anti-C, Anti-E, Anti-c, or Anti-e, whichever is applicable, in CDE or CdE
agglutinating as a minimum red blood cells from the donors
indicated in Table 2 (which may be from Group A, B, AB, or O). Anti-e cdEe, A1 cDE, B cDE, O cDE, and CcDE or
CDE or CdE
Each serum for slide or rapid tube test complies with the tests
for avidity with the cells as indicated under tests for potency ______________________________________________
above. All fresh or frozen red blood cell suspensions used for these
Table - 2 tests are prepared under specified conditions and meet
______________________________________________ specified criteria.
Serum Phenotype of Cells Monoclonal Rh (D) antibodies. Monoclonal antibodies are
______________________________________________ derived from hybridoma cell lines produced by fusing mouse
antibody produced by B lymphocyte with mouse myeloma
Anti-D cDe cells or are derived from a human B cell line through Epstein-
Anti-C Ccde Barr Virus (EBV) transformation. Each hybridoma cell line
produces homogenous antibodies of only one immunoglobulin
Anti-E cdEe class, which are identical in their chemical structure and
Anti-CD cDe, Ccde immunological activity.
Anti-DE cDe, cdEe The type of monoclonal anti-D reagents are:
Anti-CDE cdEe, cDe, Ccde 1. IgM anti-D monoclonal reagent,
Anti-c CcDEe 2. Blend of IgM and IgG monoclonal antibodies
reagent,
Anti-e cdEe
3. Blend of monoclonal IgM and polyclonal (human)
IgG anti-D.
Each serum complies with the tests for specificity by the most IgM anti-D monoclonal antibodies are highly specific and
sensitive method recommended by the manufacturer, in which saline reacting equally well at room temperature and at 37°.
not less than 4 positive and 4 negative phenotypes are included They are good for slide test or immediate spin tube tests as
(see Table 3), and confirms the absence of contaminating well as routine Rh(D) typing in tube. IgM anti D are unreliable
antibodies reactive with Mg, Wra antigens as well as other for detection of weak D (Du) by AHG test. Blend of IgM and
antigens having an incidence of 1.0 per cent or greater in the IgG (monoclonal) anti-D or blend of IgM (monoclonal) and
general population, except where some of these confirmatory polyclonal (human) IgG anti-D can be used for testing weak D
tests can not be done by the manufacturer, in which event (Du) antigen by AHG test. Mostly blended IgM and IgG
such omissions are noted. (monoclonal) anti- Rh(D) or blended monoclonal IgM and
polyclonal (human) IgG anti Rh (D) antibodies are used now
Table 3 in routine.
______________________________________________
Expiration date. The expiration date for liquid serums is not
Serum Cells later than 1 year and for dried serums not later than 5 years
______________________________________________ after date of issue from manufacturer’s cold storage (5°, 1
Anti-D CcDe, cDe, Ccde, cdEe, A1 cde, B cde, O cde, year; or 0°, 2 years), provided that the expiration date for dried
and where recommended for use by indirect serums is not later than 1 year after constitution.
antiglobulin technique, cde Bg(a+) cells from 3 Storage. Store at a temperature between 2° and 8°.
different donors
Labelling. Label each to state that the source material was not
Anti-C cDe, Ccde, cdEe, C + rhi neg. cells, A1 cde, B reactive for hepatitis B surface antigen, but that no known
cde, O cde test method offers assurance that products derived from human
Anti-E cDe, Ccde, cdEe, A1 cde, B cde, O cde blood will not transmit hepatitis. Label each to state that it is
for in vitro diagnostic use.
Anti-CD cDe, Ccde, cdEe, A1 cde, B cde, O cde, and
where recommended for detection of the G
antigen, rGr

1447
CORPUSCLES IP 2007

Concentrated Human Red Blood Description. A dark red fluid when prepared; after standing,
the red corpuscles may form a sediment, leaving a small
Corpuscles supernatant layer of yellowish plasma.
Concentrate of Human Red Blood Cells; Packed Red Cells Tests
Concentrated Red Blood Cells (RBCs) are units of whole blood Sterility (2.2.11). Complies with the tests for sterility,
with most of the plasma removed. Additive red cell preservative determined by Method B.
systems consist of a primary collection bag containing an
anticoagulant preservative with at least two satellite bags Assay. Determine the haemoglobin content by photometric
integrally attached, one is empty and one contains an additive haemoglobinometry (2.8.12).
solution (AS). Storage. Store in containers which are made of colourless and
Each 100 ml citrate phosphate dextrose (CPD) contains transparent glass, or of a suitable plastic material, are sterile
and sealed so as to exclude micro-organisms. Store at a
Citric acid (monohydrate) 0.327 g temperature between 2o and 8o.
Sodium citrate (dihydrate) 2.63 g Labelling. The label states (1) the reference number of the
Sodium acid phosphate (dihydrate) 0.251 g Whole Human Blood from which the preparation was made;
(2) the ABO and Rh groups of the Whole Human Blood; (3)
Dextrose (monohydate) 2.55 g the date of collection of the Whole Human Blood from which
Water for injection 100 ml the preparation was made; (4) the storage conditions; (5) the
date after which the preparation is not suitable for transfusion;
AS contains sodium chloride, dextrose, adenine and other
(6) that the preparation should not be used if there is any
substances that support red cell survival and function up to
visible evidence of haemolysis or other deterioration.
42 days. The volume of AS in 350 ml is 49 ml and in 450 ml is
63 ml. AS is added to the red cells remaining in the primary bag
after most of the plasma has been removed. This allows blood
centres to use or recover a maximum amount of plasma, yet
still prepare a red cell component with a final haematocrit
Cryoprecipitated Antihaemophilic
between 55.0 per cent and 65.0 per cent, a level that facilitates Factor
excellent flow rates and allows easy administration.
Cryoprecipitated Antihaemophilic Factor is a sterile, frozen
Production concentrate of human antihaemophilic factor prepared from
the Factor VIII-rich cryoprotein fraction of human venous
It may be prepared by centrifugation or undisturbed
plasma obtained from suitable whole-blood donors from a
sedimentation for the separation of plasma and anticoagulant
single unit of plasma derived from whole blood or by
solution equivalent to not less than 40.0 per cent of the total
plasmapheresis, collected and processed in a closed system.
volume of Whole Human Blood. A portion of the plasma,
It contains no preservative. It complies with the test for
sufficient to ensure optimal cell preservation, shall be left. All
potency by comparison with the Standard Antihaemophilic
surfaces that come in contact with the red cells and plasma
Factor (Factor VIII) or with a working reference that has been
shall be sterile and pyrogen-free and the entire processing of
calibrated with it, in having an average potency of not less
the blood shall be conducted in a sterile system or in a closed
than 80 Antihaemophilic Factor Units per container, made at
system by use of satellite bags. The final containers used for
intervals of not more than 1 month during the dating period.
the Concentrated Human Red Blood Corpuscles shall be the
original blood containers unless the method of processing Expiration date. The expiration date is not later than 1 year
requires a different container. Immediately after processing, from the date of collection of source material.
the containers are stored at a temperature between 2o and 8o o
Storage. Store in hermetic containers at a temperature of - 18
and not opened until immediately before transfusion. Human
Red Blood Corpuscles may be stored for a period not longer or lower.
than that for which the Whole Human Blood from which it is Labelling. Label it to indicate (1) the ABO blood group
prepared. However, if the hermetic seal is broken during designation and the identification number of the donor from
processing, the product must be used within 24 hours. From whom the source material was obtained; (2) with the type and
the tube of the blood bag sample is taken for compatibility result of a serologic test for syphilis, or to indicate that it was
and infections markers testing. non-reactive in such test; with the type and result of a test for
Concentrated Human Red Blood Corpuscles should be hepatitis B surface antigen, or to indicate that it was non-
administered only with suitable equipment meant for the reactive in such test; with a warning not to use it if there is
transfusion of blood and blood components. evidence of breakage or thawing; with instructions to thaw it
before use to a temperature between 20° and 37°, after which
Concentrated Human Red Blood Corpuscles contains not less it is to be stored at room temperature and used as soon as
than 15.5 per cent w/v of haemoglobin. possible but within 6 hours after thawing; (3) to state that it is

1448
IP 2007 DRIED HUMAN ANTIHAEMOPHILIC FRACTION

to be used within 4 hours after the container is entered; (4) to containers and dried from the frozen state. The air is removed
state that it is for intravenous administration; (5) that a filter or replaced by oxygen-free nitrogen and the containers are
is to be used in the administration equipment. sealed so as to exclude micro-organisms. No antimicrobial
preservative is added but an antiviral agent may be added
provided that it can be demonstrated to have no deleterious
effect on the final product in the amount present and to cause
Dried Human Antihaemophilic no adverse reaction in man. Heparin may be used.
Fraction For the following tests, where it is directed that a solution is to
be used, dissolve the contents of a sealed container in a volume
Dried Factor VIII Fraction; Freeze-dried Human Coagulation of the appropriate solvent equal to the volume of water for
Factor VIII injection stated on the label.
Dried Human Antihaemophilic Fraction is a preparation of Description. A white or pale yellow powder or friable solid.
antihaemophilic factor which is obtained from human plasma.
It is rich in clotting factor VIII. Identification
When the contents of a sealed container of Dried Human A. Precipitation tests with a suitable range of species specific
Antihaemophilic Fraction are dissolved in a volume of water antisera give positive results for the presence of plasma
equal to the volume of water for injection stated on the label, proteins of human origin and negative results with antisera
the resulting solution contains not less than 3.0 Units per ml, specific to plasma proteins of the other species.
not less than 0.1 Unit per mg of protein, not more than 80.0 per B. A freshly prepared solution in water causes a reduction in
cent of which is fibrinogen, and not more than 200 millimoles clotting time when treated as directed under Assay.
of sodium ions per litre.
Tests
Production
pH (2.4.24). 6.8 to 7.4, determined on the reconstituted solution.
The plasma to be used for preparing Dried Human
Antihaemophilic Fraction is obtained from blood of healthy Loss on drying (2.4.19). Not more than 2.0 per cent, determined
human donors who are, as far as can be ascertained after by drying 0.5 g over phosphorus pentoxide at a pressure not
clinical examination, laboratory tests on their blood and exceeding 3 kPa for 24 hours.
consideration of their medical history, free from detectable Haemagglutinins, anti-A and anti-B. Dissolve in water. Dilute
agents of infection transmissible by blood transfusion. The the solution with saline solution to produce a solution
examinations and tests to be carried out are decided by the containing 3 Units per ml. Carry out the test for
National Regulatory Authority. In particular, the blood must haemagglutinins anti-A and anti-B using a suitable indirect
be tested with negative results for (a) evidence of syphilitic method such as that described below.
infection; (b) hepatitis B surface antigen and (c) HIV antibodies
by suitably sensitive methods. The haemoglobin value of the Prepare in duplicate serial dilutions of the preparation under
donor’s blood is not less than 12.5 per cent w/v. examination in saline solution. To each dilution of one series
add an equal volume of a 5.0 per cent v/v suspension of group
The blood is withdrawn aseptically through a closed system A1 red blood cells previously washed three times with saline
of sterile tubing into a sterile container in which a suitable solution. To each dilution of the other series add an equal
anticoagulant solution has been placed before sterilisation. volume of a 5.0 per cent v/v suspension of group B red blood
During the withdrawal there is no interruption in the flow from cells previously washed three times with saline solution.
the donor, and the container is gently agitated. Immediately Incubate the suspensions at 37o for 30 minutes and then wash
after the withdrawal is completed, the blood is cooled to 4o; if the cells three times with saline solution. Leave the cells in
the plasma is to be stored frozen it is separated from the cellular contact with a polyvalent anti-human globulin reagent for 30
components by centrifugation and frozen to –30o or below, minutes. Without centrifuging, examine each suspension for
preferably within 12 hours of collection; if the plasma is not to agglutination under a microscope. The 1 in 64 dilutions do not
be frozen it is separated from the cellular components by show agglutination.
centrifugation as soon as possible and not later than 18 hours
after collection, and fractionation begun without delay. Hepatitis B surface antigen. Dissolve in water. Examine the
solution by a suitably sensitive method such as
Dried Human Antihaemophilic Fraction may be prepared from radioimmunoassay. Hepatitis B surface antigen is not detected.
human plasma so obtained by precipitation under controlled
conditions of pH, ionic strength and temperature with organic Abnormal toxicity (2.2.1). When dissolved in water for
solvents, or by freezing and thawing. The precipitate may be injection, complies with the test for abnormal toxicity, Method
washed by extraction with suitable solvents, dissolved in a B, injecting into each mouse a volume containing 1.5 Units
solution of sodium citrate adjusted to a pH of 6.8 to 7.2, which and into each guinea-pig a volume containing 15 Units.
may also contain sodium chloride. The solution is sterilised Pyrogens (2.2.8). When dissolved in water for injection,
by filtration through a membrane filter, distributed in sterile complies with the test for pyrogens, using a volume containing

1449
DRIED HUMAN ANTIHAEMOPHILIC FRACTION IP 2007

10 Units per kg of the rabbit’s weight in rabbits that have not petroleum mixture. Combine the extracts and evaporate to
previously received blood products. dryness at 45o at a pressure not exceeding 0.7 kPa. Dissolve
Sterility (2.2.11). Complies with the tests for sterility. the residue in 0.2V ml of ether and allow to stand at 4o until a
deposit forms. Centrifuge and evaporate the clear supernatant
Assay liquid under reduced pressure until the volume is about 100 ml
per kg of the original macerate. Allow to stand at 4o until a
For potency — Carry out the biological assay of human precipitate forms (12 to 24 hours) and centrifuge. To the clear
antihaemophilic fraction described below. The estimated supernatant liquid add 5 times its volume of acetone, centrifuge,
potency is not less than 80.0 per cent and not more than 125.0 discard the supernatant liquid, dry the precipitate and store
per cent of the stated potency. The fiducial limits of error are protected from light in a vacuum desiccator.
not less than 64.0 per cent and not more than 156.0 per cent of
Phospholipid reagent. Suspend 0.125 g of phospholipid in 5
the stated potency. ml of water, shake and stir until a uniform suspension is
Biological assay obtained. Prepare a dilution with saline solution that will give
minimum clotting times consistent with the largest clotting
The potency of human antihaemophilic fraction is determined time differences between consecutive dilutions of the Standard
by comparing the amount necessary to reduce the clotting preparation and the preparation under examination. The
time of a test mixture containing substances that cause clotting concentration usually lies between 50 and 250 µg per ml. The
of blood with the amount of the Standard Preparation necessary diluted suspension may be kept at –20º for 6 weeks.
to produce the same effect under the conditions of the Clotting factor V solution. Prepare from fresh oxalated bovine
following method of assay. plasma by fractionation at 4o with a saturated solution of
ammonium sulphate prepared at 4 o . Use the fraction
Standard preparation
precipitating between 38 per cent and 50 per cent saturation
The Standard preparation is the 4th International Standard for (which contains clotting factor V not significantly contaminated
Blood coagulation factor VIII:C, concentrate, human, with clotting factor VIII), dialysed to remove ammonium
established in 1989, consisting of an intermediate purity sulphate and diluted with saline solution to give a solution
concentrate of human blood clotting factor VIII (supplied in containing between 10 per cent and 20 per cent of the amount
ampoules containing 6.3 Units of clotting factor VIII), or of clotting factor V present in fresh normal human plasma.
another suitable preparation the potency of which has been Determine the clotting factor V content of the solution as
determined in relation to the International Standard follows. Prepare two dilutions in imidazole buffer pH 7.4 to
The Unit is the specific antihaemophilic factor contained in contain 1 volume of the solution under examination in 10
such an amount of the Standard Preparation as the Ministry volumes and 20 volumes respectively. Test each dilution as
of Health & Family Welfare, Govt. of India may from time to follows. Mix 0.1 ml each of substrate plasma deficient in
time indicate as the quantity exactly equivalent to the Unit clotting factor V, the dilution under test, thrombokinase
accepted for international use. extract and 0.025M calcium chloride. Record as the clotting
time the interval between the addition of the calcium chloride
Special reagents solution and the first indication of fibrin formation, which
may be observed visually or by mechanical means.
Normal serum reagent. Collect normal human blood in a dry,
sterile, glass bottle, shake continuously until coagulation is Similarly determine the clotting times, in duplicate, for four
complete, incubate at 37o for 3 hours, maintain at 4o overnight, dilutions of pooled normal human plasma in imidazole buffer
remove the serum, store at –20o, dry from the frozen state and pH 7.4 containing 1 volume in 10 volumes (equivalent to 100
keep in a vacuum desiccator over phosphorus pentoxide. per cent of clotting factor V), in 50 volumes (20 per cent), in
Dissolve a quantity of the dried serum calculated to have 100 volumes (10 per cent), and in 1,000 volumes (1 per cent),
been obtained from 1 ml of the serum in sufficient imidazole respectively.
buffer pH 7.4 to produce 10 ml and allow to stand at 4o for 16 To calculate the result, plot the mean of the clotting times for
to 24 hours. each dilution of human plasma on double cycle log/log paper
Phospholipid. Wash a quantity of normal human or bovine against the equivalent percentage of clotting factor V and
brain freed from meninges and blood vessels and macerate in read the percentage of clotting factor V for the two dilutions
a suitable blender. Weigh 1,000 to 1,300 g of the macerate and of clotting factor V solution by interpolation from the curve.
measure its volume (V). Extract with three quantities, each of The mean of the two results is taken as the percentage of
4V ml, of acetone, filter by suction and dry the precipitate at clotting factor V in the solution.
37o for 18 hours. Extract the dried precipitate with two Substrate plasma. Separate the plasma from 9 volumes of
quantities, each of 2V ml, of a mixture of two volumes of light human or bovine blood collected in 1 volume of a 3.8 per cent
petroleum (boiling range 30o to 40o) and 3 volumes of light w/v solution of sodium citrate or from 3.5 volumes of human
petroleum (boiling range 40o to 60o), filtering each extract or bovine blood collected in 1 volume of a solution containing
through a filter paper previously washed with the light 2.0 per cent w/v of sodium acid citrate and 2.5 per cent w/v of

1450
IP 2007 DRIED HUMAN ANTIHAEMOPHILIC FRACTION

dextrose. In the former case, prepare the substrate plasma on incubation tubes at 1-minute intervals and carry out a second
the day of collection of the blood. In the latter case, the series of determinations at 21 to 26 minutes. The period of
substrate plasma may be prepared up to 2 days after collection incubation should, if necessary, be adjusted so that the clotting
of the blood. Store at –20o. times recorded in the corresponding tests in the two series of
determinations do not differ by more than 5.0 per cent, showing
Substrate plasma deficient in clotting factor V. Preferably
use congenitally deficient plasma or, alternatively, prepare as that a stable plateau of prothrombin activator formation has
been reached.
follows. Separate the plasma from human blood collected in
one-tenth its volume of a 1.34 per cent w/v solution of sodium Carry out a blank determination using in place of the
oxalate and incubate at 37o for 24 to 36 hours. This plasma preparation under examination, an equal quantity of a mixture
should have a clotting time, when tested by the assay method of 1 volume of a 3.8 per cent w/v solution of sodium citrate
given under clotting factor V solution, of 70 to 100 seconds; and 5 volumes of saline solution. The result of the assay is
if the clotting time is less than 70 seconds, incubate the plasma not valid unless the clotting time in the blank determination is
for a further 12 to 24 hours. more than 40 seconds. Calculate the result of the assay by
Storage. Store in small amounts, at -20o or below. standard statistical methods.
For total protein. Dilute 1.0 ml to 10.0 ml with saline solution.
Suggested method Determine the assay described under Human Plasma using
5.0 ml of the dilution and beginning at the words “add 0.2 ml of
Dissolve the contents of the sealed container of the substance
a 7.5 per cent w/v solution....”.
under examination in the volume of the liquid stated on the
label and use immediately. Reconstitute the entire contents of For fibrinogen. Dilute 1.0 ml of the solution to 10.0 ml with a
one ampoule of the Standard preparation as stated on the phosphate-saline buffer pH 6.5 and ionic strength 0.15. Clot
label and use immediately. 5.0 ml of the dilution with the minimum amount of thrombin,
collect the clot and add three drops of a 30 per cent w/v solution
To the reconstituted Standard preparation and the preparation
of copper sulphate and 1 ml of nitrogen-free sulphuric acid
under examination, add sufficient imidazole buffer pH 7.4 to
and boil gently for 10 minutes; cool, add 1 g of anhydrous
produce solutions containing between 0.5 and 2 Units per ml; sodium sulphate and 10 mg of selenium, boil gently for 1 hour
these solutions are stable for 15 minutes at 20o. Using a mixture
and cool. Transfer to an ammonia distillation apparatus, add 6
of 1 volume of a 3.8 per cent w/v solution of sodium citrate
ml of a saturated solution of sodium hydroxide and pass steam
and 5 volumes of saline solution as the diluent, make from the through the flask; distil for seven minutes, collecting the
solutions three successive 2-fold dilutions in the range 1 in 16
distillate in a mixture of five ml of a saturated solution of boric
to 1 in 256 so that all the clotting times are between 17 and 35
acid, 5 ml of water, and 1 drop of a saturated solution of
seconds; the dilutions must be accurately made and used methyl red in alcohol containing 0.1 per cent of methylene
immediately.
blue, and titrate with 0.02 M hydrochloric acid.
Introduce into each of six glass incubation tubes (75 mm 100 1 ml of 0.02M hydrochloric acid is equivalent to 0.00175 g of
mm) 0.1 ml each of clotting factor V solution, phospholipid
fibrinogen.
reagent and normal serum reagent. To the first tube add 0.1
ml of the highest dilution of the Standard Preparation, place For sodium ions. To 10.0 ml of the solution add sufficient
the tube in a water-bath at 37o, add 0.1 ml of 0.05M calcium water to produce 100 ml, dilute 10.0 ml to 500 ml with water
chloride and start a stop-watch. During the next minute add and determine the content of sodium ions by Method B for
0.1 ml of the second highest dilution of the standard to a flame photometry (2.4.4), measuring at about 589 nm and using
second tube, place it in the water-bath, and add 0.1 ml of sodium solution FP suitably diluted with water as the standard
0.05M calcium chloride at exactly 1 minute by the stop-watch. solution.
Repeat the procedure with the lowest dilution of the standard Storage. Store protected from light, in an atmosphere of
and the highest to lowest dilutions of the preparation under nitrogen at a temperature below 8o. The containers are sterile
examination so that the calcium chloride solution is added at and sealed so as to exclude micro-organisms.
2, 3, 4 and 5 minutes by the stop-watch, respectively.
Labelling. The label states (1) the ABO blood group
Place in a water-bath at 37o twelve glass tubes each containing designation of the source of blood; (2) the number of Units in
0.2 ml of 0.025M calcium chloride and a further tube the container; (3) that 1 Unit is approximately equivalent to
containing about 3 ml of substrate plasma. At 14 minutes, 40 the antihaemophilic activity of 1 ml of average normal plasma;
seconds by the stop-watch, transfer 0.1 ml of the mixture from (4) the concentration of protein in g per litre and of sodium
the first incubation tube to one of the tubes containing 0.2 ml ions in millimoles per litre of the solution constituted as
of 0.025M calcium chloride solution and mix. At 15 minutes directed; (5) the maximum fibrinogen content; (6) where
add 0.2 ml of the warmed substrate plasma and, using a second applicable, the number of Units of heparin in the container; (7)
stop-watch, record as the clotting time the interval between the name and amount of any other added substance contained
the addition of the substrate plasma and the first indication of in it; (8) the volume of Water for Injections necessary to
fibrin formation, which may be observed visually or by constitute the solution; (9) the instructions for constitution
mechanical means. Repeat the procedure with the other

1451
FIBRIN SEALANT KIT IP 2007

and that reconstitution may take upto 30 minutes; (10) that if and water.
the solution is not complete or if a gel forms on constitution,
the preparation should not be used; (11) that the solution Component 1 (Fibrinogen Concentrate)
should be used as soon as possible and in any case within 3
hours of constitution and any unused solution should be Identification
discarded; (12) that the solution should be administered only
with equipment that includes a filter; (13) the storage A. Complies with the limits of the assay of fibrinogen.
conditions. B. Complies with the limits of the assay of factor XIII
(where applicable).

Tests
Fibrin Sealant Kit
pH (2.4.24). 6.5 to 8.0.
Fibrin Sealant Kit is essentially composed of two components,
namely fibrinogen concentrate (component 1), a protein Stability of solution. No gel formation appears at room
fraction containing human fibrinogen and a preparation temperature during 120 min following thawing or
containing human thrombin (component 2). A fibrin clot is reconstitution.
rapidly formed when the two thawed or reconstituted Water. Determine by semi-microdetermination (2.3.43), loss
components are mixed. Other ingredients (for example, human on drying (2.4.19) or near infrared spectrophotometry (2.4.6),
coagulation factor XIII, a fibrinolysis inhibitor or calcium ions) the water content is within the limits approved by the
and stabilisers (for example, Human albumin solution may be competent authority.
added. No antimicrobial preservative is added.
Sterility (2.2.11). Complies with the test for sterility.
Human constituents are obtained from plasma that complies
with the requirements of the monograph on Human Plasma for Assay
Fractionation. No antibiotic is added to the plasma used.
Fibrinogen (Clottable Protein)
When thawed or reconstituted as stated on the label,
component 1 contains not less than 4.0 per cent of clottable The estimated content in mg of clottable protein is not less
protein; the thrombin activity of component 2 varies over a than 70.0 per cent and not more than 130.0 per cent of the
wide range (approximately 4-1000 IU per ml). content stated on the label.
Mix 0.2 ml of the reconstituted preparation with 2 ml of a
Production suitable buffer solution pH 6.6 to 7.4 containing sufficient
human thrombin (approximately 3 IU per ml) and calcium
The method of preparation includes a step or steps that have
(0.05mol per l). Maintain at 37° for 20 minutes, separate the
been shown to remove or to inactivate known agents of
precipitate by centrifugation (5000 g, 20 minutes), wash
infection; if substances are used for inactivation of viruses
thoroughly with a 0.9 per cent solution of sodium chloride
during production, the subsequent purification procedure
and determine the protein as nitrogen by sulphuric acid
must be validated to demonstrate that the concentration of
digestion (2.3.30). Calculate the protein content by multiplying
these substances is reduced to a suitable level and any
the result by 6.0. If for a particular preparation this method
residues are such as not to compromise the safety of the
cannot be applied, use another validated method for
preparation for patients.
determination of fibrinogen.
Constituents or mixtures of constituents are passed through
a bacteria-retentive filter and distributed aseptically into sterile Factor XIII
containers. Containers of freeze-dried constituents are closed
under vacuum or filled with oxygen-free nitrogen or other Where the label indicates that the human coagulation factor
suitable inert gas before being closed. In either case, they are XIII activity is greater than 10 Units per ml, the estimated
closed so as to exclude micro-organisms. activity is not less than 80.0 per cent and not more than 120.0
per cent of the activity stated on the label.
If the human coagulation factor XIII content in component 1
is greater than 10 Units per ml, the assay of coagulation factor Make at least 3 suitable dilutions of thawed or reconstituted
XIII is carried out. component 1 and of human normal plasma (reference
preparation) using as diluent coagulation factor XIII deficient
Description. Freeze-dried constituents are hygroscopic, white plasma or another suitable diluent. Add to each dilution
or pale yellow powders or friable solids. Frozen constituents suitable amounts of the following reagents:
are colourless or pale yellow, opaque solids. Liquid
constituents are colourless or pale yellow. a. activator reagent, containing bovine or human thrombin,
a suitable buffer, calcium chloride and a suitable inhibitor
For the freeze-dried or frozen constituents, reconstitute or such as Gly-Pro-Arg-Pro-Ala-NH2 which inhibits clotting of
thaw as stated on the label immediately before carrying out the sample but does not prevent coagulation factor XIII
the Identification and the Tests, except those for solubility

1452
IP 2007 HUMAN ALBUMIN

activation by thrombin, of clottable protein), thrombin (International Units) per


container, and coagulation factor XIII, if this is greater than 10
b. detection reagent, containing a suitable factor XIIIa-specific
peptide substrate, such as Leu-Gly-Pro-Gly-Glu-Ser-Lys-Val- Units per ml; (2) where applicable, the name and volume of
solvent to be used to reconstitute the components.
Ile-Gly-NH2 and glycine ethyl ester as 2nd substrate in a
suitable buffer solution,
c. NADH reagent, containing glutamate dehydrogenase, a-
ketoglutarate and NADH in a suitable buffer solution. Human Albumin
After mixing, the absorbance changes (ÄA per min) are Human Normal Albumin; Human Albumin Solution
measured at a wavelength of 340 nm, after the linear phase of Human Albumin is a sterile non-pyrogenic solution of the
the reaction is reached. albumin component obtained from pooled human blood or
1 Unit of factor XIII is equal to the activity of 1 ml of human from normal placentae frozen immediately after collection. It is
normal plasma. obtained by fractionating source material such as blood,
plasma, serum or placentae from healthy human donors and
Calculate the activity of the test preparation by the usual tested individually for the absence of hepatitis B surface
statistical methods. The confidence limits (P = 0.95) are not antigen, HCV antibodies and HIV antibodies and complies
less than 80.0 per cent and not more than 125.0 per cent of the with other tests and requirements prescribed by the appropriate
estimated activity. national control authority. Source material obtained from
donors who do not meet all the requirements stated may be
Component 2 (Thrombin Preparation)
used provided that it has been demonstrated to the national
control authority that the process of fractionation will remove
Identification any known agent capable of adversely affecting the health of
Complies with the limits of the assay of thrombin. subjects treated with the preparation. It may be prepared from
pooled source materials by precipitation with organic solvents
Tests under controlled conditions of pH, ionic strength and
temperature or by chromatography or by any other method
pH (2.4.24). 5.0 to 8.0. which does not affect the integrity of the product and has
been shown to yield consistently a product containing not
Water. Determine by semi-microdetermination of water
less than 95.0 per cent w/v of the total protein as albumin
(2.3.43), loss on drying (2.4.19) or near infrared
which is safe for intravenous injection. Residual organic
spectrophotometry (2.4.6), the water content is within the
solvent, if present, is removed by freeze-drying or other
limits approved by the competent authority.
suitable treatment. The product is dissolved in sufficient water
Sterility (2.2.11). Complies with the test for sterility. to obtain a suitable concentration, and a suitable stabilising
agent is added to stabilise it to heat. It is prepared as a solution
Assay containing 15.0 to 25.0 per cent w/v of total protein or as an
isotonic solution containing 4.0 to 5.0 per cent w/v of total
Thrombin
protein. No antimicrobial agent is added at any stage during
The estimated activity is not less than 80.0 per cent and not preparation and all processing steps are conducted in a manner
more than 125.0 per cent of the activity stated on the label. to minimise risk of contamination from either micro-organisms
If necessary, dilute the reconstituted preparation under or other deleterious matter. The solution is sterilised by
examination to approximately 2-20 IU of thrombin per ml using filtration and distributed aseptically into containers which are
as diluent a suitable buffer pH 7.3 to 7.5, such as imidazole then sealed so as to exclude micro-organisms. The solution is
buffer solution pH 7.3 containing 1.0 per cent of human then heated to and maintained at 60o ± 0.5o for 10 hours so as
albumin or bovine albumin. To a suitable volume of the to prevent the transmission of agents of infection transmissible
dilution, add a suitable volume of fibrinogen solution (0.1 per by transfusion of blood or blood derivatives. Finally, the
cent of clottable protein) warmed to 37° and start measurement containers are stored for not less than 14 days at 30o to 32o or
of the clotting time immediately. Repeat the procedure with for not less than 4 weeks at 20o and examined visually. Those
each of at least 3 dilutions, in the range stated above, of a showing abnormalities such as abnormal colour, turbidity,
reference preparation of thrombin, calibrated in International microbial contamination, or presence of atypical particles must
Units. Calculate the activity of the test preparation by the be discarded.
usual statistical methods. The confidence limits (P = 0.95) are Albumin Solution should be tested in accordance with the
not less than 80.0 per cent and not more than 125.0 per cent of requirements decided by the National Regulatory Authority;
the estimated activity. in particular, tests for the absence of hepatitis B surface antigen
Storage. Store protected from light. and HIV antibodies are carried out by suitably sensitive
methods.
Labelling. The label states (1) the amount of fibrinogen (mg

1453
HUMAN ALBUMIN IP 2007

Human Albumin contains not less than 95.0 per cent and not the volume, V, of the eluate from the entry of the sample into
more than 105.0 per cent of the stated amount of protein. It the column to the apex of the first peak.
contains not less than 95.0 per cent and not more than 105.0 Dilute the substance under examination with the saline-
per cent of the contents of Na and K stated on the label which
phosphate solution to contain about 5.0 per cent w/v of protein,
are, in any case, not more than 160 millimoles of Na per litre
apply 2.5 ml to the column and elute under the above
and 2 millimoles of K per litre. conditions, collecting the eluate in 5-ml portions. Three peaks
Description. A clear, slightly viscous liquid, ranging in colour may appear in similar positions to those in the chromatogram
from almost colourless to greenish-yellow or amber depending obtained from the normal human serum but the relative peak
on protein concentration and the method of fractionation used. heights may be different. To the fraction eluted between
volume 0.85 V and 1.15 V, add for each 10 ml, 0.4 ml of a 7.5 per
Identification cent w/v solution of sodium molybdate and 0.4 ml of a mixture
of 1 part of nitrogen-free sulphuric acid and 30 parts of water,
A. It contains plasma proteins of human origin only as shake, centrifuge for 5 minutes and complete the Assay
determined by precipitation tests with specific antisera. described under Human Plasma beginning at the words
B. Ditermine the cellulose acetate by electrophoresis (2.4.12), “decant the supernatant...”. The weight of protein in the
using barbitone buffer pH 8.6, ionic strength 0.1 and human fraction of the eluate is not more than 3.5 per cent of the
albumin for electrophoresis RS; 96.0 per cent of the protein weight of protein in the volume of the substance under
has the mobility of human albumin. examination applied to the column.
Heat the substance under examination for 50 hours at 56.5o to
Tests 57.5o and repeat the chromatographic separation and the
Acidity or alkalinity (2.4.24). Dilute with sufficient saline determination of the weight of protein in the fraction eluted
solution to produce a solution containing 1.0 per cent w/v of between 0.85 V and 1.15 V. When expressed as a percentage of
protein; pH of the resulting solution, 6.7 to 7.3. the weight of protein in the volume of the substance under
examination applied to the column, it exceeds the percentage
Alkaline phosphatase. Not more than 0.1 Unit per g of protein, obtained before heating by not more than 1.5 per cent w/v.
determined by the following method. Transfer a mixture of 0.5
ml of the substance under examination and 0.5 ml of Protein composition. Not less than 95.0 per cent w/v of the
diethanolamine buffer pH 10.0 to a spectrophotometer cell total protein as albumin, when determined by the following
maintained at a temperature of 37o ± 0.2o and add 0.1 ml of method. Carry out Method II for cellulose acetate
nitrophenyl phosphate solution. Using a continuously electrophoresis (2.4.12), using one strip of cellulose for each
recording spectrophotometer record the absorbance of the solution.
solution at about 405 nm (2.4.7), over a period of at least 30 Test solution. Dilute the substance under examination with
seconds from the time of addition of the nitrophenyl saline solution to contain 2.0 per cent w/v of total protein.
phosphate solution. Calculate the alkaline phosphatase
activity at 37o in Units per g of protein from the expression Reference solution. Dilute human albumin for electrophoresis
118.3x/P, where x is the rate of increase of absorbance per RS with saline solution to obtain a solution containing 2.0 per
minute and P is the content of total protein in g per litre, as cent w/v of total protein.
determined in the Assay. Not more than 5.0 per cent of total protein is contained in
Haem content. Dilute with sufficient saline solution to produce bands other than the principal band in the strip obtained with
a solution containing 1.0 per cent w/v of protein; absorbance test solution. The test is not valid if the proportion of the
of the resulting solution at about 403 nm, not more than 0.15 protein in the principal band is not within the limits stated in
(2.4.7). the leaflet supplied with human albumin for electrophoresis
RS.
Denatured protein. Equilibrate a column (60 to 75 cm x 2.5 to
3.0 cm) of a gel of a cross-linked dextran suitable for Stability. The contents of the final container remain unchanged,
fractionation of proteins in the range of molecular weight from as determined by visual inspection, after heating at 57o for 50
5,000 to 3,50,000 with a lower molecular weight for complete hours, when compared to its control consisting of a sample
exclusion of globulin proteins of molecular weight between from the same lot which has not undergone this heating.
4,00,000 and 5,00,000 (Sephadex G 150 is suitable) at 20o to 25o Pyrogens (2.2.8). Complies with the test for pyrogens, using 3
with a saline-phosphate solution prepared by mixing 2 ml per kg of the rabbit’s weight, irrespective of the protein
volumes of saline solution and 1 volume of mixed phosphate content, in rabbits that have not previously received blood
buffer pH 7.0 with azide. Apply to the column 2.5 ml of normal products.
human serum, previously clarified by centrifuging, and elute
with the saline-phosphate solution at a rate of 20 ml per hour. Sterility (2.2.11). Complies with the tests for sterility.
Prepare a chromatogram by recording the absorbance (2.4.7), Abnormal toxicity (2.2.1). Complies with the test for abnormal
of the eluate at about 280 nm in relation to its volume. The toxicity, using Method B and 0.5 ml of the solution for each
chromatogram exhibits three well-defined peaks. Determine mouse and 5 ml for each guinea-pig irrespective of the protein

1454
IP 2007 HUMAN COAGULATION FACTOR IX

content. 60o for 10 hours.

Assay Labelling. The label states (1) the volume in the container; (2)
the total amount of protein in the container expressed in g per
For protein. Dilute to about 0.75 per cent w/v of total protein litre or as percentage; (3) the concentration of sodium and
with saline solution. Take 2 ml of this solution in a round- potassium ions expressed in millimoles per litre; (4) the names
bottomed centrifuge tube, add 2 ml of a 7.5 per cent w/v and concentrations of any stabilising agents and any other
solution of sodium molybdate and 2 ml of a mixture of 30 additives in the final solution; (5) the type of source material
volumes of water and 1 volume of nitrogen-free sulphuric used to manufacture the product; (6) the words “Do not use if
acid. Shake, centrifuge for 5 minutes, decant the supernatant turbid”; (7) that the contents must not be used more than 4
liquid and let the inverted tube stand on a filter paper to drain hours after the container has been penetrated and any remnant
the fluid. Carry out Method E for determination of nitrogen portion must be discarded; (8) the storage conditions; (9) the
(2.3.30), on the residue thus obtained and multiply the result date after which the solution is not intended to be used; (10)
by 6.25 to obtain the protein content. either that the preparation is suitable for administration to
patients undergoing dialysis and to premature infants or that
For sodium. Dilute to 0.01 per cent w/v of protein with water
it is not intended for such purpose.
and determine by Method A for atomic absorption
spectrophotometry (2.4.2), or by Method B for flame
photometry (2.4.4), measuring at about 589 nm and using
sodium solution FP suitably diluted with water as the standard Human Coagulation Factor IX
solution.
Human Coagulation Factor IX is a plasma protein fraction
For potassium. Dilute to 0.25 per cent w/v of protein with
containing coagulation factor IX, prepared by a method that
water and determine by Method A for atomic absorption
effectively separates factor IX from other prothrombin
spectrophotometry (2.4.2), or by Method B for flame
complex factors (factors II, VII and X). It is obtained from
photometry (2.4.4), measuring at about 767 nm and using
human plasma that complies with the monograph on Human
potassium solution FP suitably diluted with water as the
Plasma for Fractionation.
standard solution.
The potency of the preparation, reconstituted as stated on
Human Albumin intended for administration to patients the label, is not less than 20 IU of factor IX per ml.
undergoing dialysis or to premature infants complies with
the following additional test. Production
Aluminium (2.3.8). Not more than 200 µg of Al per litre.
The method of preparation is designed to maintain functional
Determine by atomic absorption spectrophotometry (2.4.2),
with a furnace as atomic generator and measuring at 309.3 nm integrity of factor IX, to minimise activation of any coagulation
factor (to minimise potential thrombogenicity) and includes a
and using as standard solutions a suitable range of dilutions
step or steps that have been shown to remove or to inactivate
in water of aluminium standard solution (10 ppm Al) further
diluted, as necessary, with a solution containing 0.17 per cent known agents of infection; if substances are used for
inactivation of viruses during production, the subsequent
w/v of magnesium nitrate and 0.05 per cent w/v of octoxinol
purification procedure must be validated to demonstrate that
10 in a solution of nitric acid containing 1 per cent w/v of
nitric acid. Prepare suitable dilutions of the preparation under the concentration of these substances is reduced to a suitable
level and that any residues are such as not to compromise the
examination and human albumin for aluminium validation
safety of the preparation for patients.
RS with water. Dilute the solutions, as necessary, with the
magnesium nitrate-octoxinol 10-nitric acid solution used The specific activity is not less than 50 IU of factor IX per mg
for dilution of the standard solution. The test is valid only if of total protein, before the addition of any protein stabiliser.
the aluminium content determined for human albumin for The factor IX fraction is dissolved in a suitable liquid. Heparin,
aluminium validation RS is within 20 per cent of the stated
antithrombin and other auxiliary substances such as a
value.
stabiliser may be included. No antimicrobial preservative is
NOTE — Wash all equipments with a solution containing added. The solution is passed through a bacteria-retentive
20.0 per cent w/v of nitric acid before use and use plastic filter, distributed aseptically into the final containers and
containers only to prepare all solutions. immediately frozen. It is subsequently freeze-dried and the
containers are closed under vacuum or under an inert gas.
Storage. Store protected from light, at a temperature between
2o and 25o. Human Albumin stored at 2o to 8o may be expected Consistency of the method
to continue to meet the requirements of the monograph for 5 The consistency of the method of production is evaluated by
years from the date on which it was heated at 60o for 10 hours. suitable analytical procedures that are determined during
Human Albumin stored at a temperature not exceeding 25o process development and which normally include (1) assay
may be expected to continue to meet the requirements of the of factor IX; (2) determination of activated coagulation factors;
monograph for 3 years from the date on which it was heated at

1455
HUMAN COAGULATION FACTOR VII IP 2007

(3) determination of activities of factors II, VII and X which IX (2.8.8).


shall be shown to be not more than 5.0 per cent of the activity
The estimated potency is not less than 80.0 per cent and not
of factor IX. more than 125.0 per cent of the stated potency. The confidence
Description. A white or pale yellow, hygroscopic powder or limits (P = 0.95) are not less than 80.0 per cent and not more
friable solid. than 125.0 per cent of the estimated potency.
Reconstitute the preparation under examination as stated Storage. Store protected from light.
on the label, immediately before carrying out the Labelling. The label states (1) the number of International
Identification, Tests (except those for solubility and water )
Units of factor IX per container; (2) the amount of protein per
and Assay.
container; (3) the name and quantity of any added substances
including, where applicable, heparin; (4) the name and volume
Identification
of the liquid to be used for reconstitution; (5) that the
It complies with the limits of the Assay. transmission of infectious agents cannot be totally excluded
when medicinal products prepared from human blood or
Tests plasma are administered.

pH (2.4.24). 6.5 to 7.5.


Osmolality (2.4.23). Minimum 240 mosmol per kg.
Human Coagulation Factor VII
Total protein. If necessary, dilute an accurately measured
volume of the preparation under examination with a 0.9 per
cent solution of sodium chloride, to obtain a solution which Human Coagulation Factor VII is a plasma protein fraction
may be expected to contain about 15 mg of protein in 2 ml. To that contains the single-chain glycoprotein factor VII and may
2.0 ml of that solution, in a round-bottomed centrifuge tube, also contain small amounts of the activated form, the two-
add 2 ml of a 7.5 per cent solution of sodium molybdate and 2 chain derivative factor VIIa. It may also contain coagulation
ml of a mixture of 1 volume of nitrogen-free sulphuric acid factors II, IX and X and protein C and protein seconds. It is
and 30 volumes of water. Shake, centrifuge for 5 minutes decant obtained from human plasma that complies with the
the supernatant liquid and allow the inverted tube to drain on monograph on Human Plasma for Fractionation.
filter paper. Determine the nitrogen in the residue by the
method of sulphuric acid digestion (2.3.30) and calculate the The potency of the preparation, reconstituted as stated on
amount of protein by multiplying the result by 6.25. the label, is not less than 15 IU of factor VII per ml.
For some products, especially those without a protein Production
stabiliser such as albumin, this method may not be applicable.
Another validated method for protein determination must The method of preparation is designed to minimise activation
therefore be performed. of any coagulation factor (to minimise potential
thrombogenicity) and includes a step or steps that have been
Activated coagulation factors (2.8.4). If necessary, dilute the
shown to remove or to inactivate known agents of infection;
preparation under examination to contain 20 IU of factor IX
if substances are used for inactivation of viruses during
per ml. For each of the dilutions the coagulation time is not
production, the subsequent purification procedure must be
less than 150 seconds.
validated to demonstrate that the concentration of these
Heparin. If heparin has been added during preparation, substances is reduced to a suitable level and that any residues
determine the amount by the assay of heparin in coagulation are such as not to compromise the safety of the preparation
factor concentrates (2.8.10). The preparation under for patients.
examination contains not more than the amount of heparin
The specific activity is not less than 2 IU of factor VII per mg
stated on the label and in any case not more than 0.5 IU of
of total protein, before the addition of any protein stabiliser.
heparin per International Unit of factor IX.
Water. Determine by semi-micro determination of water The factor VII fraction is dissolved in a suitable liquid. Heparin,
(2.3.43), loss on drying (2.4.19) or near infrared antithrombin and other auxiliary substances such as a
spectrophotometry (2.4.6), the water content is within the limits stabiliser may be added. No antimicrobial preservative is added.
approved by the competent authority. The solution is passed through a bacteria-retentive filter,
distributed aseptically into the final containers and
Sterility (2.2.11). Complies with the test for sterility. immediately frozen. It is subsequently freeze-dried and the
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject containers are closed under vacuum or under an inert gas.
per kg of the rabbit’s mass a volume equivalent to not less Consistency of the method
than 50 IU of factor IX.
The consistency of the method of production with respect to
Assay. Determine the assay of human blood coagulation factor

1456
IP 2007 HUMAN COAGULATION FACTOR VII

the activities of factors II, IX and X of the preparation, 1 IU of heparin). In each of 2 test-tubes, mix equal volumes of
expressed in International Units relative to the activity of factor the reconstituted preparation and a 0.3 per cent solution of
VII, shall be demonstrated. fibrinogen. Keep one of the tubes at 37° for 6 hours and the
other at room temperature for 24 hours. In a third tube, mix a
The consistency of the method of production with respect to
volume of the fibrinogen solution with an equal volume of a
the activity of factor VIIa of the preparation shall be
demonstrated. The activity of factor VIIa may be determined, solution of human thrombin (1 IU per ml) and place the tube
in a water-bath at 37°. No coagulation occurs in the tubes
for example, using a recombinant soluble tissue factor that
containing the preparation under examination. Coagulation
does not activate factor VII but possesses a cofactor function
specific for factor VIIa; after incubation of a mixture of the occurs within 30 seconds in the tube containing thrombin.
recombinant soluble tissue factor with phospholipids reagent Factor II
and the dilution of the test sample in factor VII-deficient
plasma, calcium chloride is added and the clotting time Determine the assay of human coagulation factor II (2.8.5).
determined; the clotting time is inversely related to the factor
The estimated content is not more than 125.0 per cent of the
VIIa activity of the test sample.
stated content. The confidence limits (P = 0.95) are not less
Description. A hygroscopic powder or friable solid that may than 90.0 per cent and not more than 111.0 per cent of the
be white, pale yellow, green or blue. estimated potency.
Reconstitute the preparation under examination as stated
Factor IX
on the label immediately before carrying out the
Identification, Tests (except those for solubility and water ) Determine the assay of human coagulation factor IX (2.8.8).
and Assay.
The estimated content is not more than 125.0 per cent of the
Identification stated content. The confidence limits (P = 0.95) are not less
than 80.0 per cent and not more than 125.0 per cent of the
It complies with the limits of the assay. estimated potency.

Tests Factor X
pH (2.4.24). 6.5 to 7.5. Determine the assay of human coagulation factor X (2.8.9).
Osmolality (2.4.23). Minimum 240 mosmol per kg. The estimated content is not more than 125.0 per cent of the
stated content. The confidence limits (P = 0.95) are not less
Total protein. If necessary, dilute an accurately measured than 90.0 per cent and not more than 111.0 per cent of the
volume of the reconstituted preparation with a 0.9 per cent estimated potency.
solution of sodium chloride to obtain a solution expected to
contain about 15 mg of protein in 2 ml. To 2.0 ml of the solution Water. Determine by semi-micro determination of water (2.3.43),
in a round-bottomed centrifuge tube add 2 ml of a 7.5 per cent loss on drying (2.4.19) or near-infrared spectrophotometry
solution of sodium molybdate and 2 ml of a mixture of 1
(2.4.6), the water content is within the limits approved by the
volume of nitrogen-free sulphuric acid and 30 volumes of
water. Shake, centrifuge for 5 minutes, decant the supernatant competent authority.
liquid and allow the inverted tube to drain on filter paper. Sterility (2.2.11). Complies with the test for sterility.
Determine the nitrogen in the residue by the method of
sulphuric acid digestion (2.3.30) and calculate the amount of Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
protein by multiplying the result by 6.25. per kg of the rabbit’s mass a volume equivalent to not less
than 30 IU of factor VII.
Activated coagulation factors (2.8.4). For each of the dilutions,
the coagulation time is not less than 150 seconds. Assay
Heparin. If heparin has been added during preparation, Determine the assay of human coagulation factor VII (2.8.6).
determine the amount present by the assay of heparin in
coagulation factor concentrates (2.8.10). The preparation under The estimated potency is not less than 80.0 per cent and not
examination contains not more than the amount of heparin more than 125.0 per cent of the stated potency. The confidence
stated on the label and in any case not more than 0.5 IU of limits (P = 0.95) are not less than 80.0 per cent and not more
heparin per International Unit of factor VII. than 125.0 per cent of the estimated potency.

Thrombin. If the preparation under examination contains Storage. Store protected from light.
heparin, determine the amount present as described in the Labelling. The label states (1) the number of International
test for heparin and neutralise the heparin by addition of Units of factor VII per container; (2) the maximum content of
protamine sulphate (10 µg of protamine sulphate neutralises International Units of factor II, factor IX and factor X per

1457
HUMAN COAGULATION FACTOR VIII (rDNA) IP 2007

container; (3) the amount of protein per container; (4) the production, and for control of bulk and final preparation. They
name and quantity of any added substances, including where are derived from representative batches of purified bulk factor
applicable, heparin; (5) the name and volume of the liquid to VIII (rDNA) that are extensively characterised by tests
be used for reconstitution; (6) that the transmission of including those described below and whose procoagulant
infectious agents cannot be totally excluded when medicinal and other relevant functional properties have been
products prepared from human blood or plasma are ascertained and compared, wherever possible, with the
administered. International Standard for factor VIII concentrate. The
reference preparations are suitably characterised for their
intended purpose and are stored in suitably sized aliquots
under conditions ensuring their stability.
Human Coagulation Factor VIII Purified bulk factor VIII (rDNA)
(rDNA) The purified bulk complies with a suitable combination of
the following tests for characterisation of integrity of the
factor VIII (rDNA). Where any substance added during
Human Coagulation Factor VIII (rDNA) is a freeze-dried preparation of the purified bulk interferes with a test, the
preparation of glycoproteins having the same activity as test is carried out before addition of that substance. Where
coagulation factor VIII in human plasma. It acts as a cofactor applicable, the characterisation tests may alternatively be
of the activation of factor X in the presence of factor IXa, carried out on the finished product.
phospholipids and calcium ions. It circulates in plasma
mainly as a two-chain glycosylated protein with 1 heavy Specific biological activity or ratio of factor VIII activity to
(relative molecular mass of about 2,00,000) and 1 light (relative factor VIII antigen
molecular mass 80,000) chain held together by divalent metal
ions. Human coagulation factor VIII (rDNA) is prepared as Determine the assay of human coagulation factor VIII (2.8.7).
full-length factor VIII (octocog alfa), or as a shortened two- The protein content, or where a protein stabiliser is present,
chain structure (relative molecular mass 90,000 and 80,000), in the factor VIII antigen content, is determined by a suitable
which the B-domain has been deleted from the heavy chain method and the specific biological activity or the ratio of factor
(moroctocog alfa). VIII activity to factor VIII antigen is calculated.
Full-length human rDNA coagulation factor VIII contains 25 Protein composition
potential N-glycosylation sites, 19 in the B domain of the
heavy chain, 3 in the remaining part of the heavy chain (relative The protein composition is determined by a selection of
molecular mass 90,000) and 3 in the light chain (relative appropriate characterisation techniques which may include
molecular mass 80,000). The different products are peptide mapping, Western blots, HPLC, gel electrophoresis,
characterised by their molecular size and post-translational capillary electrophoresis, mass spectrometry or other
modification and/or other modifications. techniques to monitor integrity and purity. The protein
composition is comparable to that of the reference preparation.
Production
Molecular size. Using size-exclusion chromatography (2.4.16),
Human coagulation factor VIII (rDNA) is produced by the molecular size distribution is comparable to that of the
recombinant DNA technology in mammalian cell culture. It is reference preparation.
produced under conditions designed to minimise microbial
contamination. Peptide mapping (2.3.47). There is no significant difference
Purified bulk factor VIII (rDNA) may contain added human between the test protein and the reference preparation.
albumin and/or other stabilising agents, as well as other Carbohydrates/sialic acid. To monitor batch-to-batch
auxiliary substances to provide, for example, correct pH and consistency, the monosaccharide content and the degree of
osmolality. sialylation or the oligosaccharide profile are monitored and
The specific activity is not less than 2,000 IU of factor VIII:C correspond to those of the reference preparation.
per mg of total protein before the addition of any protein Final lot
stabiliser, and varies depending on purity and the type of
modification of molecular structure of factor VIII. It complies with the tests under Identification, Tests and Assay.

The quality of the bulk preparation is controlled using Excipients. 80.0 per cent to 120.0 per cent of the stated content,
reference preparations. determined by a suitable method, where applicable.
Reference preparations
Description. A white or slightly yellow powder or friable mass.
During development, reference preparations are established
for subsequent verification of batch consistency during Identification

1458
IP 2007 HUMAN NORMAL IMMUNOGLOBULIN

A. It complies with the limits of the Assay. Regulatory Authority; in particular, tests for hepatitis B surface
antigen, HCV antibodies and for HIV antibodies must be
B. The distribution of characteristic peptide bands
carried out by suitable sensitive methods and must show
corresponds with that of the reference preparation (SDS- negative results in both cases. Plasma, serum or placentae
PAGE or Western blot).
obtained from donors who do not meet all the requirements
stated above may be used as source material provided that it
Tests has been demonstrated to the national authority that process
Reconstitute the preparation as stated on the label of fractionation and production removes any known agent
immediately before carrying out the Tests (except those for capable of adversely affecting the health of subjects treated
solubility and water ) and Assay. with the preparation. No antibiotic is added to the source
materials used for the preparation of immunoglobulin.
pH (2.4.24). 6.5 to 7.5.
It is prepared from pooled material of a minimum volume of 25
Osmolality (2.4.23). Minimum 240 mosmol per kg. litres by a method which has been shown (a) to be capable of
concentrating tenfold from source material at least two different
Water. Determine by semi-micro determination of water (2.3.43), antibodies, one viral and one bacterial, for which an
loss on drying (2.4.19) or near infrared spectrophotometry International Standard or Reference Preparation is available;
(2.4.6), the water content is within the limits approved by the (b) not to affect the integrity of the globulins; (c) to
competent authority. consistently yield a product which is safe for intramuscular
injection and; (d) to yield a product that does not transmit
Sterility (2.2.11). Complies with the test for sterility. viral hepatitis or any other infection.
Bacterial endotoxins (2.2.3). Less than 3 IU in the volume The liquid preparation is prepared as a stabilised solution in
that contains 100 IU of factor VIII activity. saline solution or a 2.25 per cent w/v solution of glycine or
other suitable agent and is sterilised by filtration and
Assay
distributed into previously sterilised containers which are then
Determine the assay of human coagulation factor VIII (2.8.7). sealed so as to exclude micro-organisms. An antimicrobial
preservative may be added except when the preparation is to
The estimated potency is not less than 80.0 per cent and not be freeze-dried. Any antimicrobial preservative or stabilising
more than 125.0 per cent of the stated potency. The confidence agent added must be such that neither deleterious effect on
limits (P = 0.95) are not less than 80.0 per cent and not more the final product in the amounts present nor capability to
than 120.0 per cent of the estimated potency. cause untoward reactions in human beings is demonstrated.
Storage. Store protected from light. An accelerated degradation test is carried out on the final
Labelling. The label states (1) the factor VIII content in liquid or freeze-dried preparation by heating at 37o for 4 weeks.
International Units; (2) the name and amount of any excipient; The difference between the percentages of protein eluted in
(3) the composition and volume of the liquid to be used for the fractions following the main peak as determined by size-
reconstitution. exclusion chromatography (2.4.16), before and after exposure
at 37o does not exceed 5.0 per cent.
Human Normal Immunoglobulin contains not less than 90.0
per cent and not more than 110.0 per cent of the quantity of
Human Normal Immunoglobulin protein stated on the label and in any case, not less than 10.0
Normal Immunoglobulin; Immune Human Serum Globulin; per cent w/v and not more than 18.0 per cent w/v of protein.
Human Gamma Globulin
Description. The liquid preparation is clear pale yellow or
Human Normal Immunoglobulin is a sterile solution or freeze- brownish in colour; on storage it may show turbidity or a
dried preparation containing immunoglobulins, mainly small amount of particulate matter. The freeze-dried preparation
immunoglobulin G (IgG), together with smaller amounts of is a white to slightly yellow powder or solid, friable mass.
other plasma proteins.

Production Identification
A. Precipitation tests with a suitable range of species-specific
It is obtained from source materials such as the blood, plasma,
serum or placentae frozen immediately after collection from antisera which give positive results for the presence of proteins
of human origin and negative results with antisera specific to
healthy donors who must as far as can be ascertained after
plasma proteins of the other species.
clinical examination, laboratory tests on their blood and a study
of their medical history, be free from disease transmissible by B. Examine by electrophoresis (2.4.12), using the moving
transfusion of blood or blood products. The examinations boundary technique and a 1.0 per cent w/v solution in
and tests to be carried out are decided by the National barbitone buffer solution pH 8.6 of ionic strength 0.1. At

1459
HUMAN NORMAL IMMUNOGLOBULIN IP 2007

least 90.0 per cent w/v of the protein has a mobility not greater cent of the total area of the chromatogram represents proteins
than –2.8 x 10-5 cm2V-1S-1. eluted after IgG monomer and albumin (area C).

Tests
pH (2.4.24). 6.4 to 7.2, determined in a solution prepared by
dilution of a quantity with saline solution so as to contain 1.0
per cent w/v of protein.
Protein composition. Determine by cellulose acetate
electrophoresis (2.4.12), but applying an electric field such
that the albumin band of normal human serum applied in a
control strip migrates at least 30 mm and using one strip of
cellulose acetate for each of the following solutions:
Test solution. Dilute the preparation under examination with
saline solution to produce a solution containing 5 per cent w/
v of protein.
Reference solution. Reconstitute human immunoglobulin for
electrophoresis RS with saline solution to produce a solution
containing 5 per cent w/v of protein.
Calculate the result as the mean of three measurements of the Fig.1 Typical Chromatogram for Human Normal
absorbance of each strip. In the electrophoretogram obtained Immunoglobulin
with test solution not more than 10.0 per cent of the protein is
contained in bands other than the principal band. The test is
not valid unless the proportion of protein in the principal
band in the electrophoretogram obtained with reference Stability. Heat approximately 2 ml at 57o for 4 hours in a
solution is within the limits stated in the leaflet supplied with stoppered glass tube (75 mm x 12 mm); no gelation or
human immunoglobulin for electrophoresis RS. flocculation occurs.
Molecular size. Determine by size-exclusion chromatography Pyrogens (2.2.8). Complies with the test for pyrogens, using 1
(2.4.16), applying 2 ml of the preparation under examination ml of the preparation under examination per kg of the rabbit’s
diluted with mixed phosphate buffer pH 7.0 with azide to weight.
produce a solution containing 4.0 to 5.0 per cent w/v of protein.
Sterility (2.2.11). Complies with the tests for sterility.
Chromatographic system
Abnormal toxicity (2.2.1). Complies with the test for abnormal
– a column 1 m x 25 mm packed with agarose trapped toxicity, Method A, injecting 0.5 ml into each mouse and 5 ml
within a cross-linked polyacrylamide network and into each guinea-pig.
having a linear fraction range suitable for fractionation
Other tests. Complies with the tests stated under Parenteral
of globular proteins in the range of molecular weights
Preparations (Injections).
from 20,000 to 350,000,
Assay. Dilute a suitable volume with water to produce a
– mobile phase: mixed phosphate buffer pH 7.0 with solution containing 1.0 per cent w/v of protein. Take 1.5 ml of
azide, the dilution in a round-bottomed centrifuge tube. Add 5 ml of
– flow rate 20 ml per hour (4 ml per cm2 of column water, mix, add 0.2 ml of 7.5 per cent w/v solution of sodium
cross-sectional area), molybdate and 2 ml of a mixture consisting of 1 volume
nitrogen free sulphuric acid and 30 volumes of water. Shake,
– spectrophotometer set at 280 nm. centrifuge for five minutes, decant the supernatant liquid and
Collect the eluate in fractions of about 4 ml. Examine the allow the inverted tube to drain on filter paper. To the residue
chromatogram in comparison with that in Fig.1. The sum of in the tube add three drops of a 30 per cent w/v solution of
the areas of the peaks containing IgG monomer, dimer, albumin copper sulphate and 1 ml of nitrogen-free sulphuric acid
and other proteins of similar molecular size (area B) is not less and boil gently for 10 minutes; cool, add 1 g of anhydrous
than 85.0 per cent of the total area of the chromatogram. Not sodium sulphate and 10 mg of selenium, boil gently for 1 hour
more than 10.0 per cent of the total area of the chromatogram and cool. Transfer to an ammonia distillation apparatus, add 6
represents proteins eluted ahead of IgG dimer (area A); if area ml of a saturated solution of sodium hydroxide and pass
A can be subdivided into two distinct areas, the area steam through the flask; distil for seven minutes, collecting
corresponding to larger proteins does not exceed 5.0 per cent the distillate in a mixture of 5 ml of a saturated solution of
of the total area of the chromatogram. Not more than 5.0 per boric acid, 5 ml of water, and 1 drop saturated solution of

1460
IP 2007 HUMAN PLASMA PROTEIN FRACTION

methyl red in alcohol containing 0.1 per cent of methylene Production


blue, and titrate with 0.02 M hydrochloric acid.
The plasma or serum is obtained from healthy human donors
1ml of 0.02 M hydrochloric acid is equivalent to 0.00175 g of who must, after clinical examination, laboratory tests on their
protein. blood and a study of their medical history, be free from
Freeze-dried Human Normal Immunoglobulin complies with detectable agents of infection transmissible by transfusion of
the following additional requirements. blood or blood derivatives. The examinations and tests to be
carried out are decided by the National Regulatory Authority;
Solubility rate. Add the volume of the liquid stated on the in particular tests for hepatitis B surface antigen and for HIV
label and allow it to stand for 15 minutes at a temperature of antibodies are carried out by suitable sensitive methods and
20o to 25o; it dissolves completely. must give negative results in both cases. Other disease-
Loss on drying (2.4.19). Not more than 2.0 per cent, determined causative agents that are not destroyed or removed by the
on 0.5 g, by drying over phosphorus pentoxide at a pressure processing method must not be present. Plasma or serum
not exceeding 3 Pa for 24 hours. obtained from donors who do not meet all the stated
requirements may be used as source material provided that it
Human Normal Immunoglobulin intended for use in the has been demonstrated to the national authority that the
prevention of infective hepatitis (hepatitis A) complies with process of fractionation will remove any known agent capable
the following additional requirement. of adversely affecting the health of recipients of the Human
Anti-hepatitis A activity. Determine the anti-hepatitis A activity Plasma Protein Fraction.
by comparison with the activity of the Standard preparation, The separation of the protein may be done by precipitation
using an immunoassay of suitable sensitivity and specificity. with suitable organic solvents under controlled conditions,
The stated potency is not less than 100 Units per ml. The particularly of pH, ionic strength and temperature, so that in
estimated potency is not less than the stated potency. The the final product not less than 85.0 per cent of the total protein
fiducial limits of error are not less than 80.0 per cent and not is albumin. Residual solvent, if present, may be removed by
more than 125.0 per cent. freeze-drying or other suitable treatment. Alternative methods
Standard Preparation. The Standard Preparation is the 1st of preparation which shall not affect the integrity of the product
International Reference Preparation for Hepatitis A and shall have been shown to yield consistently a product
immunoglobulin, established in 1981, consisting of freeze-dried which is safe for intravenous injection may be adopted.
material derived from fractionated plasma (supplied in The product is dissolved in water and sufficient quantities of
ampoules containing 100 Units), or another suitable a suitable stabiliser against the effect of heat, like sodium
preparation the antigen binding of which has been determined caprylate in a suitable concentration, and sufficient sodium
in relation to the International Reference Preparation. chloride to adjust the sodium ions to between 130 and 160
Storage. Store protected from light, the liquid preparation in millimoles per litre may be added but no antibiotic or
sealed, colourless, glass containers, at a temperature between antimicrobial preservative is added at any stage during
2o and 8o. Store the freeze-dried preparation under vacuum or preparation. The solution is sterilised by filtration through a
under an inert gas. bacteria-retentive filter and distributed aseptically into sterile
containers which are then closed so as to prevent microbial
Labelling. The label states (1) the volume and the protein
contamination. The solution in its final container is heated at
concentration expressed in g per litre or, for freeze-dried
60o ± 0.5o and maintained at this temperature for 10 hours. The
preparations, the total amount of protein in the container; (2)
containers are then incubated at 30o to 32o for not less than 14
the type of source material; (3) the name and quantity of any
days or at 20o to 25o for not less than 4 weeks and examined
added preservative or stabilising agent; (4) the recommended
visually for evidence of microbial contamination. Those
human dose; (5) that it is meant for intramuscular injection
showing abnormalities such as abnormal colour, turbidity,
only; (6) the storage conditions.
presence of atypical particles or microbial contamination are
discarded.
Human Plasma Protein Fraction contains not less than 95.0
Human Plasma Protein Fraction per cent and not more than 105.0 per cent of the stated amount
of total protein.
Plasma Protein Solution; Human Albumin Fraction (Saline);
Plasma Protein Fraction; PPF Description. A clear, almost colourless or pale yellow liquid;
almost odourless. On storage a dust-like precipitate may
Human Plasma Protein Fraction is a sterile isotonic aqueous develop but it disappears on shaking.
solution of proteins of plasma or serum containing albumin
and globulins. It is prepared as an isotonic solution containing Identification
4.0 to 5.0 per cent w/v of total protein. It contains no fibrinogen A. Precipitation tests with specific antisera show that the
or antibodies. preparation consists only of plasma proteins of human origin
only and gives negative results with antisera specific to plasma

1461
HUMAN PLASMA PROTEIN FRACTION IP 2007

proteins of other species. electrophoresis RS.


B. Examine by a suitable immunoelectrophoresis technique. Sodium. Not less than 95.0 per cent and not more than 105.0
Using antiserum to normal human serum, compare normal per cent of the stated amount and, in any case not more than
human serum and the preparation under examination, both 160 millimoles of Na per litre, determined by Method A for
diluted to contain 1.0 per cent w/v of protein. The main atomic absorption spectrophotometry (2.4.2), measuring at
component of the preparation under examination corresponds 589 nm and using sodium solution AAS suitably diluted with
to the main component of the normal human serum. The water to prepare the standard solutions.
solution may show the presence of small quantities of other Potassium. Not more than 50 µmol of K per g of protein,
plasma proteins.
determined by atomic absorption spectrophotometry (2.4.2),
Tests measuring at 766 nm and using potassium solution AAS,
suitably diluted with water to prepare the standard solutions.
pH (2.4.24). 6.7 to 7.3, determined in a solution prepared by
diluting with sufficient of saline solution to produce a solution Alkaline phosphatase. Not more than 0.1 Unit per g of protein,
containing 1.0 per cent w/v of protein. determined by the following method. Transfer a mixture of 0.5
ml of the substance under examination and 0.5 ml of
Polymers and aggregates. Determine by size-exclusion diethanolamine buffer pH 10.0 to a spectrophotometer cell
chromatography (2.4.16), applying 2 ml of the preparation under maintained at a temperature of 37o ± 0.2o and add 0.1 ml of
examination. nitrophenyl phosphate solution. Record the absorbance of
Chromatographic system the solution at about 405 nm (2.4.7), over a period of at least 30
seconds from the time of addition of the nitrophenyl
– a column 1 m x 25 mm packed with a cross-linked dextran phosphate solution. Calculate the alkaline phosphatase
suitable for fractionation of globular proteins in the range activity at 37o in Units per g of protein from the expression
of molecular weights from 5,000 to 3,50,000 (such as 118.3x/P, where x is the rate of increase of absorbance per
Sephadex G-150), minute and P is the content of total protein in g per litre, as
– mobile phase. mixed phosphate buffer pH 7.0 with determined in the Assay.
azide, Haem content. Dilute with sufficient saline solution to produce
– flow rate 20 ml per hour (4 ml per square centimeter of a solution containing 1.0 per cent w/v of protein; absorbance
column cross-sectional area), of the resulting solution at about 403 nm, not more than 0.15
(2.4.7).
– spectrophotometer set at 280 nm.
Sterility (2.2.11). Complies with the tests for sterility.
Collect the eluate in fractions of about 4 ml and combine the
fractions corresponding to each peak. For each combined Abnormal toxicity (2.2.1). Complies with the test for abnormal
fraction, determine by Method E for determination of nitrogen toxicity, using Method B and 0.5 ml of the solution for each
(2.3.30). mouse and 5 ml for each guinea-pig irrespectiveof the protein
content.
1 ml of 0.02M hydrochloric acid is equivalent to 0.00028 g of
nitrogen. Not more than 10.0 per cent of the total nitrogen is Pyrogens (2.2.8). Complies with the test for pyrogens, using 3
present in the combined fraction associated with non-retained ml per kg of the rabbit’s weight in rabbits that have not
proteins. previously received blood products.
Protein composition. Carry out by cellulose acetate Assay
electrophoresis (2.4.12), Method II using one strip for each
solution. For protein. Dilute to about 0.75 per cent w/v of total protein
Test solution. Dilute the preparation under examination with with saline solution. Take 2 ml of this solution in a round-
saline solution to obtain a solution containing 2.0 per cent bottomed centrifuge tube, add 2 ml of a 7.5 per cent w/v
w/v of protein. solution of sodium molybdate and 2 ml of a mixture of 30
volumes of water and 1 volume of nitrogen-free sulphuric
Reference solution. Dilute human plasma protein fraction acid. Shake, centrifuge for 5 minutes, decant the supernatant
for electrophoresis RS with saline solution to obtain a solution liquid and let the inverted tube stand on a filter paper to drain
containing 2.0 per cent w/v of protein. the fluid. Carry out Method E for determination of nitrogen
Calculate the result as the mean of three measurements of the (2.3.30), on the residue thus obtained and multiply the result
absorbance of each of the 10 strips. In the electrophoretogram by 6.25 to obtain the protein content.
obtained with test solution not more than 15.0 per cent of the Storage. Store protected from light at a temperature between
protein is contained in bands other than the principal band. 2o and 25o.
The test is not valid unless the proportion of protein in the
principal band in the electrophoretogram obtained with Labelling. The label states (1) the volume in the container; (2)
reference solution is within the limits stated in the leaflet the total amount of protein in the container expressed as a
supplied with human plasma protein fraction for percentage or in grams per litre; (3) the concentration of sodium

1462
IP 2007 HUMAN PROTHROMBIN COMPLEX

ions in millimoles per litre; (4) the names and concentrations Tests
of stabilising agents and of any other added substances
present in the final solution; (5) that the solution should not pH (2.4.24). 6.5 to 7.5.
be used if the solution is cloudy or shows a deposit which Osmolality (2.4.23). Minimum 240 mosmol per kg.
does not disappear on shaking; (6) that, once the container
has been penetrated, the contents must be used within 4 hours Total protein. If necessary, dilute an accurately measured
and any unused solution discarded; (7) the storage conditions. volume of the reconstituted preparation with a 0.9 per cent
w/v solution of sodium chloride to obtain a solution expected
to contain about 15 mg of protein in 2 ml. To 2.0 ml of the
solution in a round-bottomed centrifuge tube add 2 ml of a 7.5
Human Prothrombin Complex per cent w/v solution of sodium molybdate and 2 ml of a
mixture of 1 volume of nitrogen-free sulphuric acid and 30
Human Prothrombin Complex is a plasma protein fraction volumes of water. Shake, centrifuge for 5 minutes, decant the
containing blood coagulation factor IX together with variable supernatant liquid and allow the inverted tube to drain on
amounts of coagulation factors II, VII and X; the presence filter paper. Determine the nitrogen in the residue by the
and proportion of these additional factors depends on the method of sulphuric acid digestion (2.3.30) and calculate the
method of fractionation. It is obtained from human plasma amount of protein by multiplying the result by 6.25.
that complies with the monograph on Human Plasma for
Fractionation. Activated coagulation factors (2.8.4). If necessary, dilute the
preparation under examination to contain 20 IU of factor IX
The potency of the preparation, reconstituted as stated on per ml. For each of the dilutions, the coagulation time is not
the label, is not less than 20 IU of factor IX per ml. less than 150 seconds.

Production Heparin. If heparin has been added during preparation,


determine the amount present by the assay of heparin in
The method of preparation is designed to minimise activation coagulation factor concentrates (2.8.10). The preparation under
of any coagulation factor (to minimise potential examination contains not more than the amount of heparin
thrombogenicity) and includes a step or steps that have been stated on the label and in any case not more than 0.5 IU of
shown to remove or to inactivate known agents of infection; heparin per International Unit of factor IX.
if substances are used for inactivation of viruses during Thrombin. If the preparation under examination contains
production, the subsequent purification procedure must be heparin, determine the amount present as described in the
validated to demonstrate that the concentration of these test for heparin and neutralise it by addition of protamine
substances is reduced to a suitable level and that any residues sulphate (10 µg of protamine sulphate neutralises 1 IU of
are such as not to compromise the safety of the preparation heparin). In each of 2 test-tubes, mix equal volumes of the
for patients. reconstituted preparation and a 0.3 per cent w/v solution of
The specific activity is not less than 0.6 IU of factor IX per mg fibrinogen. Keep one of the tubes at 37° for 6 hours and the
of total protein, before the addition of any protein stabiliser. other at room temperature for 24 hours. In a third tube, mix a
volume of the fibrinogen solution with an equal volume of a
The prothrombin complex fraction is dissolved in a
solution of human thrombin (1 IU/ml) and place the tube in a
suitable liquid. Heparin, antithrombin and other auxiliary
water-bath at 37°. No coagulation occurs in the tubes
substances such as a stabiliser may be added. No antimicrobial
containing the preparation under examination. Coagulation
preservative is added. The solution is passed through a
occurs within 30 seconds in the tube containing thrombin.
bacteria-retentive filter, distributed aseptically into the final
containers and immediately frozen. It is subsequently freeze- Water. Determine by semi-micro determination of water
dried and the containers are closed under vacuum or under (2.3.43), loss on drying (2.4.19) or near-infrared spectrometry
an inert gas. (2.4.6), the water content is within the limits approved by the
competent authority.
Description. A white or slightly coloured powder or friable
solid, very hygroscopic. Sterility (2.2.11). Complies with the test for sterility.
Reconstitute the preparation under examination as stated Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
on the label immediately before carrying out the per kg of the rabbit’s mass a volume of the reconstituted
Identification, Tests (except those for solubility and water ) preparation equivalent to not less than 30 IU of factor IX.
and Assay.
Assay
Identification
Factor IX
It complies with the limits of the assay for coagulation factor
IX activity and, where applicable, those for factors II, VII and Determine the assay of human coagulation factor IX (2.8.8).
X. The estimated potency is not less than 80.0 per cent and not

1463
NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE IP 2007

more than 125.0 per cent of the stated potency. The confidence Human normal immunoglobulin for intravenous administration
interval (P = 0.95) of the estimated potency is not greater than is obtained from plasma that complies with the requirements
80.0 per cent to 125.0 per cent. of the monograph on Human plasma for fractionation. No
antibiotic is added to the plasma used.
Factor II
Production
Determine the assay of human coagulation factor II (2.8.5).
The estimated potency is not less than 80.0 per cent and not The method of preparation includes a step or steps that have
more than 125.0 per cent of the stated potency. The confidence been shown to remove or to inactivate known agents of
interval (P = 0.95) of the estimated potency is not greater than infection; if substances are used for inactivation of viruses, it
90.0 per cent to 111.0 per cent. shall have been shown that any residues present in the final
product have no adverse effects on the patients treated with
Factor VII the immunoglobulin.

If the label states that the preparation contains factor VII, The product shall have been shown, by suitable tests in
Determine the assay of human coagulation factor VII (2.8.6). animals and evaluation during clinical trials, to be well tolerated
when administered intravenously.
The estimated potency is not less than 80.0 per cent and not
more than 125.0 per cent of the stated potency. The confidence Human normal immunoglobulin for intravenous administration
interval (P = 0.95) of the estimated potency is not greater than is prepared from pooled material from not fewer than 1,000
80.0 per cent to 125.0 per cent. donors by a method that has been shown to yield a product
that (a) does not transmit infection; (b) at an immunoglobulin
Factor X concentration of 50 g per litre, contains antibodies for at least
2 of which (one viral and one bacterial) an International
Determine the assay of human coagulation factor X (2.8.9). Standard or Reference Preparation is available, the
The estimated potency is not less than 80.0 per cent and not concentration of such antibodies being at least 3 times that in
more than 125.0 per cent of the stated potency. The confidence the initial pooled material; (c) has a defined distribution of
interval (P = 0.95) of the estimated potency is not greater than immunoglobulin G subclasses; (d) complies with the test for
90.0 per cent to 111.0 per cent. Fc function of immunoglobulin.

Storage. Store protected from light. Test for Fc function of immunoglobulin

Labelling. The label states (1) the number of International Stabilised human blood. Collect group O human red blood
Units of factor IX, factor II and factor X per container; (2) into ACD anticoagulant solution. Store the stabilised blood
where applicable, the number of International Units of factor at 4° for not more than 3 weeks.
VII per container; (3) where applicable, that the preparation Phosphate buffered saline pH 7.2. Dissolve 1.022 g of
contains protein C and/or protein S; (4) the amount of protein anhydrous disodium hydrogen phosphate, 0.336 g of
per container; (5) the name and quantity of any added anhydrous sodium dihydrogen phosphate and 8.766 g of
substances, including where applicable, heparin; (6) the name sodium chloride in 800 ml of water and dilute to 1,000 ml with
and quantity of the liquid to be used for reconstitution; (7) the same solvent.
that the transmission of infectious agents cannot be totally
Magnesium and calcium stock solution. Dissolve 1.103 g of
excluded when medicinal products prepared from human blood
calcium chloride and 5.083 g of magnesium chloride in water
or plasma are administered.
and dilute to 25 ml with the same solvent.
Barbital buffer stock solution. Dissolve 207.5 g of sodium
chloride and 25.48 g of barbital sodium in 4,000 ml of water
Normal Immunoglobulin for and adjust to pH 7.3 using 1 M hydrochloric acid. Add 12.5
ml of magnesium and calcium stock solution and dilute to
Intravenous Use 5,000 ml with water. Filter through a membrane filter (pore size
Human Normal Immunoglobulin for Intravenous 0.22 µm). Store at 4° in glass containers.
Administration Albumin barbital buffer solution. Dissolve 0.150 g of bovine
Human Normal Immunoglobulin for Intravenous albumin in 20 ml of barbital buffer stock solution and dilute
Administration is a liquid or freeze-dried preparation containing to 100 ml with water.
immunoglobulins, mainly immunoglobulin G (IgG). Other Tannic acid solution. Dissolve 10 mg of tannic acid in 100 ml
proteins may be present. Human normal immunoglobulin for of phosphate-buffered saline pH 7.2. Prepare immediately
intravenous administration contains the IgG antibodies of before use.
normal subjects. This monograph does not apply to products
intentionally prepared to contain fragments or chemically Guinea-pig complement. Prepare a pool of serum from the
modified IgG. blood of not fewer than 10 guinea-pigs. Separate the serum

1464
IP 2007 NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE

from the clotted blood by centrifugation at about 4°. Store the centrifugation (1,000 g for 10 min) of the cuvette/test-tube
serum in small amounts below -70°. Immediately before starting and remove 1,900 µl of the supernatant. Replace the 1,900 µl
complement-initiated haemolysis, dilute to 125 to 200 CH50 per with albumin barbital buffer solution and repeat the whole
ml with albumin barbital buffer solution and store in an ice- of the washing procedure, finally leaving a volume of 200 µl.
bath during the test. Test samples may be stored in sealed cuvette/test-tubes at 4°
Rubella antigen. Suitable rubella antigen for for 24 hours.
haemagglutination-inhibition titre (HIT). Titre > 256 HA units. Complement-initiated haemolysis. To measure haemolysis,
Preparation of tanned human red blood cells. Separate human add 600 µl of albumin barbital buffer solution warmed to 37°
red blood cells by centrifuging an appropriate volume of to the test sample, re-suspend the cells carefully by repeated
stabilised human blood and wash the cells at least 3 times pipetting (not fewer than 5 times) and place the cuvette in the
with phosphate-buffered saline pH 7.2 and suspend at 2 per thermostatted cuvette holder of a spectrophotometer. After 2
cent v/v in phosphate-buffered saline pH 7.2. Dilute 0.1 ml of minutes, add 200 µl of diluted guinea-pig complement (125
tannic acid solution to 7.5 ml with phosphate-buffered saline to 200 CH50/ml), mix thoroughly by pipetting twice and start
pH 7.2 (final concentration 1.3 mg per litre). Mix 1 volume of immediately after the second pipetting the time-dependent
the freshly prepared dilution with 1 volume of human red recording of absorbance at 541 nm, using albumin barbital
blood cell suspension and incubate at 37° for 10 minutes. buffer solution as the compensation liquid. Stop the
Collect the cells by centrifugation (400 to 800 g for 10 minutes), measurement if absorbance as a function of time has clearly
discard the supernatant and wash the cells once with passed the inflexion point.
phosphate-buffered saline pH 7.2. Resuspend the tanned Evaluation. Determine the slope (S) of the haemolysis curve at
cells at 1.0 per cent v/v in phosphate-buffered saline pH 7.2. the approximate inflexion point by segmenting the steepest
Antigen coating of tanned human red blood cells. Take a section in suitable time intervals Ät (for example, Ät = 1 minute)
suitable volume (Vs) of tanned cells, add 0.2 ml of rubella and calculate S between adjacent intersection points,
antigen per 1.0 ml of tanned cells and incubate at 37° for 30
minutes Collect the cells by centrifugation (400 to 800 g for 10 expressed as ÄA per minute. The largest value for S serves as
minutes) and discard the supernatant, leaving a volume of (Sexp). In addition, determine the absorbance at the start of
200 µl. Add a volume of albumin barbital buffer solution measurement (As) by extrapolating the curve, which is almost
equivalent to the discarded supernatant, resuspend and collect linear and parallel to the time axis within the first few minutes.
the cells as described and repeat the washing procedure. Correct (Sexp) using the expression:
Make up the remaining 200 µl to three-quarters of Vs, thereby
obtaining the initial volume (Vi). Mix 900 µl of albumin barbital '
S exp
buffer solution with 100 µl of Vi, which is thereby reduced to S =
the residual volume (Vr), and determine the initial absorbance As
at 541 nm (A). Dilute Vr by a factor equal to A using albumin
barbital buffer solution, thereby obtaining the final adjusted Calculate the arithmetic mean of the values of S’ for each
volume Vf = Vr × A of sensitised human red blood cells and preparation. Calculate the index of Fc function (IFc) from the
adjusting A to 1.0 ± 0.1 for a tenfold dilution. expression:

( )
Antibody binding of antigen-coated tanned human red blood
cells. Prepare the following solutions in succession and in 100 × S ' − S ' c
duplicate, using for each solution a separate half-micro cuvette I Fc =
S' s − S'c
(for example, disposable type) or test-tube:
Test solutions. If necessary, adjust the immunoglobulin under _
examination to pH 7, for example by addition of 1 M sodium S’ = arithmetic mean of the corrected slope for the
hydroxide. Dilute volumes of the preparation under preparation under examination,
examination containing 30 mg and 40 mg of immunoglobulin _
with albumin barbital buffer solution and adjust the volume S’s = arithmetic mean of the corrected slope for the
to 900 µl. reference preparation,
Reference solutions. Prepare as for the test solutions using _
human immunoglobulin RS. S’c = arithmetic mean of the corrected slope for the
Complement control. 900 µl of albumin barbital buffer complement control.
solution. Calculate the index of Fc function for the preparation under
Add to each cuvette/test-tube 100 µl of sensitised human red examination; the value is not less than that stated in the leaflet
blood cells and mix well. accompanying the reference preparation.
Incubate at room temperature for 15 minutes, add 1,000 µl of Human normal immunoglobulin for intravenous administration
albumin barbital buffer solution, collect the cells by is prepared as a stabilised solution or as a freeze-dried

1465
NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE IP 2007

preparation. A stabiliser may be added. In both cases the 0.9 per cent w/v solution of sodium chloride to an
preparation is passed through a bacteria-retentive filter. The immunoglobulin concentration of 3.0 per cent w/v.
preparation may subsequently be freeze-dried and the Reference solution. Reconstitute human immunoglobulin for
containers closed under vacuum or under an inert gas. No
electrophoresis reference preparation and dilute with a 0.9
antimicrobial preservative is added either during fractionation
per cent w/v solution of sodium chloride to a protein
or at the stage of the final bulk solution. concentration of 3.0 per cent w/v.
The stability of the preparation is demonstrated by suitable
To a strip apply 4 µl of the test solution as a 10 mm band or
tests carried out during development studies. apply 0.4 µl per mm if a narrower strip is used. To another strip
Description. The liquid preparation is clear or slightly apply in the same manner the same volume of the reference
opalescent and colourless or pale yellow. The freeze-dried solution. Apply a suitable electric field such that the albumin
preparation is a hygroscopic, white or slightly yellow powder band of normal human serum applied on a control strip migrates
or solid friable mass. at least 30 mm. Stain the strips with amido black 10B solution
For the freeze-dried preparation, reconstitute as stated on for 5 minutes. Decolourise with a mixture of 10 volumes of
glacial acetic acid and 90 volumes of methanol so that the
the label immediately before carrying out the identification
background is just free of colour. Develop the transparency
and the tests, except those for solubility and water.
of the strips with a mixture of 19 volumes of glacial acetic
Identification acid and 81 volumes of methanol. Measure the absorbance
of the bands at 600 nm in an instrument having a linear response
Examine by a suitable immunoelectrophoresis technique. Using over the range of measurement. Calculate the result as the
antiserum to normal human serum, compare normal human mean of 3 measurements of each strip.
serum and the preparation under examination, both diluted to System suitability. In the electropherogram obtained with the
contain 1.0 per cent w/v of protein. The main component of reference preparation, the proportion of protein in the principal
the preparation under examination corresponds to the IgG band is within the limits stated in the leaflet accompanying
component of normal human serum. The preparation under the reference preparation.
examination may show the presence of small quantities of
other plasma proteins; if human albumin has been added as a Results. In the electropherogram obtained with the test
stabiliser, it may be seen as a major component. solution, not more than 5.0 per cent of protein has a mobility
different from that of the principal band. This limit is not
Tests applicable if albumin has been added to the preparation as a
stabiliser; for such preparations, a test for protein composition
pH (2.4.24). 4.0 to 7.4. is carried out during manufacture before addition of the
Dilute the preparation under examination with a 0.9 per cent stabiliser.
solution of sodium chloride to obtain a solution containing
1.0 per cent of protein. Molecular size. Determine by liquid chromatography (2.4.14).
Osmolality (2.4.23). Minimum 240 mosmol per kg. Test solution. Dilute the preparation under examination with a
0.9 per cent w/v solution of sodium chloride to obtain a
Total protein. Minimum 3.0 per cent w/v and between 90.0 to
concentration in the range of 0.4 to1.2 per cent w/v and
110.0 per cent of the quantity of protein stated on the label.
injection of 50 to 600 µg of protein are usually suitable.
Dilute the preparation under examination with a 0.9 per cent
Reference solution. Dilute human immunoglobulin RS with a
solution of sodium chloride to obtain a solution containing
0.9 per cent w/v solution of sodium chloride to the same
about 15 mg of protein in 2 ml. To 2.0 ml of this solution in a
protein concentration as the test solution.
round-bottomed centrifuge tube add 2 ml of a 7.5 per cent w/
v solution of sodium molybdate and 2 ml of a mixture of 1 Chromatographic system
volume of nitrogen-free sulphuric acid and 30 volumes of – a stainless steel column 60 cm x 7.5 mm or 30 cm x 7.8 mm
water. Shake, centrifuge for 5 minutes, decant the supernatant packed with hydrophilic silica gel,
liquid and allow the inverted tube to drain on filter paper. – mobile phase. dissolve 4.873 g of disodium hydrogen
Determine the nitrogen in the centrifugation residue by the phosphate dihydrate, 1.741 g of sodium dihydrogen
method of sulphuric acid digestion (2.3.30) and calculate the phosphate monohydrate, 11.688 g of sodium chloride
content of protein by multiplying the result by 6.25. and 50 mg of sodium azide in 1000 ml of water.
– flow rate 0.5 ml per minute,
Protein composition. Determine by zone electrophoresis
– spectrophotometer set at 280 nm.
(2.4.12).
In the chromatogram obtained with the reference solution, the
Use strips of suitable cellulose acetate gel as the supporting
principal peak corresponds to IgG monomer and there is a
medium and barbital buffer solution pH 8.6 as the electrolyte
peak corresponding to dimer with a relative retention to the
solution.
principal peak of about 0.85. Identify the peaks in the
Test solution. Dilute the preparation under examination with a chromatogram obtained with the test solution by comparison

1466
IP 2007 NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE

with the chromatogram obtained with the reference solution; of commercial sources.)
any peak with a retention time shorter than that of dimer
Haemolysin. Antiserum against sheep red blood cells prepared
corresponds to polymers and aggregates. in rabbits.
Results. In the chromatogram obtained with the test solution:
Guinea-pig complement. Prepare a pool of serum from the
a. relative retention: for monomer and dimer, the relative blood of not fewer than ten guinea-pigs. Separate the serum
retention to the corresponding peak in the chromatogram from the clotted blood by centrifugation at about 4°. Store the
obtained with the reference solution is 1 ± 02; serum in small amounts below -70°.
b. peak area: the sum of the peak areas of monomer and dimer Method
represent not less than 90.0 per cent of the total area of the Preparation of standardised 5 per cent sheep red blood cell
chromatogram and the sum of the peak area of polymers and
suspension. Separate sheep red blood cells by centrifuging
aggregates represents not more than 3.0 per cent of the total
an appropriate volume of stabilised sheep blood and wash
area of the chromatogram. This requirement does not apply to the cells at least three times with gelatin barbital buffer
products where albumin has been added as a stabiliser; for
solution and prepare as a 5.0 per cent v/v suspension in the
products stabilised with albumin, a test for distribution of
same solution. Measure the cell density of the suspension as
molecular size is carried out during manufacture before follows: add 0.2 to 2.8 ml of water and centrifuge the lysed
addition of the stabiliser.
solution for 5 minutes at 1,000 g; the cell density is suitable if
Anticomplementary activity. The consumption of complement the absorbance (2.4.7) of the supernatant liquid at 541 nm is
is not greater than 50.0 per cent (1 CH50/mg of immunoglobulin). 0.62 ± 0.01. Correct the cell density by adding gelatin barbital
buffer solution according to the formula:
Test for anticomplementary activity of immunoglobulin
Vix A
For the measurement of anticomplementary activity (ACA) of
immunoglobulin, a defined amount of test material (10 mg of Vf = ——————
immunoglobulin) is incubated with a defined amount of 0.62
guinea-pig complement (20 CH 50 ) and the remaining Vf = final adjusted volume,
complement is titrated; the anticomplementary activity is
expressed as the percentage consumption of complement Vi = initial volume,
relative to the complement control considered as 100.0 per A = absorbance of the original suspension at 541 nm.
cent.
The adjusted suspension contains about 1 × 109 cells per ml.
The haemolytic unit of complement activity (CH50) is the
amount of complement that, in the given reaction conditions, Haemolysin titration
will produce the lysis of 2.5 × 108 out of a total of 5 × 108 Prepare haemolysin dilutions as shown in Table 1.
optimally sensitised red blood cells.
Add 1.0 ml of 5.0 per cent sheep red blood cell suspension to
Magnesium and calcium stock solution. Prepare as stated each tube of the haemolysin dilution series, starting at the
earlier. 1:75 dilution, and mix. Incubate at 37° for 30 minutes.
Barbital buffer stock solution. Prepare as stated earlier. Transfer 0.2 ml of each of these incubated mixtures to new
tubes and add 1.10 ml of gelatin barbital buffer solution and
Gelatin solution. Dissolve 12.5 g of gelatin in about 800 ml of 0.2 ml of diluted guinea-pig complement (for example, 1:150).
water and heat to boiling in a water-bath. Cool to 20° and
Perform this in duplicate.
dilute to 10 litres with water. Filter through a membrane filter
(pore size: 0.22 µm). Store at 4°. Use clear solutions only. As the unhaemolysed cell control, prepare three tubes with
1.4 ml of gelatin barbital buffer solution and 0.1 ml of 5.0 per
Citrate solution. Dissolve 8.0 g of sodium citrate, 4.2 g of cent sheep red blood cell suspension.
sodium chloride and 20.5 g of glucose in 750 ml of water.
Adjust to pH 6.1 using a 10.0 per cent w/v solution of citric As the fully haemolysed control, prepare three tubes with 1.4
acid and dilute to 1,000 ml with water. ml of water and 0.1 ml of 5.0 per cent sheep red cell
suspension.
Gelatin barbital buffer solution. Add 4 volumes of gelatin
solution to 1 volume of barbital buffer stock solution and Incubate all tubes at 37° for 60 minutes and centrifuge at 1,000
mix. Adjust to pH 7.3, if necessary, using 1 M sodium hydroxide g for 5 minutes. Measure the absorbance (2.4.7) of the
or 1 M hydrochloric acid. Maintain at 4°. Prepare fresh supernatants at 541 nm and calculate the percentage degree
solutions daily. of haemolysis in each tube using the expression:
Stabilised sheep blood. Collect one volume of sheep blood Aa – A1
into one volume of citrate solution and mix. Store at 4° for not —————— x 100
less than 7 days and not more than 28 days. (Stabilised sheep Ab – A1
blood and sheep red blood cells are available from a number
Aa = absorbance of tubes with haemolysin dilution,

1467
NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE IP 2007

Ab = mean absorbance of the three tubes with full haemolysis, containing 2 MHU/ml and an equal volume of standardised
5.0 per cent sheep red blood cell suspension. Add the
A1 = mean absorbance of the three tubes with no haemolysis.
haemolysin dilution to the standardised cell suspension and
Plot the percentage degree of haemolysis as the ordinate mix. Incubate at 37° for 15 minutes, store at 2° to 8° and use
against the corresponding reciprocal value of the haemolysin within 6 hours.
dilution as the abscissa on linear graph paper. Determine the
optimal dilution of the haemolysin from the graph by Titration of complement. Prepare an appropriate dilution of
inspection. Select a dilution such that further increase in the complement (for example, 1:250) with gelatin barbital buffer
amount of haemolysin does not cause appreciable change in solution and perform the titration in duplicate as shown in
the degree of haemolysis. This dilution is defined as one minimal Table 2.
haemolytic unit (1 MHU) in 1.0 ml. The optimal haemolytic Add 0.2 ml of sensitised sheep red blood cells to each tube,
haemolysin dilution for preparation of sensitised sheep red mix well and incubate at 37° for 60 minutes. Cool the tubes in
blood cells contains 2 MHU per ml. an ice-bath and centrifuge at 1,000 g for 5 minutes. Measure
The haemolysin titration is not valid unless the maximum the absorbance of the supernatant liquid at 541 nm and
degree of haemolysis is 50.0 per cent to 70.0 per cent. If the calculate the degree of haemolysis (Y) using the expression:
maximum degree of haemolysis is not in this range, repeat the Ac – A1
titration with more or less diluted complement solution.
——————
Table - 1
________________________________________________ Ab – A1
Required Prepared using
dilution of
Ac = absorbance of tubes 1 to 12,
haemolysin
Gelatin barbital buffer solution Haemolysin Ab = mean absorbance of tubes with 100 per cent haemolysis,
A1 = mean absorbance of cell controls with 0 per cent
Volume Dilution Volume
haemolysis.
ml (1 : …) ml
_________________________________________________ Plot Y/(1 - Y) as the abscissa against the amount of diluted
7.5 0.65 undiluted 0.1 complement in ml as the ordinate on log–log graph paper. Fit
the best line to the points and determine the ordinate for the
10 0.90 undiluted 0.1 50.0 per cent haemolytic complement dose where Y/(1 - Y) =
75 1.80 7.5 0.2 1.0. Calculate the activity in haemolytic units (CH50/ml) from
the expression:
100 1.80 10 0.2
Cd
150 1.00 75 1.0
———
200 1.00 100 1.0
Ca x 5
300 1.00 150 1.0
400 1.00 200 1.0
600 1.00 300 1.0 Cd = reciprocal value of the complement dilution,
800 1.00 400 1.0 Ca = volume of diluted complement in ml resulting in 50.0 per
cent haemolysis,
1200 1.00 600 1.0
5 = scaling factor to take account of the number of red blood
1600 1.00 800 1.0
cells.
2400 1.00 1200 1.0
The test is not valid unless the plot is a straight line between
3200* 1.00 1600 1.0 15.0 per cent and 85.0 per cent haemolysis and the slope is
4800* 1.00 2400 1.0 0.15 to 0.40, and preferably 0.18 to 0.30.
_______________________________________________
* discard 1.0 ml of the mixture

Preparation of optimised sensitised sheep red blood cells Table - 2


(haemolytic system) _______________________________________________
Prepare an appropriate volume of diluted haemolysin Tube Number Volume of diluted Volume of gelatin

1468
IP 2007 NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE

complement in ml barbital buffer and of gelatin barbital buffer solution are added if the
(for example 1:250) solution in ml immunoglobulin concentration varies from 50 mg per ml; for
_______________________________________________ example, 0.47 ml of gelatin barbital buffer solution is added
to 0.33 ml of immunoglobulin containing 30 mg per ml to give
1 0.1 1.2 0.8 ml. Close the tubes and incubate at 37° for 60 minutes.
2 0.2 1.1 Add 0.2 ml of each incubation mixture to 9.8 ml of gelatin
barbital buffer solution to dilute the complement. Perform
3 0.3 1.0 complement titrations as described above on each tube to
4 0.4 0.9 determine the remaining complement activity (Table 2).
Calculate the anticomplementary activity of the preparation
5 0.5 0.8 under examination relative to the complement control
6 0.6 0.7 considered as 100.0 per cent, from the expression:
7 0.7 0.6 a-b
———————— x 100
8 0.8 0.5
a
9 0.9 0.4
10 1.0 0.3 a = mean complement activity (CH50/ml) of complement control,
11 1.1 0.2
12 1.2 0.1 b = complement activity (CH50/ml) of tested sample.
_______________________________________________ The test is not valid unless:
Three tube as cell - 1.3 a. the anticomplementary activities found for ACA negative
control at 0 per cent control and ACA positive control are within the limits stated
haemolysis in the leaflet accompanying the reference preparation,
_______________________________________________
b. the complement activity of the complement control (a) is in
Three tube at 100 - 1.3 ml of water the range 80 to 120 CH50 per ml.
per cent haemolysis
_______________________________________________ Prekallikrein activator. Maximum 35 IU per ml, calculated
with reference to a dilution of the preparation under examination
Test for anticomplementary activity containing 3.0 per cent w/v of immunoglobulin.

Prepare a complement dilution having 100 CH50 per ml by Test for prekallikrein activator
diluting titrated guinea-pig complement with gelatin barbital
buffer solution. If necessary, adjust the immunoglobulin under Prekallikrein activator (PKA) activates prekallikrein to kallikrein
examination to pH 7. Prepare incubation mixtures as follows and may be assayed by its ability to cleave a chromophore
for an immunoglobulin containing 50 mg per ml: from a synthetic peptide substrate so that the rate of cleavage
can be measured spectrophotometrically and the
concentration of PKA calculated by comparison with a
Table - 3 reference preparation calibrated in International Units.
______________________________________________ The International Unit is the activity of a stated amount of the
International Standard which consists of freeze-dried
Immunoglobulin Complement prekallikrein activator. The equivalence in International Units
under examination control of the International Standard is stated by the World Health
(in duplicate) Organisation.
______________________________________________
Reagents. Prekallikrein activator in albumin RS is calibrated
Immunoglobulin (50 mg per ml) 0.2 ml - in International Units by comparison with the International
Gelatin barbital buffer 0.6 ml 0.8 ml Standard.
Complement 0.2 ml 0..2 ml Buffer A. Dissolve 6.055 g of tris(hydroxymethyl)-
______________________________________________ aminomethane, 1.17 g of sodium chloride, 50 mg of
hexadimethrine bromide and 0.100 g of sodium azide in water.
Carry out the test on the immunoglobulin under examination Adjust to pH 8.0 with 2 M hydrochloric acid and dilute to
1000 ml with water.
and prepare ACA negative and positive controls using human
immunoglobulin RS, as indicated in the leaflet accompanying Buffer B. Dissolve 6.055 g of tris(hydroxymethyl)-
the reference preparation. Higher or lower volumes of sample aminomethane and 8.77 g of sodium chloride in water. Adjust
to pH 8.0 with 2 M hydrochloric acid and dilute to 1,000 ml

1469
NORMAL IMMUNOGLOBULINFOR INTRAVENOUS USE IP 2007

with water. Depending on the method used, ÄA per minutes has to be


Preparation of prekallikrein substrate corrected by subtracting the value obtained for the
corresponding blank without the prekallikrein substrate. The
To avoid coagulation activation, blood or plasma used for
the preparation of prekallikrein must come into contact results may be calculated using a standard curve, a parallel-
line or a slope ratio assay or any other suitable statistical
only with plastics or silicone-treated glass surfaces.
method. Plot a calibration curve using the values thus obtained
Draw 9 volumes of human blood into 1 volume of anticoagulant for the reference preparation and the respective
solution (ACD, CPD or 3.8 per cent w/v solution of sodium concentrations; use the curve to determine the PKA activity
citrate) to which 1 mg per ml of hexadimethrine bromide has of the preparation under examiantion.
been added. Centrifuge the mixture at 3,600 g for 5 minutes.
Separate the plasma and centrifuge again at 6,000 g for 20 Anti-A and anti-B haemagglutinins. Carry out the tests for
anti-A and anti-B haemagglutinins as stated under Dried
minutes to sediment platelets. Separate the platelet-poor plasma
human haemophilic fraction If the preparation under
and dialyse against 10 volumes of buffer A for 20 hours. Apply
the dialysed plasma to a chromatography column containing examination contains more than 3.0 per cent of
immunoglobulin, dilute to this concentration before preparing
agarose-DEAE for ion exchange chromatography which has
the dilutions to be used in the test. The 1 to 64 dilutions do
been equilibrated in buffer A and is equal to twice the volume
of the plasma. Elute from the column with buffer A at 20 ml per not show agglutination.
cm2 per hours. Collect the eluate in fractions and record the Anti-D antibodies. It complies with the test for anti-D
absorbance (2.4.7) at 280 nm. Pool the fractions containing antibodies in human immunoglobulin for intravenous
the first protein peak so that the volume of the pool is about administration.
1.2 times the volume of the platelet-poor plasma.
Test for anti-D antibodies in human immunoglobulin for
Test the substrate pool for absence of kallikrein activity by intravenous administration
mixing 1 part with 20 parts of the pre-warmed chromogenic Materials
substrate solution to be used in the assay and incubate at 37°
for 2 minutes. The substrate is suitable if the increase in Phosphate-buffered saline (PBS). Dissolve 8.0 g of sodium
absorbance is less than 0.001 per minute. Add to the pooled chloride, 0.76 g of anhydrous disodium hydrogen phosphate,
solution 0.7 per cent w/v of sodium chloride and filter using a 0.2 g of potassium chloride, 0.2 g of potassium dihydrogen
membrane filter (porosity 0.45 µm). Freeze the filtrate in portions phosphate and 0.2 g of sodium azide in water and dilute to
and store at -25°; the substrate may be freeze-dried before 1,000 ml with the same solvent.
storage. Papain solution. Use serological grade papain from a
Carry out all procedures from the beginning of the commercial source, the activity of which has been validated.
chromatography to freezing in portions during a single working Red blood cells. Use pooled red blood cells from not fewer
day. than 3 donors of group OR2R2 and 3 donors of group Orr
Method. The assay may be carried out using an automated respectively. Wash the cells 4 times with PBS or until the
enzyme analyser or a suitable microtitre plate system allowing supernatant is clear. Centrifuge the cells at 1, 800 g for 5 minutes
kinetic measurements, with appropriate software for calculation to pack. Treat the packed red cells with papain solution
of results. Standards, samples and prekallikrein substrate may according to the manufacturer’s instructions. Store in
be diluted as necessary using buffer B. Alsever’s solution for not more than 1 week.
Microtitre plates. Use V-bottomed rigid micro-titre plates.
Incubate diluted standards or samples with prekallikrein
substrate for 10 minutes such that the volume of the undiluted Reference standards. Immunoglobulin (anti-D antibodies
sample does not exceed 1/10 of the total volume of the test) reference preparation and Immunoglobulin (anti-D
incubation mixture to avoid errors caused by variation in ionic antibodies test negative control) reference preparation are
strength and pH in the incubation mixture. Incubate the suitable for use as the reference preparation and negative
mixture or a part thereof with at least an equal volume of a control, respectively.
solution of a suitable synthetic chromogenic substrate, known
Method
to be specific for kallikrein (for example, N-benzoyl-L-prolyl-
L-phenylalanyl-L-arginine 4-nitroanilide acetate or D- Reference preparation and negative control solutions.
prolyl-L-phenylalanyl-L-arginine-4-nitroanilide- Reconstitute the reference preparation and the negative
dihydrochloride), dissolved in buffer B. Record the rate of control according to instructions. Dilute the reconstituted
change in absorbance per minute for 2 to 10 minutes at the preparations with an equal volume of PBS containing 0.2 per
wavelength specific for the substrate used. Prepare a blank cent w/v of bovine albumin and then prepare a further 7 serial
for each mixture of sample or standard using buffer B instead two-fold dilutions using PBS containing 0.2 per cent w/v of
of prekallikrein substrate. bovine albumin to give a total dilution range from 1/2 to 1/
256. Make 2 independent sets of dilutions for each preparation.
Add 20 µl of each dilution to the microtitre plate.

1470
IP 2007 PLASMA FOR FRACTIONATION

Test solutions. Initially dilute the test samples to give a starting G present in the preparation; (7) where applicable, the amount
immunoglobulin G (IgG) concentration of 2.5 per cent w/v of albumin added as a stabilizer; (8) the maximum content of
using PBS containing 0.2 per cent w/v of bovine albumin and immunoglobulin A.
then prepare a further 7 serial two-fold dilutions using PBS
containing 0.2 per cent w/v of bovine albumin to give a total
dilution range from 1/2 to 1/256. Make 2 independent sets of
dilutions for each test sample. Add 20 µl of each dilution to Plasma for Fractionation
the microtitre plate.
Human Plasma for Fractionation
Prepare 3.0 per cent v/v suspensions of papain-treated D-
positive (OR2R2) and D-negative (Orr) red cells in PBS Human Plasma for Fractionation is the liquid part of human
containing 0.2 per cent w/v of bovine albumin. Add 20 µl of blood remaining after separation of the cellular elements from
D-positive cells to one dilution series of each of the test sample, blood collected in a receptacle containing an anticoagulant,
the reference preparation and the negative control, and 20 µl or separated by continuous filtration or centrifugation of
of D-negative cells to the other dilution series of each of the anticoagulated blood in an apheresis procedure; it is intended
test samples, the reference preparation and the negative for the manufacture of plasma-derived products.
control. Mix by shaking the plate on a shaker for 10 seconds.
Production
Centrifuge the plate at 80 g for 1 minutes to pellet the cells.
Donors
Place the plate at an angle of approximately 70°. Read after 4 to
5 minutes (or until the cells have streamed in the wells Only a carefully selected, healthy donor who, as far as can be
containing the negative control and the wells where the D- ascertained after medical examination, laboratory blood tests
negative cells have been added). A cell button at the bottom and a study of the donor’s medical history, is free from
of the well indicates a positive result. A stream of cells detectable agents of infection transmissible by plasma-derived
represents a negative result. products may be used.
Record the endpoint titre as the reciprocal of the highest Immunisation of donors
dilution that gives rise to a positive result.
Immunisation of donors to obtain immunoglobulins with
The titre of the preparation under examination is not greater specific activities may be carried out when sufficient supplies
than the titre of the reference preparation. of material of suitable quality can not be obtained from
Water. Determine by semi-micro determination of water naturally immunised donors. Recommendations for such
(2.3.43), loss on drying (2.4.19) or near infrared immunisations are formulated by the World Health
spectrophotometry (2.4.6), the water content is within the limits Organisation (Requirements for the collection, processing
approved by the competent authority. and quality control of blood, blood components and plasma
derivatives, WHO Technical Report Series, No. 840, 1994 or
Sterility (2.2.11). Complies with the test for sterility. subsequent revision).
Pyrogens (2.2.8). Complies with the test for pyrogens. Inject
per kg of the rabbit’s mass a volume equivalent to 0.5 g of Records
immunoglobulin but not more than 10 ml per kg of body mass. Records of donors and donations made are kept in such a way
that, while maintaining the required degree of confidentiality
Antibody to hepatitis B surface antigen concerning the donor’s identity, the origin of each donation
Minimum 0.5 IU per g of immunoglobulin, determined by a in a plasma pool and the results of the corresponding
suitable immunochemical method. acceptance procedures and laboratory tests can be traced.
Storage. For the liquid preparation, store in a colourless glass Laboratory tests
container, protected from light, at the temperature stated on
the label. For the freeze-dried preparation, store in an airtight Laboratory tests are carried out for each donation to detect
colourless glass container, protected from light, at a the following viral markers:
temperature not exceeding 25°. 1. Antibodies against human immunodeficiency virus 1 (anti-
Labelling. The label states (1) for liquid preparations, the HIV-1),
volume of the preparation in the container and the protein 2. Antibodies against human immunodeficiency virus 2 (anti-
content expressed in grams per litre; (2) for freeze-dried HIV-2),
preparations, the quantity of protein in the container; (3) the
amount of immunoglobulin in the container; (4) the route of 3. Antibodies against hepatitis C virus (anti-HCV),
administration; (5) for freeze-dried preparations, the name or 4. Hepatitis B surface antigen (HBsAg).
composition and the volume of the reconstituting liquid to be Pending complete harmonisation of the laboratory tests to be
added; (6) the distribution of subclasses of immunoglobulin
carried out, the competent authority may require that a test for

1471
PLASMA FOR FRACTIONATION IP 2007

alanine aminotransferase (ALT) also be carried out. be achieved, but units of plasma with a lower activity may
still be suitable for use in the production of coagulation
The test methods used are of suitable sensitivity and
specificity and comply with the regulations in force. If a repeat- factor concentrates. The aim of good manufacturing practice
is to conserve labile proteins as much as possible.
reactive result is found in any of these tests, the donation is
not accepted. Total protein. Carry out the test using a pool of not less than
10 units. Dilute the pool with a 0.9 per cent solution of sodium
Individual plasma units
chloride to obtain a solution containing about 15 mg of protein
The plasma is prepared by a method that removes cells and in 2 ml. To 2.0 ml of this solution in a round-bottomed centrifuge
cell debris as completely as possible. Whether prepared from tube add 2 ml of a 7.5 per cent solution of sodium molybdate
whole blood or by plasmapheresis, the plasma is separated and 2 ml of a mixture of 1 volume of nitrogen-free sulphuric
from the cells by a method designed to prevent the introduction acid and 30 volumes of water. Shake, centrifuge for 5 minutes,
of micro-organisms. No antibacterial or antifungal agent is decant the supernatant liquid and allow the inverted tube to
added to the plasma. The containers comply with the drain on filter paper. Determine the nitrogen in the residue by
requirements for plastic containers for blood and blood the method of sulphuric acid digestion (2.3.30) and calculate
components (6.2). The containers are closed so as to prevent the protein content by multiplying the quantity of nitrogen by
any possibility of contamination. 6.25. The total protein content is not less than 5.0 per cent.
If 2 or more units are pooled prior to freezing, the operations
Factor VIII
are carried out using sterile connecting devices or under
aseptic conditions and using containers that have not Carry out the test using a pool of not less than 10 units. Thaw
previously been used. the samples under examination, if necessary, at 37°. Determine
When obtained by plasmapheresis, plasma intended for the the assay of factor VIII (2.8.7), using a reference plasma
recovery of proteins that are labile in plasma is frozen by calibrated against the International Standard for human
cooling rapidly in a chamber at - 30° or below as soon as coagulation factor VIII in plasma. The activity is not less than
possible and at the latest within 24 hours of collection. 0.7 IU per ml.
When obtained from whole blood, plasma intended for the Pooled plasma
recovery of proteins that are labile in plasma is separated During the manufacture of plasma products, the first
from cellular elements and is frozen by cooling rapidly in a homogeneous pool of plasma (for example, after removal of
chamber at -30° or below as soon as possible and at the latest cryoprecipitate) is tested for HBsAg, HCV antibodies and for
within 24 hours of collection. HIV antibodies using test methods of suitable sensitivity and
When obtained from whole blood, plasma intended solely for specificity; the pool must give negative results in these tests.
the recovery of proteins that are not labile in plasma is The plasma pool is also tested for hepatitis C virus RNA using
separated from cellular elements and frozen in a chamber at - a validated nucleic acid amplification technique (2.8.1). A
20° or below as soon as possible and at the latest within 72 positive control with 100 IU per ml of hepatitis C virus RNA
hours of collection. and, to test for inhibitors, an internal control prepared by
It is not intended that the determination of total protein and addition of a suitable marker to a sample of the plasma pool
factor VIII shown below be carried out on each unit of plasma. are included in the test. The test is invalid if the positive
They are rather given as guidelines for good manufacturing control is non-reactive or if the result obtained with the internal
practice, the test for factor VIII being relevant for plasma control indicates the presence of inhibitors. The plasma pool
intended for use in the preparation of concentrates of labile complies with the test if it is found non-reactive for hepatitis C
proteins. virus RNA.
The total protein content of a unit of plasma depends on the Description. Before freezing, a clear to slightly turbid liquid
serum protein content of the donor and the degree of dilution without visible signs of haemolysis; it may vary in colour
inherent in the donation procedure. When plasma is obtained from light yellow to green.
from a suitable donor and using the intended proportion of Storage. Frozen plasma is stored in conditions designed to
anticoagulant solution, a total protein content complying maintain the temperature at or below -20°; for accidental
with the limit of 5 per cent is obtained. If a volume of blood or reasons, the storage temperature may rise above -20° on one
plasma smaller than intended is collected into the or more occasions during storage but the plasma is
anticoagulant solution, the resulting plasma is not nevertheless considered suitable for fractionation if all the
necessarily unsuitable for pooling for fractionation. The aim following conditions are fulfilled (1) the total period of time
of good manufacturing practice must be to achieve the during which the temperature exceeds -20° does not exceed 72
prescribed limit for all normal donations. hours; (2) the temperature does not exceed -15° on more than
Preservation of factor VIII in the donation depends on the one occasion; (3) the temperature at no time exceeds -5°.
collection procedure and the subsequent handling of the Labelling. The label enables each individual unit to be traced
blood and plasma. With good practice, 0.7 IU/ml can usually to a specific donor.

1472
IP 2007 PLATELET CONCENTRATE

Platelet Concentrate 22°, label it also to state that a continuous gentle agitation
shall be maintained, or where labelled for storage at 1° to 6°, to
Platelets separated from whole blood within 4 to 6 hours of state that such agitation is optional. Label it also with the type
collection and suspended in 40 to 50 ml of plasma are and result of a serologic test for syphilis, or to indicate that it
designated as platelet concentrate. was nonreactive in such test; with the type and result of a test
for hepatitis B surface antigen, or to indicate that it was
Production nonreactive in such test; with a warning that it is to be used as
soon as possible but not more than 6 hours after entering the
Platelet Concentrate is prepared from units of whole blood container; to state that a filter is to be used in the administration
that have not been allowed to cool below 20°. Platelet rich equipment; and to state that the instruction circular provided
plasma (PRP) is separated within 4-6 hours after completion is to be consulted for directions for use.
of the phlebotomy. The final component should contain
resuspended platelets in an amount of plasma adequate to
maintain an acceptable pH - generally 40 to 70 ml is used.

Tests
Whole Human Blood
Whole Blood (Human)
Four PC per month should be assayed for pH and for platelet,
RBC and leucocyte counts. Whole Human Blood is blood drawn aseptically from selected
human donors and mixed with a suitable anticoagulant.
PRP Whole Human Blood is the final mixture of blood and
Counts 3.0 - 4.5 x 105 per µl (approximately) anticoagulant solution contains not less than 9.7 per cent
Volume 170 - 250 ml (approximately) w/v of haemoglobin, calculated from the haemoglobin content
of the donor’s blood and the dilution due to the anticoagulant
Total Count 9 x 1010 per bag (approximately)
solution. It is obtained from healthy donors who must:
(a) be in the age range of 18 to 60 years and be in good health
PRC as indicated in part by normal temperature and blood pressure
within normal limits;
Counts 8 - 10 x 105 per µl 350 ml bag
(approximately) (b) not be pregnant, if females;
10 - 12 x 105 per µl 450 ml bag (c) not have undergone major surgery within 6 months of
(approximately) donation;
Volume 40 - 50 ml per bag (d) as far as can be ascertained after clinical and laboratory
examination and the study of medical history of the donor be
Platelet counts > 5.5 x 1010 per bag in 75 per cent of bags free from disease transmissible by blood transfusion;
( if prepared from 450 ml bags )
(e) have blood containing not less than 12.5 per cent w/v of
> 4.2 x 1010 per bag in 75 per cent of bags haemoglobin;
( if prepared from 350 ml bags )
(f) be free from acute respiratory diseases;
Leucocyte count < 0.12 x 109 per bag.
(g) be free from any infectious skin disease at the site of
RBC counts < 1.2 x 109 per bag. phlebotomy;
pH < 6.3 at the end of 5 days storage. (h) have no history of malarial fever within 12 months of
donation;
Expiration time. The expiration time is not more than 72 hours
from the time of collection of the source material. (i) have no history of viral hepatitis or of close contact with an
individual having viral hepatitis within 12 months of donation
Storage. Store at 20° to 22° in polyvinylchloride plastic bags. and have blood that has given negative results in tests for the
Preserve at the temperature relevant to the volume of presence of hepatitis-B antigen;
resuspension plasma, either between 20° and 22° or between
1° and 6°, the latter except during shipment, when the (j) have blood that has been tested with negative results
temperature may be between 1° and 10°. for evidence of syphilitic infection, HCV antibodies, HIV
antibodies and malarial parasites.
Labelling. In addition to the labelling requirements of Whole
Blood applicable to this product, label it to state the volume of The examinations and tests to be carried out are decided by
original plasma present, the kind and volume of anticoagulant the National Regulatory Authority.
solution present in the original plasma, the blood group The frequency of donations of whole blood shall not exceed
designation of the source blood, and the hour of expiration on once every 3 months with a maximum volume of 1.5 litres in
the stated expiration date. Where labelled for storage at 20° to any consecutive 12 month period.

1473
WHOLE HUMAN BLOOD IP 2007

The blood is drawn aseptically through a closed system into haemolysis, with a greyish layer between the two consisting
a suitable sterile container containing a specific amount of of leucocytes and thrombocytes. A layer containing emulsified
Anticoagulant Citrate Dextrose Solution (ACD Solution) or fat may form on the surface.
Anticoagulant Citrate Phosphate Dextrose Solution (CPD
Solution) which is placed before the container is sterilised. Tests
The quantity of anticoagulant solution should not exceed
22.0 per cent v/v of the final volume of the mixture. No Sterility (2.2.11). Complies with the tests for sterility,
antimicrobial preservative is added. determined by Method B.
During the withdrawal of blood the container is gently agitated Assay. Determine the haemoglobin content by photometric
to ensure thorough mixing. When withdrawal is complete the haemoglobinometry (2.8.12).
container is immediately sealed and cooled to 2o to 8o. It is not
opened until immediately before transfusion. With every Storage. Store in colourless, transparent and sterile containers
container of blood, a separate sample mixed with the into which it was originally drawn. The containers should be
appropriate quantity of anticoagulant solution, is collected provided with a hermetic, contamination-proof closure. Store
for compatibility and other tests; this small container is firmly at a temperature between 2o to 8o.
attached to the main container. Labelling. The label states (1) the distinctive code number by
Whole Human Blood in containers from which samples have reference to which the details of the donor are available; (2)
been removed for tests should not be used for transfusion. the ABO group with the approved colour scheme for different
Consequently, it is not intended that the Tests and the Assay groups as specified by the National Regulatory Authority; (3)
should be carried out on the contents of the container. The the Rh group; (4) the total volume of fluid, the proportion of
Blood Bank or the service collecting the blood is responsible blood, and the nature and volume of anticoagulant solution;
for ensuring that the conditions in which the blood is collected (5) the date on which the blood was withdrawn; (6) the expiry
and stored are such that, if and when tested, the blood will date which should not exceed 21 days from the date of
comply with the requirements of the monograph. withdrawal of blood; (7) the storage conditions; (8) that the
blood must not be used for transfusion if there is any visible
Blood group. Determine the blood group (in the sample evidence of haemolysis or other deterioration; (9) for blood of
accompanying each donation) under the ABO system (2.8.11), group O whether haemolysins are present or not and if they
by examination of both corpuscles and serum, and under the are, that the blood must be administered only to recipients of
Rh system (2.8.11), by examination of the corpuscles. blood group O; (10) that the blood has given negative results
Description. Deep red fluid which, on standing separates into in the tests for the presence of malarial parasites, hepatitis-B
a lower layer of sedimented-red cells and a yellowish, almost antigen, syphilis and HIV antibodies and any other tests
clear upper layer of plasma, free from visible signs of prescribed by the National Regulatory Authority.

1474
INDIAN PHARMACOPOEIA 2007 MONOGRAPHS

BIOTECHNOLOGY PRODUCTS

General Monographs
r-DNA Biotechnology Products of Therapeutic Value ....
Monographs
Erythropoietin concentrated solution ....
Granulocyte Colony Stimulating Factor, Human ( Filgrastim ) ....
Interferon alfa-2 concentrated solution ....
Sereptokinase ....

1475
IP 2007 r-DNA BIOTECHNOLOGY PRODUCTS OF THERAPEUTIC VALUE

r-DNA Biotechnology Products of hybridoma cells using various techniques yield monoclonal
antibodies.
Therapeutic Value
r-DNA derived products suffer from a danger of micro-
This monograph states the general requirements for the heterogeneity in the expressed protein and therefore require
manufacture of products of recombinant DNA technology extensive process validation. In addition, the concern of
that are produced by genetic modification in which the DNA adventitious viruses , presence of host cell DNA, presence of
coding for the required product is introduced into a suitable oncogenic genes, residual host proteins, endotoxins, residual
cell line or microorganism by means of a plasmid or viral vector. media proteins etc calls for stringent methods of testing to
The DNA is expressed and translated into protein of interest ensure their removal to acceptable levels.
in the genetically modified organism. r-DNA derived products are not significantly different from
Therapeutic proteins are derived from several processes. One other therapeutic proteins derived from natural sources after
of them is the recombinant DNA technology, the products the final purification step and therefore basic requirements of
of which are often referred to as r-DNA products. The process validation of the process, environmental conditions, and
involves isolation of a specific active gene and inserting into aseptic techniques during manufacturing, quality assurance
a host cell. The host cell expresses the protein encoded in the and quality control do not differ in approach from conventional
transferred gene. The host cell could be a microorganism or pharmaceutical products. Handling of genetically modified
an animal cell. Large-scale propagation yields large quantities organisms in the production process require adherence to the
of crude protein. Purification of the crude protein using multiple rules governing environmental safety.
techniques derives a safe, pure and biologically active product. r-DNA products are produced in Prokaryotic or Eukaryotic
The product being a protein may cause immunological systems. The choice of the system primarily depends upon
sensitization in the recipients and therefore needs a high the size of the protein and the extent of glycosylation required
degree of characterization. Testing of the product for safety, to make them biologically active. The choice of bacteria in the
identity, strength, quality and purity are similar to those applied Prokaryotic system is E.Coli due to the extensive information
to other pharmaceutical products, although the tests available of this bacterium.
themselves may require some modification due to the The disadvantages in use of the organism are:
pretentious nature of the active compound. 1) The proteins are produced in the reduced state.
Recombinant DNA technology products are produced by 2) Need to remove the N formyl methionine sequence.
genetic manipulation of a cell line or microorganism usually 3) Product degradation during the production process due
through a plasmid transfer or viral vector. The recovery of the to the presence of protease enzyme.
desired product expressed by such genetically modified
4) Need for endotoxin removal step.
organisms is by multistage purification. Monoclonal
antibodies too fall under this category. Eukaryotic cells including yeasts on the other hand have the
ability to produce proteins that are properly folded due to its
The cell or microorganism used for expressing the protein is glycosylation ability. Primary cell lines have a short life span.
referred to as the host cell and the transformed cell after the Cloning, selection, amplification, master cell bank creation,
gene insertion is referred to as the host-vector system. scaling up requires several passages. Primary Cell lines do
The host cells could be Prokaryotic or Eukaryotic cells. The not last these many passages. Use of immortal cell lines has
differences in their metabolic pathway result in differences in helped overcome the problem but brought in new challenges
the type proteins expressed by them. Prokaryotic cells do not in the form of oncogenes, potential viral and retroviral
have the ability to do glycosylation. Therefore, proteins contamination, and presence of animal derived growth
expressed by the Prokaryotic cells may require post translation promoters etc., which are required to be removed by various
modification to make them biologically active. purification steps.
Monoclonal antibodies are derived from a single clone of B Exhaustive analysis, validation of the process and safety tests
lymphocyte .They differs from polyclonal antibodies that are are done on the Master Banks are done to rule out presence of
a mixture of antibodies of several antigenic epitopes. B- adventitious agents. Cells are used only after this for large-
lymphocytes have a short life span and hence need to be scale production.
immortalized if they are to be used for production for a long Yeast offers an advantage against the complexities of using
period. Fusion with myeloma cells make them immortal. Such animal cell lines but have the disadvantage that they maintain
fused hybridoma cells inherit the property of producing the plasmids extra chromosally creating a danger of losing the
desired antibody from the B lymphocyte and immortal property inserted gene. However, their ability to produce glycosylated
from the Myeloma cells. Large scale culturing of these protein makes them a better choice than Escherichia coli.

1
r-DNA BIOTECHNOLOGY PRODUCTS OF THERAPEUTIC VALUE IP 2007

Monoclonal antibodies are produced in two ways depending iii. The method of transfer of the expression gene into the
on the source of the lymphocyte – murine or Human. host cell.
Antibodies of Murine origin are produced by selecting the iv. Methods employed in the clone selection.
lymphocytes from the spleen of previously inoculated mice or
v. Amplification role of various components like promoter,
rats and fusing them with an immortal cell line like myeloma
enhancers, antibiotic resistance gene etc.
cells. Such hybridoma cells are cloned, propagated in large
scale and used for antibody production. Antibody produced vi. A complete annotated sequence of the plasmid indicating
is as per the chromosomal information acquired during the those regions that have been sequenced.
hybridoma formation process. Characterization of the cell bank system
Large-scale propagation of bacteria and Yeast are often The master cell bank is a collection of vials containing
referred to as Fermentation. During the process the growth homogeneous suspension of the original cells already
rate, purity of the culture and yields are monitored. Nucleotide transformed by the expression vector containing the desired
sequence analysis or DNA restriction mapping checks the gene. One or more of the Master Cell Bank vials may be
genetic stability of the plasmid. Peptide mapping of the amplified for creating a Working Cell Bank. The cell banks
expressed protein is carried out for confirmation. Proteolytic should be characterized for relevant phenotypic and
degradation of the expressed protein is a concern and therefore genotypic markers, by expressing the recombinant protein
optimization of the fermentation process is essential to and confirming the nucleotide sequence.
minimize it. Recovery of the protein often requires lysis of the
cells. Proteases released during the process of cell lysis calls The number of passages from the Master Bank level or the
for rapid processing to minimize the damage caused by it. cell doubling permitted during the production process will be
determined from the data derived from long tem cultivation.
Large-scale cell culture is quite well established but poses it Based on the analysis of the molecular integrity of the protein-
own challenges like genetic stability, protein folding and expressing gene, the phenotypic and the genotypic
culture conditions. Genetic stability cannot be rapidly checked characteristics of the host cell, passage or cell doubling limits
as in the case of Prokaryotic as the gene is incorporated in the are established.
cell genome. Peptide mapping of the expressed protein is the
only check but lacks sensitivity to detect minor genetic Analysis of the cell bank systems for copy number, insertions
mutations. Manufacturers are required to demonstrate the and deletions and the number of integration sites is done.
purity of the cultures against contaminating organisms When the expression system is extra chromosomal, the
including mycoplasma and adventitious viruses, at the end of percentage of the host cells retaining the expression system
purification. The advantage of cell culture is that the protein should be determined. The nucleic acid sequence should be
is directly secreted into the medium and therefore cell identical and should correspond to that expected for the
separation is sufficient to achieve a significant level of protein sequence.
purification. Characterization of the host-vector system and the final
purified protein are done to ensure consistent production of
Production the r-DNA protein.
Production is based on a validated stable host –vector
combination using a seed-lot system. The seed-lot system Validation of the cell banks
uses a master cell bank and a working cell bank derived from Validation of the cell banks shall include:
the master seed lot of the host-vector combination. The i. Stability by measuring viability and the retention of the
validation to determine the suitability shall include the vector.
following.
ii. Identity of the cells by phenotypic features.
Characterization of the Host-Vector System iii. Establishing that the cell banks are free from potentially
Characterization of the host-vector system is used for oncogenic or infective adventitious agents (viral,
establishing its suitability for production. Characterization of bacterial, fungal or mycoplasma).
the host-vector system involves several tests that are iv. For mammalian cells, details of the tumorogenic potential
performed to establish the purity. of the cell bank shall be obtained.
Characterization includes documentation of: Validation of the purification process
i. The origin of the nucleotide sequence coding for the The ability of the different steps of the extraction and
protein. purification process to remove and/or inactivate contaminating
ii. The methods used to prepare the DNA coding. substances derived from the host cell or culture medium,

2
IP 2007 ERYTHROPOIETIN CONCENTRATED SOLUTION

including, virus particles, proteins, nucleic acids and added liquid chromatography, capillary electrophoresis or sodium
substances, must be validated. Validation studies should dodecyl sulphate polyacrylamide gel electrophoresis.
demonstrate that the production process routinely meets the
following criteria: Host-cell-derived proteins
i. Removal of extraneous agents from the product. Host-cell-derived proteins are detected by immunochemical
ii. Removal of vector, host-cell, culture medium and reagent- methods, using, polyclonal antisera rose against protein
derived contaminants below the acceptable levels. components of the host-vector system. Liquid-phase
displacement assays (radioimmunoassay), liquid-phase direct-
iii. Capability to reduce the DNA below acceptable levels.
binding assays and direct-binding assays using antigens
iv. Adequate stability of any partially purified product if immobilized on nitrocellulose (or similar) membranes (for
stored during the process. example, dot-immunoblot assays, Western blots) may be used
v. Minimum yields in the process. for the assay.

Characterization of the substance The Antisera are raised against a preparation of antigens
derived from the host organism, into which has been inserted
The identity, purity, potency and stability of the final bulk the vector used in the manufacturing process that lacks the
product are established initially by carrying out a wide range specific gene coding for the product. This host cell is cultured,
of chemical, physical, immunochemical and biological tests. and proteins are extracted, using conditions identical to those
Prior to release, each batch of the product is tested by the used for culture and extraction in the manufacturing process.
manufacturer for identity and purity and an appropriate assay Partly purified preparations of antigens, using some of the
is carried out. purification steps in the manufacturing process, may also be
Production Consistency used for the preparation of antisera.

Suitable tests for demonstrating the consistency of the Host-cell- and vector-derived DNA
production and purification are performed. The tests include, Residual DNA is detected by hybridization analysis, using
especially characterization tests, in-process controls and final- suitably sensitive, sequence-independent analytical
product tests, for example: techniques or other suitably sensitive analytical techniques.
However, the method used must be validated to ensure
Amino-acid composition
parallelism with the DNA standard used, linearity of response
Partial amino acid sequence analysis test for confirmation of and non-interference of either the drug substance or excipients
the correct N-terminal processing and detect loss of the C- of the formulation at the dilutions used in the assay.
terminal amino acids. The limits for the presence of these are as currently prescribed
Peptide mapping by W.H.O. or competent authorities.

Peptide mapping using chemical and/or enzymatic cleavage


of the protein product must be done that no significant
difference between the test protein and reference preparation Erythropoietin Concentrated Solution
is found. Two-dimensional gel electrophoresis, capillary
electrophoresis or liquid chromatography may be used for
analysis. Peptide mapping can also be used to demonstrate APPRLICDSR VLERYLLEAK EAENITTGCA
correct disulphide bonding.
EHCSLNENIT VPDTKVNFYA WKRMEVGQQA
Determination of molecular mass
VEVWQGLALL SEAVLRGQAL LVNSSQPWEP
Cloned-gene Retention
LQLHVDKAVS GLRSLTTLLR ALGAQKEAIS
The relevant authority approves the minimum amount in
percentage of the cells containing the vector or the cloned PPDAASAAPL RTITADTFRK LFRVYSNFLR
gene after cultivation.
GKLKLYTGEA CRTGD
Total Protein. The yield of protein is determined.
Chemical Purity Mol. Wt. 30,600 (approx)
The purity of the protein product is analyzed in comparison Erythropoietin Concentrated Solution contains a family of
with a reference preparation by a suitable method such as closely-related glycoproteins which are not different from the

3
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2007

naturally occurring human erythropoietin (urinary Pour into a suitable gel cassette, with approximate dimensions
erythropoietin) in terms of the amino acid sequence (165 amino of 15 cm x 15 cm x 0.05 cm. Insert a suitable sample well comb
acids) and average glycosylation pattern, at a concentration and allow to polymerize.
of 0.5 mg per ml to 10 mg per ml. It has a potency of not less
Use as the anode solution the anolyte for isoelectric focusing
than 100000 IU per mg of active substance.
pH 3 to 5 and as the cathode solution the catholyte for
Production isoelectric focusing pH 3 to 5. Allow prefocusing to take
place for 1 hour at a constant power of 10 W, with maximum
Erythropoietin is produced in rodent cells in vitro by a method voltage and current settings of 2000 V and 100 mA respectively.
based on recombinant DNA technology. All batches are tested
Dilute the test solution to 0.5 mg per ml with water. Apply to
as described below, prior to release.
the gel 10 µl of each solution. Carry out focusing for a further
Host cell-derived proteins 30 minutes at the same power supply settings. Take the gel
out of the focusing chamber, and transfer the proteins onto a
The limit is as prescribed by WHO.
membrane suitable for immobilization of proteins (such as
Host cell-derived proteins (HCP). This must be routinely Polyvinylidene Fluoride), using commercially available
monitored using a scientifically accepted method that electrotransfer equipment and following the manufacturer’s
demonstrates that: instructions. After electrotransfer, incubate the membrane in a
1. the assay is sensitive (a useful target for the limit of neutral isotonic buffer containing a suitable blocking agent
detection is 1 to 100 ppm) (for example, 50 g per litre of dried milk), for 1 hour, followed by
2. the assay is specific for the HCP (defined by proprietary incubation in the same blocking solution with a suitable dilution
detection reagents such as antibodies elicited against of a polyclonal anti-erythropoietin antibody. Detect the
process-specific contaminating HCP) consistently found erythropoietin-bound antibody using a suitable enzyme- or
in the product from a specific manufacturing / purification radiolabelled antibody (for example, an alkaline phosphatase-
process. conjugated second antibody). The precise details of blocking
details of blocking agents, concentrations and incubation times
Acceptable limits should be set empirically based on the above should be optimized using the principles set out in
by the manufacturer. Immunochemical methods.
Host cell- and vector-derived DNA The test is not valid unless the distribution of bands in the
The limit is as prescribed by the regulatory authority. electrophoretogram obtained with the reference solution
contains at least 6 well separated bands. If necessary, the
Description. A clear and colourless solution. voltage settings and duration may be altered to optimize the
separation of the isoforms.
Identification
In the electrophoretogram obtained with the test solution, the
A. It gives the appropriate response when examined using the pH range of the bands observed corresponds to that of the
conditions described under Assay. electrophoretogram obtained with the reference solution. The
B. Determine by capillary electrophoresis (capillary isoelectric predominant bands correspond to isoforms 4, 5, 6 and 7.
focusing) (2.4.32). Additional, fainter bands corresponding to isoforms 1, 2, 3
Test solution. Dilute if necessary the preparation in water and and 8 may also be present. Other bands are present in no more
desalt the preparation. Using a membrane filtration system than trace amounts.
suitable for desalting proteins, desalt the sample. Make up C. Determine by capillary electrophoresis (capillary zone
the volume to the original volume with water. electrophoresis) (2.4.32).
Reference solution. Dissolve erythropoietin RS in water to All the solutions should be filtered through a 0.45 µm membrane
produce a solution containing 1mg per ml and desalt as above. filter before use.
The isoelectric focusing procedure may be carried out using a Test solution. Dilute the substance under examination with
0.5mm thick polyacrylamide slab gel containing ampholytes water or concentrate it to obtain a concentration of 1 mg per
covering the pH of range 3 to 10, prepared as follows. ml. Desalt 0.25 ml of the solution by passage through a micro-
Mix 9 g of urea, 6.0 ml of 30 per cent acrylamide / concentrator cartridge provided with a membrane with a
bisacrylamide solution, 1.05 ml of pH 3 to 5 ampholyte, 0.45 molecular mass cut-off of not more than 10000. Add 0.2 ml of
ml pH 3 to 10 ampholyte and 13.5 ml of water. Degas. Add 15 water to the sample and desalt again. Repeat the desalting
µl of tetramethylethylene diamine and 0.3 ml of a 100 g per procedure once more. Dilute the sample with water, determine
litre freshly prepared solution of ammonium persulphate. its protein concentration as described under Tests and adjust

4
IP 2007 ERYTHROPOIETIN CONCENTRATED SOLUTION

to a concentration of approximately 1 mg per ml with water. Preconditioning of the capillary. Rinse the capillary for 60
Reference solution. Dissolve erythropoietin RS in water to minutes with 0.1 M sodium hydroxide filtered through a 0.45
produce a solution containing 1 mg per ml. Desalt the sample µm membrane filter and for 60 minutes with CZE buffer. Apply
as described for the test solution. voltage for 12 hours (20 kV).

Capillary system Between-run rinsing. Rinse the capillary for 10 minutes with
– material. uncoated fused silica, water, for 5 minutes with 0.1 M sodium hydroxide filtered
– size. effective length = about 100 cm, Ø = 50 µm, through a 0.45 µm membrane filter and for 10 minutes with
– temperature. 35º, CZE buffer.
– spectrophotometer set at 214 nm, Migration. Apply a field strength of 143 V/cm (15.4 kV for
– injection. under pressure or vacuum. capillaries of 107 cm total length) for 80 min, using CZE buffer
CZE buffer concentrate (0.1 M sodium chloride, 0.1 M tricine, as the electrolyte in both buffer reservoirs.
0.1 M sodium acetate). Dissolve 0.584 g of sodium chloride, System suitability. In the electropherogram obtained with the
1.792 g of tricine and 0.820 g of anhydrous sodium acetate in reference solution, a pattern of well- separated peaks
water and dilute to 100.0 ml with the same solvent. corresponding to the peaks in the reference electropherogram
1 M putrescine solution. Dissolve 0.882 g of putrescine in 10 of erythropoietin RS is seen, and the largest peak is at least 50
ml of water. Distribute in 0.5 ml aliquots. times greater than the baseline noise. If necessary, adjust the
sample load to give peaks of sufficient height. Identify the
CZE buffer. (0.01 M tricine, 0.01 M sodium chloride, 0.01 M peaks corresponding to isoforms 1 to 8. The peak
sodium acetate, 7 M urea, 2.5 mM putrescine). Dissolve 21.0 corresponding to isoform 1 is detected; the resolution between
g of urea in 25 ml of water by warming in a water-bath at 30º. the peaks corresponding to isoforms 5 and 6 is not less than 1.
Add 5.0 ml of CZE buffer concentrate and 125 ml of 1 M Repeat the separation at least 3 times. The baseline is stable,
putrescine solution. Dilute to 50.0 ml with water. Using dilute showing little drift, and the distribution of peaks is qualitatively
acetic acid, adjust the pH to 5.55 at room temperature and and quantitatively similar to the distribution of peaks in the
filter through a 0.45 µm membrane filter. reference electropherogram of erythropoietin RS. The relative
Set the auto sampler to store the samples at 4° during standard deviation of the migration time of the peak
analysis. corresponding to isoform 2 is less than 2 per cent.

Reference electropherogram of Erythropoietin

5
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2007

Identify the peaks corresponding to isoforms 1 to 8 in the Casting the Stacking gel: 4 per cent
electropherogram obtained with the test solution by Decant the water-saturated butanol and rinse the separating
comparison with the electropherogram obtained with the gel with water. (If the gel has not polymerized and flows out,
reference solution. Calculate the percentage content of each discard and prepare fresh). Place the comb in position above
isoform from the corresponding peak area. The percentages the separating gel. Prepare the stacking mix by adding the
are within the following ranges: reagents in the same sequence as given in the table below:
Isoform Number Content (per cent) Solutions For 5 ml For 10 ml
1 0 – 15 (1 gel) (2 gels)
2 0 – 15 Water 1.8 ml 3.6 ml
3 0 – 20 Acrylamide (30 per cent) 0.6 ml 1.2 ml
4 10 – 35 0.5 M Tris Cl pH. 6.8 2.5 ml 5.0 ml
5 15 – 40 20 per cent SDS 25 µl 50 µl
6 10 – 35
Ammonium persulphate 50 µl 100 µl
7 0 – 20 (10 per cent w/v) (APS)
8 0 – 15 TEMED 5 µl 10 µl
D. Immunoblotting. Determine by electrophoresis (sodium
Mix the above ingredients by swirling gently and pour over
dodecyl sulphate polyacrylamide gel electrophoresis) (SDS-
the separating gel. Avoid bubbles. Keep aside for at least 45
PAGE)(2.4.12).
minutes for polymerization at room temperature.
Use a 1-mm thick spacer with six-well comb, a well capacity of
Preparation of samples and loading
45 l and plate size of 100 x 120 mm
Samples include:
Assemble the gel casting as recommended by the
1. Test sample 1.0 µg
manufacturer.
2. Reference standard RS 1.0 µg
Use the composition of gel given below:
3. Prestained Marker 20 µl
— Separating gel. 12 per cent
Test solution. Take X µl of the test sample and add an equal
— Add the reagents into a clean glass/plastic container in volume of 2X non-reducing sample buffer where,
the same sequence as given in the table below:
X (µl) = 1µg per (concentration of the test sample in mg per ml)
Solutions For 7 ml For 14 ml Reference solution. Dilute a part of 1mg per ml reference
(for 1gel) (for 2 gels) standard RS stock 10 times to get a final concentration of 0.1
Water 2.16 ml 4.32 ml mg per ml with water as follows:
Acrylamide (30per cent) 2.8 ml 5.6 ml 10 µl of 1 mg per ml reference standard RS + 90 ml water.
1.5 M Tris-Cl pH.8.8 1.75 ml 3.4 ml To 10 µl of 0.1 mg per ml reference standard RS solution add
10 ml of 2X non-reducing sample buffer.
20 per cent SDS 35 ml 70 ml
Prestained marker: Take 20 ml of prestained marker,
Ammonium persulphate 70 ml 140 ml reconstituted as recommended by the manufacturer.
(10 per cent w/v)
Loading of samples
TEMED 3 ml 6 ml
Boil the samples for two minutes, centrifuge, bring to room
Mix the above contents by swirling gently (do not mix temperature and load the entire volume on the gel.
vigorously) in the container and pour into the casting unit Sample Samples Amount of protein Total Vol. to be
using a 1-ml pipette to 1 to 1.5 cm from the top edge of the No. loaded/well (µg) loaded. (µl)
plate. After addition of ammonium persulphate and TEMED,
mix the solution and pour quickly (in less than 1minute) or it 1 Test Sample 1 –
will begin to polymerize. Pour 200 – 1000 µl of water-saturated 2 Reference 1 20
butanol on the top of separation gel. Keep aside for at least 45
3 Marker – 20
minutes for polymerization at room temperature.

6
IP 2007 ERYTHROPOIETIN CONCENTRATED SOLUTION

Running the gel 17. Incubate for 1hour with gentle agitation at room
temperature.
1. Fix the gel apparatus in the running unit according to the
manufacturer’s instructions. 18. Wash the membrane with 1 X transfer buffer saline with
change of buffer in-between at room temperature with
2. Chill the 1X running buffer to 4º for at least 1 hour.
gentle agitation. (3 times X 5 minutes)
3. Pour 1X running buffer in the upper as well as lower
19. Discard the transfer buffer saline. Add 25 ml of goat anti-
chambers of the running unit.
rabbit ALP conjugated antibody (secondary antibody)
4. Set parameters on the Power pack as follows: solution. Incubate for 1 hour with gentle shaking at room
— Constant Voltage: 130 V temperature.
— Max Current: 200 mA. 20. Wash the membrane with 1 X transfer buffer saline (3
5. Run until dye front just goes out at the gel bottom (usually times X 5 minutes)
about 1.5 hrs) 21. Develop the color by adding 5 ml of the substrate solution.
(Usually it takes 15 – 20 minutes).
Transfer, blotting and development of membrane:
22. Stop the reaction by pouring off the substrate solution
1. Disassemble the running unit according tohe and washing it with water before the color of the
manufacturer’s instructions. background gets dark.
2. Cut Nitrocellulose Membrane (NCM) having the same 23. Scan the blot while it is wet.
dimensions as of the gel.
24. Air dry the blot and preserve it.
3. Equilibrate the membrane in chilled transfer buffer for 15
– 20 minutes. The molecular mass markers are resolved on the membrane
into discrete bands with a linear relationship between distance
4. Equilibrate the gel in chilled transfer buffer for 15 – 20
migrated and logarithm10 of the molecular mass.
minutes.
5. Soak the nylon pads in transfer buffer and place on the To evaluate the linear relationship between the distance
pad on the cathode. migrated and logarithm10 of the molecular mass, calculate
6. Take 5 sheets of a suitable filter paper, cut to the size of a) log molecular weights corresponding to marker bands
the gel and soak in chilled transfer buffer. Place them on b) migration distance of protein band
the nylon pad placed on cathode plate. c) plot (a) vs (b) and perform linear regression analysis
7. Carefully place the equilibrated gel on the filter papers. The graph should be linear
8. Place the equilibrated membrane onto the gel. The single broad band of the test solution and of reference
9. Roll a clean glass rod dipped in transfer buffer on top of standard RS match in position and intensity.
the membrane to get rid of any air bubbles trapped
E. Peptide mapping (2.3.47).
between the gel and the membrane.
10. Place 5 sheets of a suitable filter paper soaked in chilled Test solution. Dilute the substance under examination in tris-
transfer buffer over the membrane. Once again roll the acetate buffer solution pH 8.5 to a concentration of 1.0 mg
glass rod to remove any air bubbles. per ml. Equilibrate the solution in tris-acetate buffer solution
pH 8.5 using a suitable procedure (such as dialysis against
11. Close the cassette. tris-acetate buffer solution pH 8.5, or membrane filtration using
12. Place the cassette in the running unit. the procedure described under Identification C, but
13. Connect the electrodes with the Power pack and set reconstituting the desalted sample with tris-acetate buffer
following parameters for transfer: solution pH 8.5). Transfer the dialyzed solution to a
— Current: 200 mA polypropylene centrifuge tube. Freshly prepare a solution of
trypsin for peptide mapping at a concentration of 1 mg per ml
— Voltage: 100 V in water, and add 5 ml to 0.25 ml of the dialysed solution. Cap
— Time: 1 hour. the tube and place in a water-bath at 37º for 18 hours. Remove
14. After the transfer is over, disassemble the cassette. the sample from the water-bath and stop the reaction
15. Transfer the membrane to the blocking solution. Incubate immediately by freezing.
the membrane in the blocking buffer for 1 hour with gentle Reference solution. Dissolve the contents of a vial of
shaking at room temperature. erythropoietin RS in 0.25 ml of water. Prepare as for the test
16. Discard the blocking buffer. Add 25 ml of rabbit anti solution, ensuring that all procedures are carried out
r-Hu EPO antibody (Primary antibody) solution. simultaneously, and under identical conditions.

7
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2007

Chromatographic system cartridge using the protocol provided by the manufacturer.


– a stainless steel column 25 cm x 4.6 mm, packed with Run 15 sequencing cycles, using the reaction conditions for
butylsilyl silica gel (5-10 µm), proline when running the second and third cycles.
– mobile phase: A. 0.06 per cent v/v solution of
Identify the phenylthiohydantoin (PTH)-amino acids released
trifluoroacetic acid,
at each sequencing cycle by reverse-phase liquid
B. to 100 ml of water add 0.6 ml of
chromatography. The procedure may be carried out using the
trifluoroacetic acid and dilute to 1000 ml with
column and reagents recommended by the manufacturer of
acetonitrile,
the sequencing equipment for the separation of PTH-amino-
– a linear gradient programme using the conditions given
acids.
below,
– spectrophotometer set at 214 nm, The separation procedure is calibrated using:
– a 50 µl loop injector. — the mixture of PTH-amino acids provided by the
Time Flow rate Mobile phase A Mobile phase B manufacturer of the sequencer, with the gradient
(min) (ml/min) (per cent v/v) (per cent v/v) conditions adjusted as indicated to achieve optimum
0 – 10 0.75 100 0 resolution of all amino acids,
10 – 125 0.75 100 →39 0 → 61 — a sample obtained from a blank sequencing cycle obtained
125 – 135 1.25 39 → 17 61→83 as recommended by the equipment manufacturer.
135 – 145 1.25 17 →0 83→ 100 If a sample does not comply with the limits set for the n-1 and
145 – 150 1.25 100 0 n-2 sequences repeat the analysis.

Equilibrate at initial conditions for at least 15 minutes. Carry Tests


out a blank run using the above-mentioned gradient.
The chromatogram obtained with each solution is qualitatively Protein. 80 per cent to 120 per cent of the stated amount.
similar to the reference chromatogram of erythropoietin RS Test solution. Dilute the substance under examination in a
digest. 0.4 per cent w/v solution of ammonium hydrogen carbonate
The profile of the chromatogram obtained with the test or the appropriate blank solution.
solution corresponds to that of the chromatogram obtained When examined in the range 250 nm to 400 nm (2.4.7), the
with the reference solution. solution shows an absorption maximum between 276 nm and
F. Determine by N-terminal sequence analysis 282 nm. Calculate the content of erythropoietin taking the
specific absorbance to be 7.43.
The first 15 amino acids are: Alanine - Proline - Proline -
Arginine - Leucine - Isoleucine - (no recovered peak) - Aspartic Dimers and related substances of higher molecular mass
acid - Serine - Arginine - Valine - Leucine - Glutamic acid -
Arginine - Tyrosine. A. Determine by size-exclusion chromatography (2.4.16).

Perform the Edman degradation using an automated solid- Test solution. Dilute the substance under examination in the
phase sequencer, operated in accordance with the mobile phase to obtain a concentration of 0.2 mg per ml.
manufacturer’s instructions. Reference solution. To 0.02 ml of the test solution add 0.98 ml
Desalt the equivalent of 50 µg of erythropoietin. For example, of the mobile phase (2 per cent).
dilute a volume of the substance under examination containing Chromatographic system
50 µg of the active substance in 1 ml of a 0.1 per cent v/v – a stainless steel column 6 cm x 7.5 mm, packed with
solution of trifluoroacetic acid. Pre-wash a C18 reverse-phase hydrophilic silica gel, of a grade suitable for fractionation
sample preparation cartridge according to the instructions of globular proteins in the molecular mass range of 20
supplied by the manufacturer and equilibrate the cartridge in 000 to 200 000,
a 0.1 per cent v/v solution of trifluoroacetic acid. Apply the – mobile phase: Dissolve 1.15 g of anhydrous disodium
sample to the cartridge, and wash successively with a 0.1 per hydrogen phosphate, 0.2 g of potassium dihydrogen
cent v/v solution of trifluoroacetic acid containing 0 per cent, phosphate and 23.4 g of sodium chloride in 1 litre of
10 per cent and 50 per cent v/v of acetonitrile according to water (1.5 mM potassium dihydrogen phosphate, 8.1
the manufacturer’s instructions. Lyophilise the 50 per cent v/ mM disodium hydrogen phosphate, 0.4M sodium
v acetonitrile eluate. chloride, pH 7.4); adjust the pH to 7.4 if necessary,
Redissolve the desalted sample in 50 µl of a 0.1 per cent v/v – flow rate. 0.5 ml per minute,
solution of trifluoroacetic acid and couple to a sequencing – spectrophotometer at 214 nm,

8
IP 2007 ERYTHROPOIETIN CONCENTRATED SOLUTION

– a 100 µl loop injector. Place the comb in position above the separating gel.
Inject the test solution and the reference solution. Continue Prepare the stacking mix by adding the reagents in the same
the chromatography for 1 hour. The area of the principal peak sequence as given in the table below:
in the chromatogram obtained with the reference solution is
Solutions For 5 ml For 10 ml
1.5 to 2.5 per cent of the area of the principal peak in the
(1 gel) (2 gels)
chromatogram obtained with the test solution. The total area
of any peaks eluted before the principal peak is not more than Water 1.8 ml 3.6 ml
the area of the principal peak in the chromatogram obtained
Acrylamide (30 per cent) 0.6 ml 1.2 ml
with the reference solution (2 per cent).
0.5 M Tris Cl pH. 6.8 2.5 ml 5.0 ml
B. Determine by electrophoresis (sodium dodecyl sulphate
polyacrylamide gel electrophoresis (SDS-PAGE) (2.4.12). 20 per cent SDS 25 µl 50 µl
Casting the Gels Ammonium persulphate 50 µl 100 µl
(10 per cent w/v) (APS)
— Use a 1-mm thick spacer with eight-well comb.
— The well capacity is 65 µl. TEMED 5 µl 10 µl
— Assemble the gel - casting unit according to the
Mix the above contents by swirling gently and pour over the
manufacturer’s instructions.
separating gel. Avoid bubbles.
— Use the assembly of plate size, 160 x 160 mm.
Keep aside for at least 45 minutes for polymerization at room
a) Casting the separating gel: 12 per cent temperature.
Add the reagents into a clean glass/plastic container in the Preparation of samples
same sequence as given in the table below:
Samples include:
Solutions For 20 ml For 40 ml 1. Test sample 10 µg
(1 gel) (2 gels)
2. Reference standard RS 10 µg
Water 6.17 ml 12.34 ml 3. Marker for non-reducing gel 20 µl
Acrylamide (30 per cent) 8.0 ml 16.0 ml Test solution. To a volume containing 10 µg of protein add an
1.5 M Tris Cl pH. 8.8 5.0 ml 10.0 ml equal volume of 2X non-reducing sample buffer.
20 per cent SDS 100 µl 200 µl Reference solution. Take 10 µl of the reference standard RS
from 1mg per ml stock in a micro-centrifuge tube and add 10 µl
Ammonium persulphate 200 µl 400 µl
of 2X non-reducing sample buffer.
(10 per cent w/v) (APS)
Molecular weight marker. Take 20 µl of low molecular weight
TEMED 8 µl 16 µl
markers for SDS-PAGE, which is reconstituted according to
Mix the above ingredients by swirling gently and pou the manufacturer’s instruction:
Mix the above contents by swirling gently (do not mix Sample loading
vigorously) in the container and pour into the casting unit Keep all the samples in a boiling water-bath for 2 minutes.
using a 1-ml pipette till 1 - 1.5 cm from the top edge of the plate. Centrifuge, bring to room temperature and load the entire
After addition of ammonium persulphate and TEMED, the volume on to the gel.
solution should be mixed and poured quickly (less than
Sample Samples Amount of protein Total Vol. to be
1minute) or it will begin to polymerize.
No. loaded/well (µg) loaded. (µl)
Pour 200 – 1000 µl of water-saturated butanol on the top of
the separation gel. 1 Test Sample 1 –
Keep aside for at least 45 minutes for polymerization at room 2 Reference 1 20
temperature (RT). 3 Marker – 20
b) Casting the Stacking gel: 4 per cent
Running the gel
Decant the water-saturated butanol and rinse the separating
gel with water. (If the gel has not polymerized and flows out, Fix the gel apparatus in the running unit as given in the
discard and prepare fresh) instruction manual of the manufacturer.

9
ERYTHROPOIETIN CONCENTRATED SOLUTION IP 2007

Chill the 1X running buffer to 4° for at least 1 hour. Reference solution (a). Dissolve a suitable amount of N-
acetylneuraminic acid in water to produce a solution
Pour 1X running buffer in the upper as well as lower chambers
containing 0.1 mg per ml.
of the running unit.
Reference solution (b). To 0.8 ml of reference solution (a) add
Set parameters on the Power pack as follows:
0.2 ml of water.
Constant Voltage: 130 V
Reference solution (c). To 0.6 ml of reference solution (a) add
Max Current: 200 mA.
0.4 ml of water.
Run until the dye front reaches the bottom of the gel (usually
Reference solution (d). To 0.4 ml of reference solution (a) add
about 5.0 hrs. The dye front should have run at least 80 per
0.6 ml of water.
cent of the gel).
Reference solution (e). To 0.2 ml of reference solution (a) add
Staining of Gel
0.8 ml of water.
Stop the run. Disassemble the casting unit and transfer the
Reference solution (f). Use water
gel carefully into a staining tray. Do not touch the gel with
naked hands; wear gloves while handling the gel. Carry out the test in triplicate. Transfer 100 ml each of the test
and reference solutions to 10-ml glass test tubes. To each
Detect proteins in the gel by silver staining.
tube add 1.0 ml of resorcinol reagent. Stopper the tubes and
Scanning incubate at 100º for 30 minutes. Cool on ice. To each tube, add
Scan and save the image of the stained gel. 2.0 ml of a mixture of 12 volumes of butanol and 48 volumes of
butyl acetate. Mix vigorously, and allow the 2 phases to
Gel drying separate. Ensuring that the upper phase is completely clear,
The stained gel is placed in between two cellophane sheets remove the upper phase, taking care to exclude completely
and clamped with the gel dryer frames (taking care that no air any of the lower phases. Measure the absorbance. of all
bubbles are present in between the two cellophane sheets). samples at 580 nm (2.4.7).
The set up is placed in the gel dryer apparatus and left at least Using the calibration curve generated by the reference
2 hrs for drying. solutions, determine the content of sialic acids in each of the
two test solutions and calculate the mean. Calculate the
The dye front is run for at least 80 per cent of the total gel
number of moles of sialic acids per mole of erythropoietin
length. Molecular weight markers are resolved on the gel into
assuming that the relative molecular mass of erythropoietin is
discrete bands, with a linear relationship between distance
30 600 and that the relative molecular mass of N-
migrated and logarithm10 of the molecular mass.
acetylneuraminic acid is 309.
To evaluate the linear relationship between distance migrated
System suitability. The individual replicates agree to within
and logarithm10 of the molecular mass, calculate
±10 per cent of each other; the value obtained from reference
a) log molecular weights corresponding to marker bands solution (a) is between 1.5 and 2.5 times that obtained with
b) migration distance of protein bands test solution (a).
c) plot (a) vs (b) and perform linear regression analysis. Bacterial endotoxins (2.2.3). Not more than 20 Endotoxin Units
in the volume that contains 100 000 IU of erythropoietin.
The graph should be linear.
Assay. The activity of the preparation is compared with that
The single diffuse band of the test solution and of the reference of erythropoietin RS and expressed in International Units
solution match in position and intensity. (IU).
Sialic acids. Not less than 10 mol of Sialic acids (calculated as The estimated potency is not less than 80 per cent and not
N-acetylneuraminic acid) per mole of erythropoietin, more than 125 per cent of the stated potency. The fiducial
determined in the following manner. limits of error of the estimated potency (P = 0.95) are not less
Test solution (a). Dilute the preparation under examination in than 64 per cent and not more than 156 per cent of the stated
the mobile phase used in the test for dimers and related potency.
substances of higher molecular mass to obtain a concentration Determine by Method A or Method B.
of 0.3 mg/ml.
A. In polycythaemic mice
Test solution (b). To 0.5 ml of test solution (a) add 0.5 ml of the
mobile phase used in the test for dimers and related substances The activity of the preparation is estimated by examining,
of higher molecular mass. under given conditions, its effect in stimulating the

10
IP 2007 ERYTHROPOIETIN CONCENTRATED SOLUTION

incorporation of 59Fe into circulating red blood cells of mice radiolabelled [59Fe] ferric chloride solution. The order of the
made polycythaemic by exposure to reduced atmospheric injections must be the same as that of the erythropoietin
pressure. injections, and the time interval between administration of the
erythropoietin and the radiolabelled ferric chloride solution
The following schedule, using treatment in a hypobaric
must be the same for each animal. After a further 48 h,
chamber, has been found to be suitable.
anaesthetise each animal by injection of a suitable anaesthetic,
Induce polycythaemia in female mice of the same strain, record body weights and withdraw blood samples (0.65 ml)
weighing 16 g to 18 g. Place the mice in a hypoxic chamber and into haematocrit capillaries from the bifurcation of the aorta.
reduce the pressure to 0.6 atmospheres. After 3 days at 0.6 After determining the packed cell volume for each sample,
atmospheres, further reduce the pressure to 0.4-0.5 measure the radioactivity.
atmospheres and maintain the animals at this pressure for a
Calculate the response (percentage of iron-59 in total
further 11 days (the partial vacuum is interrupted daily for a
circulating blood) for each mouse using the expression:
maximum of 1 h at about 11:00 a.m., in order to clean the cages
and feed the animals). At the end of the specified period,
A s × M × 7.5
return the mice to normal atmospheric conditions. Randomly
distribute the mice into cages, each containing 6 animals, and A t × Vs
mark them.
Where, As = radioactivity in the sample,
Test solution (a). Dilute the substance under examination in At = total radioactivity injected,
phosphate-albumin buffered saline pH 7.2 to obtain a
7.5 = total blood volume as per cent body weight,
concentration of 0.2 IU per ml.
M = body weight, in grams,
Test solution (b). Mix equal volumes of test solution (a) and Vs = sample volume.
phosphate-albumin buffered saline pH 7.2.
Calculate the potency by the usual statistical methods for a
Test solution (c). Mix equal volumes of test solution (b) and parallel line assay. Eliminate from the calculation any animal
phosphate-albumin buffered saline pH 7.2. where the packed cell volume is less than 54 per cent, or where
Reference solution (a). Dissolve erythropoietin RS in the body weight is more than 24 g.
phosphate-albumin buffered saline pH 7.2 to obtain a
B. In normocythaemic mice
concentration of 0.2 IU per ml.
The assay is based on the measurement of stimulation of
Reference solution (b).Mix equal volumes of reference
reticulocyte production in normocythaemic mice.
solution (a) and phosphate-albumin buffered saline pH 7.2.
Test solution (a). Dilute the substance under examination in
Reference solution (c). Mix equal volumes of reference
phosphate-albumin buffered saline pH 7.2 to obtain a
solution (b) and phosphate-albumin buffered saline pH 7.2.
concentration of 80 IU per ml.
Radiolabelled ferric [59Fe] chloride solution, concentrated.
Test solution (b). Mix equal volumes of test solution (a) and
Use a commercially available solution of [59Fe] ferric chloride
phosphate-albumin buffered saline pH 7.2.
(approximate specific activity: 100-1000 MBq per mg of Fe).
Radiolabelled [59Fe] ferric chloride solution. Dilute the Test solution (c). Mix equal volumes of test solution (b) and
concentrated radiolabelled [59Fe] ferric chloride solution in phosphate-albumin buffered saline pH 7.2.
sodium citrate buffer solution pH 7.8 to obtain a solution Reference solution (a). Dissolve erythropoietin RS in
with an activity of 3.7 × 104 Bq per ml. phosphate-albumin buffered saline pH 7.2 to produce a
The concentrations of the test solutions and reference solution containing 80 IU per ml.
solutions may need to be modified, based on the response Reference solution (b). Mix equal volumes of reference
range of the animals used. solution (a) and phosphate-albumin buffered saline pH 7.2.
3 days after returning the animals to atmospheric pressure, Reference solution (c). Mix equal volumes of reference
inject each animal subcutaneously with 0.2 ml of one of the solution (b) and phosphate-albumin buffered saline pH 7.2.
solutions. The 6 animals in each cage must each receive one
The exact concentrations of the test solutions and reference
of the 6 different treatments (3 test solutions and 3 reference
solutions may need to be modified, based on the response
solutions), and the order of injection must be separately
range of the animals used.
randomised for each cage. A minimum of 8 cages is
recommended. 2 days after injection of the test or reference At the beginning of the assay procedure, randomly distribute
solution, inject each animal intraperitoneally with 0.2 ml of mice of a suitable age and strain (8-week old B6D2F1 mice are

11
FILGRASTIM CONCENTRATED SOLUTION IP 2007

suitable. Other strains like Swiss Albino, Balb/C of suitable Filgrastim Concentrated Solution contains not less than 1.5
age can also be used) into 6 cages. A minimum of 8 mice per mg of protein per ml, and not less than 1.0 x 108 IU of filgrastim
cage is recommended. Inject each animal subcutaneously with per mg of protein.
0.5 ml of the appropriate treatment (one solution per cage) and
Prior to release, the following tests are carried out on each
put the animal in a new cage. Combine the mice in such a way
batch of the final bulk product, unless the regulatory authority
that each cage housing the treated mice contains one mouse
has granted exemption.
out of the 6 different treatments (3 test solutions and 3 reference
solutions, 6 mice per cage). 4 days after the injections, collect Description. A clear, colourless to slightly yellowish liquid.
blood samples from the animals and determine the number of
reticulocytes using the following procedure. Identification
The volume of blood, dilution procedure and fluorescent A. It shows the biological activity as decribed under Assay.
reagent may need to be modified to ensure maximum B. Determine by capillary electrophoresis (capillary isoelectric
development and stability of fluorescence. focusing) (2.4.32).
Colorant solution, concentrated. Use a solution of thiazole In the test for impurities with charges different from that of
orange suitable for the determination of reticulocytes. Prepare filgrastim the principal band in the electropherogram obtained
at a concentration twice that necessary for the analysis. with the test solution is similar in position to the principal
Proceed with the following dilution steps. Dilute whole blood band in the electropherogram obtained with the reference
500-fold in the buffer used to prepare the colorant solution. solution.
Dilute this solution 2-fold in the concentrated colorant C. In the test for impurities of molecular masses higher than
solution. After staining for 3-10 min, determine the reticulocyte that of filgrastim, the retention time, of the principal peak
count microfluorometrically in a flow cytometer. The percentage obtained with the test solution is similar to that of the principal
of reticulocytes is determined using a biparametric histogram: peak obtained with the reference solution.
number of cells/red fluorescence (620 nm). D. In the test for impurities with molecular masses differing
Calculate the potency by the usual statistical methods for a from that of rG-CSF under both reducing and non-reducing
parallel line assay. conditions, the principal band in the electropherogram obtained
with test solution (a) is similar in position to the principle
Storage. Store in an airtight container at a temperature below
band in the electropherogram obtained with reference solution
-20º. Avoid repeated freezing and thawing.
(a).
Labelling. The label states (1) the erythropoietin content in E. Determine by peptide mapping (2.3.47).
mg per ml; (2) the activity in International Units per ml; (3) the
name and the concentration of any other excipients. Test solution. Introduce 50 µl of a 0.05 M sodium phosphate
buffer pH 8.0 into a polypropylene tube. Add a volume of the
substance under examination corresponding to 25 µg of
protein, and 25 µl of a 0.1 mg per ml solution of Glu-C2 protease;
dilute to 1 ml with water, stopper the tube and incubate at
Filgrastim Concentrated Solution about 37º for 18 hours. Add 125 µl of a 7.64 per cent w/v
solution of guanidine hydrochloride and mix well. Add 10 µl
Granulocyte Colony Stimulating Factor Solution
of a 1.542 per cent w/v solution of dithiothreitol and mix well.
Place the capped tube in boiling water for 1 minute. Allow to
MTPLGPASSL PQSFLLKCLE QVRKIQGDGA ALQEKLCATY KLCHPEELVL LGHSLGIPWA cool to room temperature.
PLSSCPSQAL QLAGCLSQLH SGLFLYQGLL QALEGISPEL GPTLDTLQLD VADFATTIWQ
Reference solution. Prepare at the same time and in the same
QMEELGMAPA LQPTQGAMPA FASAFQRRAG GVLVASHLQS FLEVSYRVLR HLAQP
manner as for the test solution but use G-CSF RS instead of
the test preparation under examination.
C845H1343N223O243S9 Mol. Wt. 18799
Chromatographic system
Filgrastim Concentrated Solution is a solution of a protein – a stainless steel column 10 cm x 2 mm, packed with
having the structure of the granulocyte colony-stimulating octadecylsilyl silica gel (5 µm) with a pore size of 20 nm,
factor (G-CSF) produced and secreted by various human blood – mobile phase: A. Dilute 0.5 ml of trifluoroacetic acid to
cell types. The protein stimulates the differentiation and 950 ml with water add 50 ml of acetonitrile and mix,
proliferation of leucocyte stem cells into mature granulocytes. B. Dilute 0.5 ml of trifluoroacetic acid to 50 ml with
It is produced by a method based on rDNA technology, using water; add 950 ml of acetonitrile,
bacteria as host cells. – flow rate. 0.2 ml per minute,

12
IP 2007 FILGRASTIM CONCENTRATED SOLUTION

– A linear gradient programme using the conditions given sulphate polyacrylamide gel electrophoresis) (2.4.12) under
below, both reducing and non-reducing conditions.
– spectrophotometer set at 215 nm,
Gel dimensions. 1.0 mm thick. 160 X 160 mm
– a 10 µl loop injector.
Resolving gel. 13 per cent Acrylamide
Time Mobile phase A Mobile phase B Sample buffer A.
(min) (per cent v/v) (per cent v/v) Sample buffer B (reducing conditions).
0 – 8.0 3–6 97– 94 Test solution (a). Dilute the preparation under examination
8 – 25 6 – 34 94 – 66 with water to produce a solution containing 0.1 mg per ml. To
25 – 40 34 – 90 66 – 100 50 volumes of this solution, add 13 volumes of concentrated
SDS-PAGE sample buffer.
40 – 45 90 10
45 – 46 90 – 3 10– 97 Test solution (b). Prepare in the same manner as test solution
(a) but using concentrated SDS-PAGE sample buffer for
46 – 65 3 97
reducing conditions.
Equilibrate the column at the initial conditions for at least 30
Reference solution (a). Dilute the preparation under
minutes.
examination with water to obtain a concentration of 0.1 mg
Inject the test solution and the reference solution. per ml. To 50 volumes of this solution, add 13 volumes of
The chromatograms obtained with the reference solution and concentrated SDS-PAGE sample buffer.
the test solutions are qualitatively similar. Reference solution (b.) Prepare as for reference solution (a),
The profile of chromatogram obtained with the reference but using concentrated SDS-PAGE sample buffer for reducing
solution and the test solution corresponds to that of the conditions.
chromatogram obtained with the reference solution. Reference solution (c). Use a solution of molecular mass
F. Determine by N-Terminal Sequencing. marker suitable for calibrating SDS-PAGE gels in the range of
14 400 to 94 000. Dissolve in sample buffer of sample buffer
Perform the Edman degradation using an automated solid- (reducing conditions), as appropriate.
phase sequencer, operated in accordance with the
manufacturer’s instructions. Reference Protein Sample SDS-PAGE
solution amount Volume Sample
Load about 1ml of the test preparation to a sequencing cartridge (µg) (µl) Buffer
using the protocol provided by the manufacturer. Run 16
sequencing cycles, noting, if appropriate the presence of (a) 100 20 20
praline at positions 3, 6 and 111. (b) 50 20 20
Identify PTH-amino acids released at each sequencing cycle (c) 20 20 20
by RPHPLC. The procedure may be carried out using the (d) 10 20 20
column and reagents recommenced by the manufacturer of (e) 5 20 20
the sequencing equipment for the separation of PTH-amino
acids. (f) 2 20 20
(g) 1 20 20
The separation procedure is calibrated using:
a) the mixture of PTH-amino acids provided by the
Sample treatment: boil for 5 minutes.
manufacturer of the sequencer, with the gradient
conditions adjusted as indicated to achieve optimum Apply 20 µl of each reduced and non-reduced solutions to
resolution of all amino acids; separate gels.
b) a sample obtained from a blank sequencing cycle obtained Detection: Silver staining as described below.
as recommended by the equipment manufacturer.
Immerse the gel for 30 minutes in a mixture of 10 volumes of
The first 16 amino acids should match the sequence given in acetic acid, 40 volumes of water and 50 volumes of methanol.
the beginning starting with Met. Transfer the gel to 5 per cent methanol and shake for 5 minutes.

Tests Repeat this washing step thrice. Replace the 5 per cent v/v
solution of methanol with 0.2 g per litre sodium thiosulphate.
Impurities with molecular masses differing from that of Wash the gel thrice in water for 30 seconds each. Transfer the
Filgrastim. Determine by electrophoresis (sodium dodecyl gel to a 0.2 per cent w/v solution of silver nitrate.

13
FILGRASTIM CONCENTRATED SOLUTION IP 2007

This solution is prepared immediately before use. Place the Test solution. Dilute the preparation under examination with
gel on shaker for 25 minutes. Wash the gel for 1 minute. Repeat mobile phase A to obtain a concentration of 0.3 mg per ml.
this washing step thrice. Transfer the gel into a mixture
Reference solution (a). Dilute G-CSFRS with the same mobile
containing 30 g per litre solution of sodium carbonate, 0.05
phase to obtain a concentration of 0.3 mg per ml.
per cent v/v solution of formaldehyde and 0.2 g per litre
solution of sodium thiosulphate in water. Protein bands Reference solution (b). To 570 µl of reference solution (a),
become visible during this step. Keep the gel in the solution add 6.8 µl of a 0.45 per cent v/v solution of hydrogen peroxide;
until sufficiently stained and then stop the staining by soaking mix and incubate at 25° for 1hour, then add 2.5 mg of
the gel in a 14 g per litre solution of disodium edetate. methionine RS.
The test is not valid unless the proteins of the molecular weight Chromatographic system
marker are distributed along 80 per cent of the gel and over the – a stainless steel column 15 cm x 4.6 mm, packed with
required separation range (the range covering the product octadecylsilyl silica gel (5 µm) with a pore size of 20 nm
and its dimer or the product and its related impurities). and a guard column 7.5 cm x 7.5mm packed with
In the electropherogram obtained with test solution (a) no octadecylsilyl silica gel,
band is more intense that the principal band in the – column temperature. 65º,
electropherogram obtained with reference solution (f). – mobile phase: A. dilute 1 ml of trifluoroacetic acid to
500 ml with water and add 499 ml of acetonitrile,
Impurities with charges differing from that of Filgrastim. B. dilute 1 ml of trifluoroacetic acid to 950 ml with
Determine by capillary electrophoresis (capillary isoelectric acetonitrile and add 49 ml of water,
focusing) (2.4.32). – flow rate. 1 ml per minute,
Test solution. Dilute the preparation under examination to – a linear gradient programme using the conditions given
produce a solution containing 0.3 mg per ml. below,
– spectrophotometer set at 215 nm,
Reference solution (a). A solution of filgrastim RS containing – a 50 µl loop injector.
0.3 mg per ml.
Time Mobile phase A Mobile phase B
Reference solution (b). A solution of filgrastim RS containing (min) (per cent v/v) (per cent v/v)
0.06 mg per ml.
0–4 92 8
Reference solution (c). Use an isoelectric point (pI) calibration 4 – 19 92 → 72 8 → 28
solution, in the pI range of 2.5-6.5, prepared according to
manufacturer’s instructions. 19 – 19.1 72 → 0 28 → 100
19.1 – 21 0 100
Focusing:
– pH gradient. 4.5 - 8.0, 21 – 21.1 0 → 92 100 → 8
– catholyte. 1 M sodium hydroxide, 21.1 – 25 92 8
– anolyte: 0.04 M glutamic acid in a 0.0025 per cent v/v Inject the test solution and reference solutions (a) and (b).
solution of phosphoric acid,
– Application 20 µl. Relative retention with reference to filgrastim (retention
time=about 12 minutes.): oxidized filgrastim 2= about 0.95
Detection. Proceed as described in Isoelecric Focusing
(2.4.33). The profile of the chromatogram obtained with reference
solution (b) is similar to that of the chromatogram of oxidized
Detect the product and its dimer or the product and its related
filgrastim supplied with filgrastim RS. Two peaks
impurities.
corresponding respectively to oxidized filgrastim 2 elute before
In the electropherogram obtained with reference solution (c), the principal peak, the second peak not being completely
relevant isoelectric point markers are distributed along the separated form the principal peak.
entire length of the gel.
In the chromatogram obtained with the test solution, the area
In the electropherogram obtained with reference solution (a), of any peak other than the principal peak is not greater than
the pI of the principal band is 5.7-6.3. 2.0 per cent of the total area of all the peaks. The sum of the
No band is more intense that the principal band in the areas of any peaks other than the principal peak is not greater
electropherogram obtained with reference solution (b). than 3.5 per cent of the total area of all of the peaks.
Related Proteins. Determine by liquid chromatography Dimers and Related Substance of Higher Molecular Mass.
(2.4.14). Determine by size exclusion chromatography (2.4.14).

14
IP 2007 INTERFERON ALFA-2 CONCENTRATED SOLUTION

Solution A. Dissolve 4.1 g of sodium acetate in 400 ml of The formazan is then measured spectrophotometrically. The
water, adjust to pH 4.0 with acetic acid and dilute to 500 ml potency of the test preparation is determined by comparison
with water. of the dilutions of the test preparation with the dilutions of
Test solution. Dilute the preparation under examination with the appropriate International Standard of rG-CSF or with a
solution A to obtain a concentration of 0.4 mg per ml. reference preparation calibrated in International Units, which
yield the same response (50 per cent maximal stimulation).
Reference solution. Dilute filgrastim RS with solution A to
obtain a concentration of 0.4 mg per ml. The International Unit is the activity contained in a stated
amount of the appropriate International Standard. The
Resolution solution. Mix a sample of the reference solution
equivalence in International Units of the International standard
for about 30 seconds using a vortex mixer.
is stated by the World Health Organization.
Chromatographic system
Add 50 µl of the dilution medium to all wells of a 96 –well
– a stainless steel column 30 cm x 7.8 mm, packed with
microtitre plate. Add an additional 50 µl of this solution to the
hydrophilic silica gel
wells designed for blanks. Add 50 µl of each solution to be
– column temperature. 30°,
tested in triplicate (test preparation and reference preparation
– mobile phase: dissolve 7.9 g of ammonium hydrogen
at a concentration of about 800 IU per ml, plus a series of 10
carbonate in 1000 ml of water and adjust to pH 7.0 with
twofold dilutions to obtain a standard curve). Prepare a
phosphoric acid; dilute to 2000 ml with water,
suspension of NFS-60 cells containing 7x105 cells per ml.
– flow rate. 0.5 ml per minute,
immediately before use, add 2-mercaptoethanol to a final
– spectrophotometer set at 215 nm,
concentration of 0.1 mM, and add 50 µl of the prepared cell
– a 200 µl loop injector.
suspension to each well, maintaining the cells in a uniform
Relative retention times with reference to filgrastim monomer suspension during addition.
(retention time=about 19 minutes): aggregates = about 0.60; Incubate the plate at 36.0º- 38.0º for a minimum of 24 hours in
filgrastim oligomer 1= about 0.75; filgrastim oligomer 2= about a humidified incubator using 6 ± 1 per cent CO2. Add 20 ml of
0.80; filgrastim dimer=about 0.85. a 5.0 g per litre sterile solution of tetrazolium bromide to each
Inject the resolution solution. The retention time of filgrastim well and re-incubate 4 hours. Estimate the quantity of formazan
monomer is 17 minutes to 20 minutes. The resolution between produced using a microtitre well plate reader at 490 nm.
the peaks due to filgrastim dimer and filgrastim monomer is Analyze the data by fitting a sigmoidal dose-response curve
not less than 4.0. to the data obtained and by using a suitable stastical method,
Calculate the content of dimer, oligomers and aggregates. The for example the 4-parameter or parallel line models.
sum of the peaks with retention times less that that of the The estimated potency is not less than 80 per cent and not
principal peak is not more than 2.5 per cent. more than 125 per cent of the stated potency. The confidence
Bacterial Endotoxins. Not more than 2 Endotoxin Unit per mg limits (P=0.95) of the estimated potency are not less than 74
of protein. per cent and not more than 136 per cent of the stated potency.
Assay. A. Protein - Determine by liquid chromatography (2.4.14) Storage. Store protected from light in a refrigerator (2º to 8º).
as described under the test for Related proteins. Labeling. The label states the content, in mg of protein per ml;
Inject the test solution and reference solution (a). the potency, in IU per mg of protein.
Calculate the content of filgrastim.
B. Potency- Determination of the biological activity of rG-
CSF concentrated solution is based on the stimulation of
Interferon Alfa-2 Concentrated
NFS60 cells (murine myeloblastic cell line) by rG-CSF. Solution
The following method uses the conversion of tetrazolium CDLPQTHSLG SRRTLMLLAQ MRX1ISLFSCL KDRHDFGFPQ
bromide (MTS) as a staining method. Alternative methods of
quantifying cell proliferation, such as measurement of EEFGNQFQKA ETIPVLHEMI QQIFNLFSTK DSSAAWDETL
intracellular ATP by luciferase bioluminescence have also been LDKFYTELYQ QLNDLEACVI QGVGVTETPL MKEDSILAVR
found suitable, and may be used as the assay readout, subject
to appropriate validation. KYFQRITLYL KEKKYSPCAW EVVRAEIMRS FSLTSNLQES

NFS-60 cells are incubated with varying dilution of test and LRSKE
reference preparations of rG-CSF. They are then incubated
with a solution of MTT. This cytochemical stain is converted alfa-2a: C860H1353N227O255S9 Mol. Wt.19,241
by cellular dehydrogenases to a purple formazan product. alfa-2b: C860H1353N229O255S9 Mol. Wt.19,269

15
INTERFERON ALFA-2 CONCENTRATED SOLUTION IP 2007

Interferon alfa-2 concentrated solution is a solution of an r- Horizontal Electrophoresis


DNA derived therapeutic protein which exhibits non-specific
antiviral activity, at least in homologous cells. Interferon alfa- Select and use a suitable horizontal isoelectric focusing
2 concentrated solution also exerts antiproliferative and apparatus with facility for connecting a circulating bath chiller
immunomodulator activity. Two different types of alfa-2 capable of maintaining 10°. Select gels for isoelectric focusing
interferon, varying in the amino acid residue at position 23, with a pH gradient from 3.5 to 9.5
are found and are named as alfa-2a and alfa-2b. Use phosphoric acid as anode solution (98 g per litre
phosphoric acid) and 1 M sodium hydroxide as the cathode
Designation Residue at position 23 (X1) solution. Using filter paper apply 15 µl of the test solution and
alfa-2a Lys the reference solution to the gel close to the cathode.
alfa-2b Arg Start the isoelectric focusing at 1500 V and 50 mA. Turn off
the power after 30 minutes. Remove the application filters and
This monograph applies to interferon alfa-2a and 2b reconnect the power supply for 1 hour. Keep the power
concentrated solutions. constant during the focusing process.
Interferon alfa-2 concentrated solution contains not less than Immerse the gel in a solution containing 115 g per litre of
1.4 × 108 IU per mg of protein and not less than 0.5 mg of trichloroacetic acid and 34.5 g per litre of sulphosalicylic
interferon alfa-2 per ml. acid in water and agitate the container gently for 60 minutes.
Prepare a mixture of 32 volumes of glacial acetic acid, 100
Production
volumes of ethanol and 268 volumes of water. Transfer the
Interferon alfa-2 concentrated solution is produced by a gel to the mixture and soak for 5 minutes.
method based on recombinant DNA (rDNA) technology. It is Immerse the gel for 10 minutes in a staining solution prewarmed
produced under controlled conditions designed to ensure to 60°.The staining solution is prepared by adding 1.2 g per
sterility of the product. litre of acid blue 83 to the mixture of glacial acetic acid,
Interferon alfa-2 concentrated solution complies with the ethanol and water.
following additional requirements. Wash the gel several times to destain with the mixture of glacial
acetic acid, ethanol and water and keep the gel in this mixture
Host-cell-derived proteins
for about 12-24 hours until the background is clear.
The limit is approved by WHO guidelines. Add glycerol 10 per cent v/v to the mixture of glacial acetic
Host-cell- or vector-derived DNA acid, ethanol and water. Soak the gel for 1 hour in the solution.
The limit is approved by WHO guidelines. The principal bands of the electropherogram obtained with
the test solution correspond in position to the principal bands
Description. A clear,colourless or slightly yellowish liquid. of the electropherogram obtained with the reference solution.
Plot the migration distances of the isoelectric point markers
Identification versus their isoelectric points and determine the isoelectric
points of the principal components of the test solution and
A. It shows the expected biological activity as described under
the reference solution. They do not differ by more than 0.2 pI
Assay for potency.
unit. The test is not valid unless the isoelectric point markers
B. Determine by isoelectric focusing. are distributed along the entire length of the gel and the
isoelectric points of the principal bands in the electropherogram
Test solution. Dilute the preparation under examination with
obtained with the reference solution are between 5.8 and 6.3.
water to obtain a solution containing 0.5 mg protein per ml.
C. Examine the electropherograms obtained under reducing
Reference solution. A 0.5 mg per ml solution of interferon conditions in the test for impurities of molecular masses differing
alfa-2 RS in water. from that of interferon alfa-2. The principal band in the
Isoelectric point calibration solution pI range 3.0 to 10.0. electropherogram obtained with test solution (a) corresponds
Prepare and use according to the manufacturer’s instructions. in position to the principal band in the electropherogram
obtained with reference solution (a).
Isoelectric focusing is carried out using either horizontal
electrophoresis system or by vertical electrophoresis system D. Determine by peptide mapping (2.3.47).
as per the procedure described below or by any appropriate Test solution. Dilute the preparation under examination with
validated method. water to produce a solution containing 0.5 mg protein per ml.

16
IP 2007 INTERFERON ALFA-2 CONCENTRATED SOLUTION

Transfer 25 µl to a microfuge tube of 1.5 ml capacity. Add 1.6 µl Sample buffer (reducing conditions). Mix equal volumes of
of 1 M phosphate buffer solution pH 8.0, 2.8 µl of a freshly water and concentrated SDS PAGE sample buffer for reducing
prepared 1.0 mg per ml solution of trypsin in water (suitable conditions containing 2-mercaptoethanol as the reducing
for peptide mapping) and 3.6 µl of water and mix vigorously. agent.
Cap the tube and place it in a water-bath at 37º for 18 hours.
Test solution (a).Dilute the preparation under examination in
Add 100 µl of a 573 g per litre solution of guanidine
the sample buffer to obtain a solution containing a
hydrochloride and mix well. Add 7 µl of 154.2 g per litre solution
concentration of 0.5 mg protein per ml.
of dithiothreitol and mix well. Place the capped tube in boiling
water for 1 minute and cool to room temperature. Test solution (b). Dilute 0.2 ml of test solution (a) to 1 ml with
Reference solution. A 0.5 mg per ml solution of the appropriate the sample buffer.
interferon alfa-2 RS prepared at the same time and in the same Reference solution (a). Prepare a 0.625 mg per ml solution of
manner as the test solution. the appropriate interferon alfa-2 RS in the sample buffer.
Chromatographic system Reference solution (b). Dilute 0.2 ml of reference solution (a)
– a stainless steel column 100 cm x 4.6 mm, packed with to 1 ml with the sample buffer.
octadecylsilyl silica gel (5 µm) with a pore size of 30 nm,
Reference solution (c). Dilute 0.2 ml of reference solution (b)
– mobile phase A.1 ml of trifluoroacetic acid dilute to
to 1 ml with the sample buffer.
1000 ml with water,
B. Add 1 ml of trifluroacetic acid to Reference solution (d). Dilute 0.2 ml of reference solution (c)
100 ml of water and dilute to 1000 ml with acetonitrile, to 1 ml with the sample buffer.
– flow rate. 1 ml per minute,
Reference solution (e). Dilute 0.2 ml of reference solution (d)
– a linear gradient programme using the conditions given
to 1 ml with the sample buffer.
below,
– spectrophotometer set at 214 nm, Reference solution (f). Use a solution of molecular mass
– a 100 µl loop injector. standards suitable for calibrating SDS-PAGE gels in the range
Time Mobile Mobile Comment 15 kDa to 67 kDa.
phase A phase B Place the test and reference solutions, contained in covered
(min) (per cent v/v) (per cent v/v) test-tubes, on a water-bath for 2 minutes.
0.8 100 0 isocratic
Apply 10 µl of reference solution (f) and 50 µl of each of the
8.68 100 → 04 0 → 60 linear gradient
other solutions to the stacking gel wells. Perform the
68.72 40 60 isocratic electrophoresis under the conditions recommended by the
72.75 40 → 100 60 → 0 linear gradien manufacturer of the equipment. Detect proteins in the gel by
75.08 100 0 re-equilibration silver staining.
Equilibrate the column with mobile phase A for at least 15 The test is not valid unless (1) the validation criteria are met;
minutes maintaining the temperature of the column at 30º. (2) a band is seen in the electropherogram obtained with
Inject the test solution and the reference solution. The reference solution (e); (3) a gradation of intensity of staining
chromatogram obtained with each solution should be is seen in the electropherograms obtained, respectively, with
qualitatively similar to the chromatogram of interferon alfa-2. test solution (a) and test solution (b) and with reference
The profile of the chromatogram obtained with the test solutions (a) to (e).
solution should also correspond to that of the chromatogram The electropherogram obtained with test solution (a) under
obtained with the reference solution. reducing conditions may show, additional bands but no such
band should be more intense than the band obtained with
Tests
reference solution (d). Further, not more than 3 such bands
Impurities of molecular masses differing from that of should be more intense than the principal band obtained with
interferon alfa-2. Determine by electrophoresis (sodium reference solution (e).
dodecyl sulphate polyacrylamide gel electrophoresis (SDS- The electropherogram obtained with test solution (a) under
PAGE) (2.4.12). The test is performed under both reducing non-reducing conditions may show, in addition to the principal
and non-reducing conditions, using resolving gels of 14 per band, less intense bands with molecular masses higher than
cent acrylamide and silver staining as the detection method. the principal band. No such band is more intense than the
Sample buffer (non-reducing conditions). Mix equal volumes principal band in the electropherogram obtained with reference
of water and concentrated SDS PAGE sample buffer. solution (d) and not more than three such bands are more

17
INTERFERON ALFA-2 CONCENTRATED SOLUTION IP 2007

intense than the principal band in the electropherogram 1.0. Consider only the peaks whose retention time is 0.7 to 1.4
obtained with reference solution (e). relative to that of the principal peak.
Related proteins. Determine by liquid chromatography (2.4.14). In the chromatogram obtained with the test solution, the area
of any peak, apart from the principal peak, is not greater than
0.25 per cent w/w hydrogen peroxide solution. Dilute
3.0 per cent of the total area of all of the peaks. The sum of the
hydrogen peroxide solution with water to obtain 0.25 per
areas of any peaks other than the principal peak is not greater
cent w/w solution.
than 5.0 per cent of the total area of all of the peaks.
Test solution. Dilute the preparation under examination with
Bacterial endotoxins (2.2.3). Not more than 100 Endotoxin
water to obtain a solution containing 0.5 mg protein per ml.
Units per mg of protein.
Reference solution. To a volume of the test solution, add a
suitable volume of the 0.25 per cent hydrogen peroxide solution Assay
to give a final hydrogen peroxide concentration of 0.005 per Protein
cent, and allow to stand at room temperature for 1 hour, to
generate about 5 per cent oxidised interferon. Add 12.5 mg of Test solution. Dilute the preparation under examination with
L-methionine per ml of the solution. Allow to stand at room water to obtain a concentration of about 0.5 mg of interferon
temperature for 1 hour. Store the solutions for not longer than alfa-2 per ml.
24 hours at a temperature of 2º to 8º. Reference solutions. Prepare a stock solution of 0.5 mg per ml
Chromatographic system of bovine albumin. Prepare eight dilutions of the stock
– a stainless steel column 25 cm x 4.6 mm, packed with solution containing between 3 µg per ml and 30 µg per ml of
octadecylsilyl silica gel (5 µm) with a pore size of 30 nm, bovine albumin.
– mobile phase: A. To 700 ml of water add 2 ml of Prepare 30-fold and 50-fold dilutions of the test solution.
trifluoroacetic acid and 300 ml of acetonitrile,
Prepare a mixture of 2.0 ml of a 2.0 per cent w/v solution of
B. To 200 ml of water add 2 ml of
copper sulphate in water, 2.0 ml of a 4.0 per cent w/v solution
trifluoroacetic acid and 800 ml of acetonitrile,
of sodium tartrate in water and 96.0 ml of a 4.0 per cent w/v
– flow rate. 1 ml per minute,
solution of sodium carbonate in 0.2 M sodium hydroxide.
– a linear gradient programme using the conditions given
below, Add 1.25 ml of the above mixture to the test-tube containing
– spectrophotometer set at 210 nm, 1.5 ml of water to prepare the blank, 1.25 ml to the test tube
– a 100 µl loop injector containing different dilutions of the sample and 1.25 ml to the
Time Mobile Mobile Comment test tube with the reference solution.
phase A phase B Mix after each addition and after approximately 10 minutes,
(min) (per cent v/v) (per cent v/v) add to each test-tube 0.25 ml of a mixture of equal volumes of
0.1 72 280 isocratic water and phosphomolybdotungstic reagent. Mix after each
1.5 72 → 67 28 → 33 linear gradient addition. After 30 minutes, measure the absorbance of each
solution at 750 nm (2.4.7) using the blank as the compensation
5 – 20 67 → 63 33 → 37 linear gradient
liquid.
20 – 30 63 → 57 37 → 43 linear gradient
Draw a calibration curve from the absorbances of the eight
30 – 40 57 → 40 43 → 60 linear gradient
reference solutions and the corresponding protein contents
40 – 42 40 60 isocratic and read from the curve the content of protein in the test
42 – 50 40 → 72 60 → 28 linear gradien solution.
50 – 60 72 28 re-equilibration
Potency
Equilibrate the column with the mobile phases in the initial
gradient ratio for at least 15 minutes. The potency of interferon alfa-2 is estimated based on its
ability to protect cells against a viral cytopathic effect compared
Inject alternatively the test solution and the reference solution. to the protection accorded by an appropriate International
Interferon alfa-2 elutes at a retention time of about 20 minutes Standard of human recombinant interferon alfa-2 or of a
in the chromatogram. With the reference solution a peak related reference preparation calibrated in International Units.
to oxidized interferon appears at a retention time of about 0.9 The International Unit is the activity contained in a stated
relative to the principal peak. amount of the appropriate International Standard. The
The test is not valid unless the resolution between the peaks equivalence in International Units of the International Standard
corresponding to oxidised interferon and interferon is at least is stated by the World Health Organisation.

18
IP 2007 STREPTOKINASE BULK SOLUTION

Carry out the assay by a suitable method, based on the demonstrate that the product, if tested, would comply with
following design. the following test.
Use, an established cell line sensitive to the cytopathic effect Abnormal toxicity
of a suitable virus, responsive to interferon.
Inject into each mouse a quantity of the preparation under
The following cell cultures and virus have shown to be suitable: examination (diluted, if necessary, with water for injections)
MDBK cells (ATCC No. CCL22), or Mouse L cells (NCTC containing 50,000 IU of streptokinase activity in 0.5 ml, the
clone 929; ATCC No.CCL 1) as the cell culture and vesicular injection lasting 15-20 seconds.
stomatitis virus VSV, Indiana strain (ATCC No. VR-158) as the
Description. A clear, colourless liquid.
infective agent; or human diploid fibroblast FS-71 cells
responsive to interferon as the cell culture, and
encephalomyocarditis virus (ATCC No.VR-129B) as the
Identification
infective agent. A. Place 0.5 ml of citrated human plasma in a haemolysis
Incubate in at least three groups, cells with three or more tube maintained in a water-bath at 37º. Add 0.1 ml of a dilution
different concentrations of the preparation under examination of the preparation under examination containing 10,000 IU of
and one with the reference preparation in a microtitre plate. streptokinase activity per ml in phosphate buffer pH 7.2 and
Include appropriate controls of untreated cells in each group. 0.1 ml of a solution of human thrombin RS containing 20 IU
per ml in phosphate buffer pH 7.2. Mix immediately. A clot
Choose the concentrations of the preparations such that the forms and lyses within 30 minutes. Repeat the procedure using
lowest concentration produces some protection and the citrated bovine plasma. The clot does not lyse within 60
largest concentration produces less than maximal protection minutes.
against the viral cytopathic effect.
B. Dissolve 0.6 g of agar in 50.0 ml of barbitone buffer pH 8.6,
Add the cytopathic virus after the cells have established to all heating until a clear solution is obtained. Use glass plates 50
wells except the control wells. mm square (transparency mounts) free from traces of grease.
Determine the cytopathic effect of virus quantitatively and Using a pipette, apply to each plate 4 ml of the agar solution.
calculate the potency of the preparation to be examined by Maintain the plates horizontal. Allow to cool. Pierce a cavity 6
the usual statistical methods for a parallel line assay. mm in diameter in the centre of the agar and an appropriate
number of cavities (not exceeding 6) at distances of 11 mm
The estimated potency is not less than 80 per cent and not
from the central cavity. Remove the residual agar from the
more than 125 per cent of the stated potency. The confidence
cavities using a cannula connected to a vacuum pump. Using
limits of the estimated potency (P= 0.95) are not less than 64
pipettes graduated in microlitres, place in the central cavity
per cent and not more than 156 per cent of the stated potency.
about 80 µl of goat or rabbit antistreptokinase serum containing
Storage. Store protected from light, at or below –20º. 10,000 units of antistreptokinase activity per ml; place in each
Labelling. The label states (1) The type of interferon (alfa-2a of the surrounding cavities about 80 µl of a dilution of the
or alfa-2b); (2) the type of production. preparation under examination containing 125,000 IU of
streptokinase activity per ml. Allow the plates to stand in a
humidified tank for 24 hours. Only one precipitation arc appears
and it is well defined and localised between the application
Streptokinase Bulk Solution point of the serum and each cavity containing the solution of
the preparation under examination.
Streptokinase Bulk Solution is a fibrinolytic enzyme present
in certain strains of haemolytic Streptococcus group C. It has
Tests
the property of combining with human plasminogen to form
plasminogen activator. Streptokinase is also produced by a pH (2.4.24). 6.8 to 7.5, determined in a solution prepared by
method based on recombinant DNA technology using bacteria diluting the preparation under examination in carbon dioxide-
or suitable genetically engineered host cells. free water to produce a solution containing 5000 IU of
Streptokinase Bulk Solution has a potency of not less than streptokinase activity per ml.
96,000 IU per mg of protein. Streptodornase. Not more than 10 IU of streptodornase
activity per 100,000 IU of streptokinase activity.
Production
Test solution. Dilute the preparation under examination in
If intended for use in the manufacture of parenteral imidazole buffer pH 6.5 to obtain a solution containing 150,000
preparations, the method of manufacture is validated to IU of streptokinase activity per ml.

19
STREPTOKINASE BULK SOLUTION IP 2007

Reference solution. Dissolve in imidazole buffer pH 6.5 a Potency


reference preparation of streptodornase, calibrated in
International Units against the International Standard of The potency of streptokinase is determined by comparing its
streptodornase, to obtain a solution containing 20 IU of capacity to activate plasminogen to form plasmin with the
streptodornase activity per ml. The equivalence in International same capacity of a reference preparation of streptokinase
Units of the International Standard is stated by the regulatory calibrated in International Units; the formation of plasmin is
authority. determined using a suitable chromogenic substrate.

To each of 8 numbered centrifuge tubes, add 0.5 ml of a 0.1 per The International Unit is the activity of a stated amount of the
cent solution of sodium deoxyribonucleate in imidazole buffer International Standard for streptokinase. The equivalence in
pH 6.5. To tube number 1 and tube number 2 add 0.25 ml of International Units of the International Standard is stated by
imidazole buffer pH 6.5, 0.25 ml of the test solution and, the regulatory authority.
immediately, 3.0 ml of 2.5 per cent w/v of perchloric acid. Mix, Reference and test solutions. Prepare 2 independent series of
centrifuge at about 3000 rpm for 5 minutes and measure the 4 dilutions of each of the substance under examination and of
absorbances of the supernatant liquids at 260 nm (2.4.7), using the reference preparation of streptokinase in tris
as the compensation liquid a mixture of 1.0 ml of imidazole (hydroxymethyl) aminomethane sodium chloride buffer pH
buffer pH 6.5 and 3.0 ml of 2.5 per cent w/v solution of 7.4, in the range of 0.5-4.0 IU per ml. Prepare and maintain all
perchloric acid (absorbances A1 and A2). To the other 6 solutions at 37º.
tubes (numbers 3 to 8) add 0.25 ml, 0.25 ml, 0.125 ml, 0.125 ml,
0 ml and 0 ml respectively of imidazole buffer solution pH 6.5; Substrate solution. Mix 1.0 ml of tris (hydroxymethyl)
add to each tube 0.25 ml of the test solution and 0 ml, 0 ml, aminomethane buffer pH 7.4 with 1.0 ml of chromophore
0.125 ml, 0.125 ml, 0.25 ml and 0.25 ml respectively of the substrate. Add 5 µl of a 10 per cent w/v solution of polysorbate
reference solution. Mix the contents of each tube and heat at 20. Keep at 37º in a water-bath. Immediately before commencing
37º for 15 minutes. To each tube add 3.0 ml of 2.5 per cent w/v the activation assay, add 45 µl of a 1 mg per ml solution of
of perchloric acid, mix and centrifuge. Measure the human plasminogen.
absorbances of the supernatant liquids at 260 nm (2.4.7), using Analyse each streptokinase dilution, maintained at 37º, in
the compensation liquid described above (absorbances A3 to duplicate. Initiate the activation reaction by adding 60 µl of
A8). The absorbances comply with the following test. each dilution to 40 µl of substrate solution. For blank wells,
use 60 µl of tris (hydroxymethyl) aminomethane sodium
(A5 + A6 + A7 + A8) chloride buffer solution pH 7.4 instead of the reference and
(A3 + A4) − (A1 + A2) < − (A3 + A4)
2 test solutions. Allow the reaction to proceed at 37º for 20
minutes and read the absorbance at 405 nm (2.4.7). If a suitable
Streptolysin thermostated plate reader is available, this may be used to
monitor the reaction. Alternatively, it may be necessary to
In a haemolysis tube, use a quantity of the preparation under
stop the reaction after 20 minutes using 50 µl of a 50 per cent
examination containing 500,000 IU of streptokinase activity
v/v solution of glacial acetic acid. Best results are obtained
and dilute to 0.5 ml with a mixture of 1 volume of phosphate
when the absorbance for the highest streptokinase
buffer pH 7.2 and 9 volumes of a 0.9 per cent w/v solution of
concentration is between 0.1 and 0.2 (after blank subtraction).
sodium chloride. Add 0.4 ml of a 2.3 per cent w/v solution of
If necessary, adjust the time of incubation in order to reach
sodium thioglycollate. Heat in a water-bath at 37º for 10
this range of absorbances.
minutes. Add 0.1 ml of a solution of a reference preparation of
human antistreptolysin O containing 5 IU per ml. Heat at 37º Calculate the regression of the absorbance on log
for 5 minutes. Add 1 ml of rabbit erythrocyte suspension. concentrations of the solutions of the substance under
Heat at 37º for 30 minutes. Centrifuge at about 1000 rpm. In the examination and of the reference preparation of streptokinase
same manner, prepare a haemolysis tube in which the solution and calculate the potency of the substance under examination
of the preparation under examination has been replaced by 0.5 using the usual statistical methods for parallel-line assays.
ml of a mixture of 1 volume of phosphate buffer pH 7.2 and 9
The estimated potency is not less than 90 per cent and not
volumes of a 0.9 per cent w/v solution of sodium chloride.
more than 111 per cent of the stated potency. The confidence
Measure the absorbances of the supernatant liquids at 550
limits (P = 0.95) of the estimated potency are not less than 80
nm (2.4.7). The absorbance of the test solution is not more
per cent and not more than 125 per cent of the stated potency.
than 50 per cent than that of the reference solution.
Streptokinase Bulk Solution intended for use in the
Protein. Determine the nitrogen content (2.3.30).
manufacture of parenteral preparations without a further
1 mg of N is equivalent to 6.25 mg of protein. appropriate procedure for the removal of bacterial

20
IP 2007 STREPTOKINASE BULK SOLUTION

endotoxins complies with the following additional Labelling. The label states (1) the number of International
requirement. Units of streptokinase activity per mg, calculated on the dried
basis; (2) the name and quantity of any added substance; (3)
Bacterial endotoxins (2.2.3). Less than 0.02 Endotoxin Unit
where applicable, that the substance is free from bacterial
per 100 IU of streptokinase activity.
endotoxins; (4) where applicable, that the substance is suitable
Storage. Store protected from light and at a temperature of for use in the manufacture of parenteral preparations.
about - 20°. If it is intended for the manufacture of parenteral
preparations, the container should be sterile and sealed so as
to exclude micro-organisms.

21
INDIAN PHARMACOPOEIA 2007 GENERAL MONOGRAPHS

VETERINARY PRODUCTS

General Monographs
Dip Concentrates ..................................................................................................
Intramammary Infusions .........................................................................................
Premixes ................................................................................................................
Veterinary Aerosols ................................................................................................
Veterinary Diagnostics ............................................................................................
Veterinary Oral Liquids ..........................................................................................
Veterinary Oral Powders .......................................................................................
Veterinary Parenteral Preparations ..........................................................................
Veterinary Tablets ..................................................................................................
Monographs
Non Biological .......................................................................................................
Biological ...............................................................................................................
Veterinary Vaccines ..............................................................................................

1499
IP 2007 GENERAL MONOGRAPHS

Veterinary Preparations suitable vehicle. They may contain stabilizing, emulsifying,


suspending and thickening agents. If a sediment is formed in
a suspension, it is readily dispersible on shaking. In emulsions,
General Requirements
phase separation may occur but this is readily miscible on
The general requirements relating to a specific type of dosage shaking.
form of an active pharmaceutical ingredient or ingredients, There are two main types of Intramammary Infusions. One is
that have been given in the chapter on General Monographs intended for administration to lactating animals as qualified
on Dosage Forms of Active Pharmaceutical Ingredients apply by the term Lactating Cow/Buffalo and the other, qualified as
to all veterinary dosage forms or preparations of the type Non-lactating or Dry Cow/Buffalo, is intended for
defined. However, a valid interpretation of the appropriateness administration to animals at the end of lactation or during the
of a test or requirement should be done in the context of the
non-lactating period for the prevention or treatment of infection
monograph as a whole and of the relevant General Notices.
during the dry period.
The requirement for compliance with the tests given under
each dosage form or preparation is indicated in each Intramammary Infusions are prepared by dissolving or
monograph of a drug product or preparation under the heading suspending the sterile medicaments in the sterilized vehicle
‘Other tests’. These tests are mandatory and are additional to using aseptic precautions, unless a process of terminal
the tests given in the individual monograph. sterilisation is employed.
Containers. Intramammary Infusions are usually supplied in
single dose containers for administration into a single teat
canal of an animal. If supplied in multiple dose containers,
Dip Concentrates aqueous preparations contain an antimicrobial preservative
Dip concentrates are preparations for the prevention and in adequate concentration except when the preparation itself
treatment of ectoparasitic infestations of animals. They contain has antimicrobial properties. The containers are made as far
one or more medicaments, usually in the form of wettable as possible from materials that meet the requirements for
powders, pastes or solutions from which diluted suspensions Parenteral Preparations intended for use in human beings.
or emulsions are prepared by appropriate dilution with the
The containers are sealed so as to exclude micro-organisms
recommended liquid. The diluted preparations are applied by
and each container is fitted with a smooth, tapered nozzle to
complete immersion of the animal or by spraying, as
appropriate. They contain suitable antimicrobial preservatives. facilitate the introduction of the infusion into the teat canal.
The containers are sterilised and filled aseptically unless the
Labelling. The label states (1) the name(s) and proportion(s) preparation is subjected to a process of terminal sterilisation.
of medicament(s); (2) the name and proportion of any added
antimicrobial preservative; (3) the name and quantity of the Tests
diluent and the manner of preparing the diluted dip solution
or spray; (4) any special precautions to be taken for use of the Sterility. Intramammary Infusions comply with the test for
preparation; (6) the storage conditions; (6) the date after which sterility (2.2.11), using Method A or B, as appropriate, using
the preparation is not intended to be used. the contents of 10 containers mixed thoroughly before use in
If the preparation contains an organophosphorus compound the test. Use for each medium 0.5 to 1.0 g or 0.5 to 1.0 ml, as
the label also states (1) that the preparation contains an appropriate, of the mixed sample.
organophosphorus compound; (2) and special precautions Storage. Store in sterile, single dose or multiple dose, tamper-
on the use of the preparation.
evident containers.
Labelling. The label states (1) the strength in terms of the
weight or the number of Units of activity of the active
Intramammary Infusions ingredient(s) or that may be expressed from the container using
normal techniques; (2) whether the preparation is intended
Intramammary Infusions for Veterinary Use;
for use in lactating cow/buffalo or in dry or non-lactating
Intramammary Injections. cow/buffalo; (3) for Intramammary Infusions (Non-lactating
Intramammary Infusions are sterile products intended for or Dry Cow/Buffalo), that the preparation is not intended for
injection into the mammary gland through the teat canal. They use in lactating animals; (4) in the case of infusions in multiple
are solutions, emulsions or suspensions or semi-solid dose containers, the name of any added antimicrobial
preparations containing one or more active ingredients in a preservative.

1501
GENERAL MONOGRAPHS IP 2007

Premixes Labelling. The label states (1) that the aerosol is intended for
external use only; (2) the instructions for use; (3) any special
Premixes are mixtures of one or more active ingredients with precautions in the use of the preparation.
suitable bases intended for mixing with feedstuffs before
administration to the animals. They are used to dilute
medicament(s) with the feed and are usually issued as pellets,
granules or powders. If issued as granules, these are free- Veterinary Diagnostics
flowing and free from aggregates. Suitable precautions are
Veterinary Diagnostics are antigenic materials of bacterial or
taken during manufacture for ensuring that the premix is
viral origin employed for various tests. These will also include
homogeneous.
polyclonal or monoclonal antibodies. The preparations are
Unless otherwise stated in the individual monograph, the examined for their purity at various critical stages of
concentration of the premix in medicated feedstuffs is not less production. The diagnostic kits may be prepared using
than 0.5 per cent. bacterial or viral antigens and antisera.

Tests Tests
Loss on drying (2.4.19). Not more than 15.0 per cent, Veterinary Diagnostics, reconstituted where necessary,
determined on 3 g by drying in an oven at 105º for 2 hours. comply with the following tests unless otherwise stated in
Labelling. The label states (1) the strength in terms of the the individual monograph.
amount of active ingredient(s) as a percentage; (2) the category
of animal for which the premix is intended to be used; (3) the Identification
directions for the preparation of the medicated feed; (4) where Unless otherwise stated in the individual monograph,
applicable, the minimum interval between the stoppage of Veterinary Diagnostics give specific reaction when injected
feeding of the diluted premix and the slaughter of the animal into the skin of a healthy white guinea-pig or rabbit that has
for human consumption; (5) any special precautions to be not been previously treated with any material that will interfere
taken for use of the premix; (6) the storage conditions; (7) the with the test but fails to produce this reaction when mixed
date after which the preparation is not intended to be used. with a sufficient quantity of the specific antitoxin or antiserum.
Sterility. Unless otherwise stated, Veterinary Diagnostics
comply with the test for sterility (2.2.11), except that in the
Veterinary Aerosols case of preparations containing living bacteria there may be
growth of the organism from which the diagnostic was
Veterinary Sprays prepared.
Veterinary Aerosols are solutions, suspensions or emulsions Use suitable solid media for streaking the preparation under
of one or more active ingredients intended for use by external examination and incubate at 32º to 37º for 72 hours for detecting
application. They may contain auxiliary substances such as bacteria and at 20º to 25º for 72 hours for detecting fungi. The
solvents, solubilising agents, emulsifying agents and media selected will depend upon the nature of the product to
suspending agents. They are delivered in the form of an aerosol be tested. The contents of each randomly selected sealed
by the actuation of an appropriate valve or by means of a container of the preparation under examination or portions or
suitable atomizing device that is either an integral part of the dilutions thereof, as appropriate, are used for the test.
container or is supplied separately.
Other tests to determine the nature and identity of
They may be presented in special containers under pressure contaminating microorganisms, if any, detected during the
of a gas and contain propellants or mixtures of propellants. test include examination for mobility of the organisms,
The medicaments are released from the container in the form fermentation reactions, thermo-agglutination tests and dye
of an aerosol upon actuation of an appropriate valve. inhibitor tests (in the case of Brucella cultures).
Veterinary Aerosols supplied in special pressurized containers Unless otherwise stated in the monograph, the preparation
comply with the appropriate requirements for Inhalation passes the test if no growth of microorganisms, other than
Preparations. The following requirements also apply for any those from which the veterinary diagnostic was prepared, is
veterinary aerosol that is the subject of an individual observed in any of the media during the incubation period.
monograph. Repeat the tests if growth of organisms, other than those from
Containers. A suitable atomizing device may form part of the which the veterinary diagnostic was prepared, is observed.
container or is supplied separately. The vaccine passes the test if no growth of microorganisms,

1502
IP 2007 GENERAL MONOGRAPHS

other than those from which the diagnostic was prepared, is given in the chapter on General Monographs on Dosage Forms
observed in any of the media. The preparation fails the test if of Active Pharmaceutical Ingredients.
growth of a microorganism that was seen after the first test,
other than those from which the veterinary diagnostic was
prepared, is observed. If growth of a different microorganism
is observed, the test may be repeated a second time. The Veterinary Tablets
preparation passes the test if no growth of a microorganism, Veterinary tablets are usually solid, circular cylinders the end
other than those from which the veterinary diagnostic was surfaces of which are flat or biconvex and the edges of which
prepared, is observed in any of the media. are bevelled except that those weighing 5 g of more may be
The number of containers recommended to be drawn by the elongated or biconical.
manufacturer for performing the test for sterility depends on
the environmental conditions of manufacture, the volume of Tests
preparation per container and any other special considerations Disintegration (2.5.1). The test may have to be suitably
applicable to the preparation concerned. For preparations modified in the case of large tablets; the discs may have to be
intended for veterinary use, 1 per cent of the containers in a omitted because they would otherwise be dislodged from the
batch, with a minimum of three and a maximum of ten, is disintegration tubes. It may also be necessary to adjust the
considered a suitable number assuming that the preparation volume of the disintegration medium so that the tablet does
has been manufactured under appropriately validated not break the surface of the medium at the top of the up-
conditions designed to exclude contamination. stroke, care being taken to apply the minimum practical volume
Storage. Store protected from light in a refrigerator (2º to 8º) of liquid for this purpose. For certain tablets where the diameter
unless otherwise stated in the individual monograph. of the tablet may not permit adequate movement of the
disintegration medium, the apparatus and the method should
Labelling. The label states (1) the name and quantity of any
be suitably modified.
antibacterial substance added; (2) for a dried preparation, the
nature and quantity of the liquid to be used for reconstitution.

Veterinary Vaccines
Veterinary Oral Liquids
Vaccines for Veterinary Use
Veterinary oral liquids intended for administration in large
animals may also be called Drenches. Vaccines are a heterogeneous class of medicinal products
containing immunogenic substances capable of inducing
specific, active and protective host immunity against
infectious diseases. They may be prepared from bacteria,
Veterinary Oral Powders viruses, parasites or other organisms or their toxins. Vaccines
Veterinary Oral Powders are intended for oral administration, may contain live attenuated or avirulent microorganisms or
usually after dilution in drinking water or the feed. They may these may consist of killed or inactivated microorganisms.
be in the form of soluble or wettable powders. Some vaccines consist of antigenic fractions or substances
produced by the same pathogenic organisms but rendered
Labelling. The label states (1) for single dose containers, the
harmless whilst retaining their immunogenecity. Vaccines may
name and quantity of active medicament(s) per container; (2)
be prepared from one species or from two or more species of
for multiple dose containers, the name and quantity of active
microorganisms.
medicament(s) by weight; (3) the name of any added
antimicrobial preservative(s); (4) the directions for use of the Vaccines may be prepared by the method described in the
preparation. individual monograph or by any other appropriate method
provided the identity of the antigens is maintained and the
preparations are free from microbial contamination and
Veterinary Parenteral Preparations extraneous agents. Suitable adjuvants may be added during
the preparation of the vaccines. The addition of antibiotics
Veterinary Parenteral Preparations prepared with oily vehicles during the manufacturing process is normally restricted to
are not meant for intravenous administration but are suitable cell culture fluids and other media, egg inocula and material
for intramuscular or subcutaneous use. harvested from skin or other tissues. A suitable bactericide
Veterinary Parenteral Preparations comply with the appropriate may be added to sterile and inactivated vaccines. The final
requirements for Parenteral Preparations (Injections) that are products are distributed aseptically into sterile containers that

1503
GENERAL MONOGRAPHS IP 2007

are then sealed to exclude extraneous microorganisms. Unless Live viral vaccines. Live viral vaccines are prepared using
otherwise indicated in the monograph, the final vaccine may avirulent or attenuated strains of the specific viruses that are
be filled into single dose or multiple dose containers; however, capable of stimulating immunogenic response against
inactivated vaccines in multiple dose containers must pathogenic strains of the same or of antigenically related
invariably contain a bactericide. viruses.
Bacterial vaccines. Bacterial vaccines are either suspensions Inactivated viral vaccines. Inactivated viral vaccines contain
of live or killed bacteria or sterile antigenic extracts or viruses that have been inactivated by suitable chemical or
derivatives of bacteria pathogenic to animals. They may be physical means in such a way that the preparations retain
simple vaccines prepared from one species or may be combined adequate immunogenicity or they are suspensions of
or polyvalent vaccines prepared by blending two or more immunogenic components of such viruses.
simple vaccines from different species or strains. Bacterial
Combined vaccines. Combined vaccines consist of two or
vaccines may be prepared from cultures grown on suitable
more antigens, combined by the manufacturer at the final
solid or liquid media. The identity, antigenic potency and purity
formulation stage or mixed immediately before administration.
of each bacterial culture must be carefully controlled.
Such vaccines are intended to protect against either more
Bacterial vaccines are suspensions of varying degrees of than one disease, or against one disease caused by different
opacity in colourless or slightly coloured liquids or they may strains or serotypes of the same organism.
be freeze-dried so that the water content is not more than 3.0
Stability. Stability is the ability of a vaccine to retain its
per cent w/w unless otherwise stated in the individual
chemical, physical, microbiological and biological properties
monograph. They may be standardised in terms of international
within specified limits throughout its shelf life.
opacity units or, where appropriate, by numbers of live or
killed bacteria determined by viable count or by direct cell Adjuvants. Substances that are intended to enhance relevant
count. immune response and subsequent clinical efficacy of the
Live bacterial vaccines. Live bacterial vaccines are prepared vaccine.
from avirulent or attenuated strains of the specific bacteria
Tests
that are capable of stimulating immune response against
pathogenic strains of the same or of antigenically related Vaccines comply with the tests prescribed in the individual
species of bacteria. monographs including, where applicable, the following:
Inactivated bacterial vaccines. Inactivated bacterial vaccines Aluminium (2.3.9). Where an aluminium adsorbent has been
are either prepared from bacteria or their immunogenic used in the vaccine, not more than 1.25 mg of aluminium (Al)
components that have been inactivated in a suitable way that per single dose, unless otherwise stated.
they retain adequate immunogenicity.
Calcium (2.3.11). Where a calcium adsorbent has been used
Bacterial toxoids. Bacterial toxoids are prepared from toxins in the vaccine, not more than 1.3 mg of calcium (Ca) per single
by diminishing their toxicity to a very low level or by dose, unless otherwise stated.
completely eliminating it by physical or chemical means whilst
retaining adequate immunising potency. The toxins are Formaldehyde (2.3.20). Where formaldehyde has been used
obtained from selected strains of specific microorganisms, in the preparation of the vaccine, not more than 0.2 g/l of free
grown in suitable media devoid of agents capable of inducing formaldehyde is present in the final product, unless otherwise
undesirable immunological reactions in animals. Bacterial stated.
toxoids may be liquid or may be prepared by adsorbing on Phenol (2.3.36). Where phenol has been used in the
suitable agents such as aluminium phosphate, aluminium preparation of the vaccine, not more than 2.5 g/l is present in
hydroxide or any other suitable adsorbents. Bacterial toxoids the final product, unless otherwise stated.
are clear or slightly opalescent liquids, colourless or slightly
Water (2.3.43). For freeze-dried vaccines, not more than 3.0
yellow. Adsorbed toxoids may be white or greyish-white
per cent, unless otherwise stated.
suspensions or paleyellow liquids with sediment at the bottom
of the container. Freeze-dried preparations are greyish-white Thiomersal (2.3.12) (2.3.48). Where thiomersal has been used
or yellowish-white powders or pellets. in the preparation of the vaccine, not more than 0.02 per cent
Viral vaccines. Viral vaccines are suspensions of viruses or w/v.
preparations obtained from tissues or blood of animals Extraneous pathogens. Unless otherwise stated in the
artificially infected with viruses pathogenic to animals or from individual monograph, live viral vaccines other than those
cultures in fertile eggs, or from cell or tissue cultures, they intended for poultry comply with the following test. The
may be live, inactivated / killed and may be freeze-dried. mixture obtained after neutralisation of the vaccine with specific

1504
IP 2007 GENERAL MONOGRAPHS

antiserum does not cause cytopathic effects in cell cultures being examined contains an adjuvant, inject 2 ml of the vaccine
known to be sensitive to agents pathogenic for the species in subcutaneously into each guinea pig. Observe the animals
which the vaccine is intended to be used. for 7 days. None of the animals shows significant local or
systemic reaction. If one animal dies or shows signs of ill
Sterility (2.2.11). Unless otherwise stated in the individual
health during the observation period repeat the test. None of
monograph, use method A. Incubate the media for not less
the animals of the second group dies or shows signs of ill
than 14 days at 30º to 37º in the test for detecting bacteria and
health. This test may be omitted if a safety test is carried out
at 20º to 25º in the test for detecting fungi. However, for live
on animals of the species for which the vaccine is intended.
bacterial vaccines growth of the organism from which the
vaccine was prepared is permitted. Potency. Determine the potency of the vaccine using the
method described in the individual monograph. The vaccine
The number of containers to be drawn for the test should be 1
complies with the level of immune response specified in the
per cent of the containers in a batch, with a minimum of 3 and
monograph. A combined vaccine complies with the level
a maximum of 10, assuming that the preparation has been
specified in the respective monographs for each individual
manufactured under appropriately validated conditions
component. If the immunogenicity test (potency test) has been
designed to exclude contamination.
performed with satisfactory results on representative batch
Safety Test. Unless otherwise stated in the individual of live vaccine from the same seed lot, it may be omitted as a
monograph, vaccines other than live viral vaccines intended routine control test during production of other batches of the
for poultry comply with the following test. vaccine prepared from the same seed lot.
Inject at least 2 healthy, susceptible animals of one of the Storage. Store protected from light in a refrigerator (2º to 8º),
species in which the vaccine is intended to be used by the unless otherwise directed. Do not freeze. Store freeze-dried
route recommended by the manufacturer for field use. The vaccines at a temperature not exceeding 20º.
quantity to be injected in each animal is twice the appropriate
Labelling. The label states (1) the potency of the preparation;
vaccinating dose. Observe the animals for not less than 7
(2) the route of administration and dose; (3) the date up to
days. No animal exhibits an abnormal reaction.
which the product is expected to remain within specifications;
Abnormal toxicity. Where stated in the individual monograph (4) the storage conditions.
vaccines comply with the following test.
Inject 0.5 ml subcutaneously into each of five mice and 2 ml
intraperitoneally into each of two guinea pigs. If the vaccine

1505
IP 2007 ACEPROMAZINE MALEATE

Acepromazine Maleate liquid and allow the impregnating solvent to ascend almost to
the top. Use the plate immediately after removing it from the
tank. Apply to the plate 1 µl of each solution. After
CH3 development, dry the plate in air and examine in ultraviolet
N light at 365 nm. The principal spot in the chromatogram
H3 C O
H COOH obtained with the test solution corresponds to the spot in the
N chromatogram obtained with the reference solution. A
CH3 ,
secondary spot due to maleic acid is also observed in both
H COOH chromatograms. Spray the plate with ethanolic sulphuric acid
S
(10 per cent v/v). The spot in the chromatogram obtained
with the test solution corresponds to the spot in the
C19H22N2OS, C4H4O4 Mol. Wt. 442.5 chromatogram obtained with the reference solution.
Acepromazine Maleate is 2-acetyl-10-(3-
D. Dissolve 5 mg in 2 ml of sulphuric acid; a yellow colour is
dimethylaminopropyl)phenothiazine maleate.
produced which changes to deep orange on warming for 2
Description. A yellow, crystalline powder. minutes.
Acepromazine Maleate contains not less than 98.5 per cent E. Dissolve 0.2 g in a mixture of 3 ml of water and 2 ml of 5 M
and not more than 101.0 per cent of C19H22N2OS,C4H4O4, sodium hydroxide and shake with three quantities, each of 3
calculated on the dried basis. ml, of ether. Discard the ether extracts. Add 2 ml of bromine
solution to the aqueous solution, warm in a water-bath for 10
Identification minutes, heat to boiling, cool and add 0.25 ml to a solution of
Tests B and D may be omitted if tests A, C, E and F are carried 10 mg of resorcinol in 3 ml of sulphuric acid; a bluish-black
out. Tests A and E may be omitted if tests B, C, D and F are colour is produced on heating for 15 minutes in a water-bath.
carried out. F. Melting range ( 2.4.21). 136° to 139°.
NOTE — Carry out the tests in subdued light.
Tests
A. Dissolve 20 mg in 2 ml of water, add 3 ml of 2 M sodium
hydroxide, extract with 5 ml of cyclohexane and remove the pH (2.4.24). 4.0 to 4.5, determined in a 1.0 per cent w/v solution.
solvent under reduced pressure. The residue complies with Related substances. Determine by thin-layer chromatography
the following test. (2.4.17), coating the plate with silica gel GF254.
Determine by infrared absorption spectrophotometry (2.4.6).
Solvent mixture. A mixture of 95 volumes of methanol and 5
Compare the spectrum with that obtained with acepromazine
volumes of diethylamine.
RS or with the reference spectrum of acepromazine.
Mobile phase. A mixture of 75 volumes of hexane, 17 volumes
B. When examined in the range 230 nm to 360 nm (2.4.7), a
of 2-butanol and 8 volumes of diethylamine.
0.002 per cent w/v solution in 0.1 M hydrochloric acid, exhibits
maxima at about 244 nm and 280 nm; absorbance at about 244 Test solution. Dissolve 0.2 g of the substance under
nm, about 1.1 and at about 280 nm, about 0.82. examination in 10 ml of the solvent mixture.
C. Determine by thin-layer chromatography (2.4.17), coating Reference solution. A 0.010 per cent w/v solution of the
the plate with kieselguhr G. substance under examination in the solvent mixture.
Mobile phase. A mixture of 100 volumes of light petroleum ( Apply to the plate 10 µl of each solution. After development,
40° to 60° ), 2 volumes of diethylamine and 6 to 8 volumes of dry the plate in air and examine in ultraviolet light at 254 nm.
2-phenoxyethanol. Shake and use the supernatant liquid. Any secondary spot in the chromatogram obtained with the
Test solution. Dissolve 0.2 g of the substance under test solution is not more intense than the spot in the
examination in 100 ml of dichloromethane. chromatogram obtained with the reference solution.

Reference solution. A 0.2 per cent w/v solution of Sulphated ash (2.3.18) Not more than 0.2 per cent.
acepromazine maleate RS in dichloromethane. Loss on drying (2.4.19). Not more than 0.1 per cent, determined
on 1.0 g by drying in an oven at 105°.
Impregnate the dry plate by placing it in a tank containing a
shallow layer of a mixture of 85 volumes of acetone, 10 volumes Assay. Weigh accurately about 0.4 g, dissolve in 50 ml of
of 2-phenoxyethanol and 5 volumes of polyethyleneglycol acetic anhydride. Titrate with 0.1 M perchloric acid, using
300 so that the plate dips about 5 mm below the surface of the crystal violet solution as indicator. Carry out a blank titration.

1507
ACEPROMAZINE INJECTION IP 2007

1 ml of 0.1 M perchloric acid is equivalent to 0.04425 g of chromatogram obtained with the reference solution. A
C19H22N20S,C4H4O4. secondary spot due to maleic acid is also observed in both
chromatograms. Spray the plate with ethanolic sulphuric acid
Storage. Store protected from moisture.
(10 per cent v/v). The spot in the chromatogram obtained
with the test solution corresponds to the spot in the
chromatogram obtained with the reference solution.
Acepromazine Injection D. To a volume containing 25 mg of acepromazine add 2 ml of
Acepromazine Maleate Injection 5 M sodium hydroxide and shake with three quantities, each
of 3 ml, of ether. Discard the ether extracts. Add 2 ml of bromine
Acepromazine Injection is a sterile solution of Acepromazine solution to the aqueous solution, warm in a water-bath for 10
Maleate in Water for Injections. minutes, heat to boiling, cool and add 0.25 ml to a solution of
Acepromazine Injection contains not less than 92.5 per cent 10 mg of resorcinol in 3 ml of sulphuric acid; a bluish-black
and not more than 107.5 per cent of the stated amount of colour is produced on heating for 15 minutes in a water-bath.
acepromazine, C19H22N2OS.
Tests
Identification
pH (2.4.24). 4.5 to 5.5.
NOTE — Carry out the tests in subdued light. Other tests. Complies with the tests stated under Parenteral
A. To a volume containing 20 mg of acepromazine, add 2 ml of Preparations (Injections).
water and 3 ml of 2 M sodium hydroxide, extract with two Assay. To an accurately measured volume containing 40 mg
quantities, each of 5 ml, of cyclohexane and remove the solvent of acepromazine add 5 ml of 1 M sodium hydroxide and extract
under reduced pressure. The residue complies with the with three or more quantities, each of 50 ml, of dichloromethane
following test. until the dichloromethane extract is colourless. Wash the
Determine by infrared absorption spectrophotometry (2.4.6). extracts with the same 10 ml of water and filter through a plug
Compare the spectrum with that obtained with acepromazine of absorbent cotton previously moistened with
RS or with the reference spectrum of acepromazine. dichloromethane. Evaporate the combined extracts to dryness,
dissolve the residue in 15 ml of acetic anhydride. Titrate with
B. To 5 mg of the residue obtained in test A, add 2 ml of 0.02 M perchloric acid, using crystal violet solution as
sulphuric acid; a yellow colour is produced which changes indicator. Carry out a blank titration.
to deep orange on warming for 2 minutes.
1 ml of 0.02 M perchloric acid is equivalent to 0.006529 g of
C. Determine by thin-layer chromatography (2.4.17), coating
C19H22N2OS.
the plate with kieselguhr G.
Storage. Store protected from light.
Mobile phase. A mixture of 100 volumes of light petroleum
(40° to 60°), 2 volumes of diethylamine and 6 to 8 volumes of Labelling. The label states the strength in terms of the
2-phenoxyethanol. Shake and use the supernatant liquid. equivalent amount of acepromazine.
Test solution. Extract a volume containing 20 mg of
acepromazine with two quantities, each of 5 ml, of
dichloromethane and use the combined extracts. Acepromazine Tablets
Reference solution. A 0.2 per cent w/v solution of Acepromazine Maleate Tablets
acepromazine maleate RS in dichloromethane. Acepromazine Tablets contain not less than 92.5 per cent and
Impregnate the dry plate by placing it in a tank containing a not more than 107.5 per cent of the stated amount of
shallow layer of a mixture of 85 volumes of acetone, 10 volumes acepromazine, C19H22N2OS.
of 2-phenoxyethanol and 5 volumes of polyethyleneglycol
300 so that the plate dips about 5 mm below the surface of the Identification
liquid and allow the impregnating solvent to ascend almost to NOTE — Carry out the tests in subdued light.
the top. Use the plate immediately after removing it from the
tank. Apply to the plate 1 µl of each solution. After A. To a quantity of the powdered tablets containing 20 mg of
development, dry the plate in air and examine in ultraviolet acepromazine add 2 ml of water and 3 ml of 2 M sodium
light at 365 nm. The principal spot in the chromatogram hydroxide. Extract with two quantities, each of 5 ml, of
obtained with the test solution corresponds to the spot in the cyclohexane and remove the solvent under reduced pressure.
The residue complies with the following test.

1508
IP 2007 AMITRAZ

Determine by infrared absorption spectrophotometry (2.4.6). Test solution. Shake a quantity of the powdered tablets
Compare the spectrum with that obtained with acepromazine containing 50 mg of acepromazine with 10 ml of
RS or with the reference spectrum of acepromazine. dichloromethane, filter, evaporate to dryness and dissolve
the residue in 5 ml of methanol containing 0.5 per cent v/v of
B. To 5 mg of the residue obtained in test A add 2 ml of sulphuric
strong ammonia solution.
acid; a yellow colour is produced which changes to deep
orange on warming for 2 minutes. Reference solution. Dilute 1 ml of the test solution to 100 ml
with methanol containing 0.5 per cent v/v of strong ammonia
C. Determine by thin-layer chromatography (2.4.17), coating
solution.
the plate with kieselguhr G.
Apply to the plate 10 µl of each solution. After development,
Mobile phase. A mixture of 100 volumes of light petroleum
dry the plate in air and examine in ultraviolet light at 254 nm.
(40° to 60°), 2 volumes of diethylamine and 6 to 8 volumes of
Any secondary spot in the chromatogram obtained with the
2-phenoxyethanol. Shake and use the supernatent liquid.
test solution is not more intense than the spot in the
Test solution. Extract a quantity of powdered tablets containing chromatogram obtained with the reference solution.
20 mg of acepromazine with two quantities, each of 5 ml, of
Other tests. Comply with the tests stated under Tablets.
dichloromethane and use the combined extracts.
Assay. Weigh and powder 20 tablets. Weigh accurately a
Reference solution. A 0.2 per cent w/v solution of
quantity of the powder containing 60 mg of acepromazine,
acepromazine maleate RS in dichloromethane.
add 5 ml of water and extract with three or more quantities,
Impregnate the dry plate by placing it in a tank containing a each of 50 ml, of dichloromethane until the dichloromethane
shallow layer of a mixture of 85 volumes of acetone, 10 volumes extract is colourless. Wash the extracts with the same 10 ml of
of 2-phenoxyethanol and 5 volumes of polyethyleneglycol water and filter through a plug of absorbent cotton previously
300 so that the plate dips about 5 mm below the surface of the moistened with dichloromethane. Evaporate the combined
liquid and allow the impregnating solvent to ascend almost to extracts to dryness, dissolve the residue in 15 ml of acetic
the top. Use the plate immediately after removing it from the anhydride. Titrate with 0.02 M perchloric acid, using crystal
tank. Apply to the plate 1 µl of each solution. After violet solution as indicator. Carry out a blank titration.
development, dry the plate in air and examine in ultraviolet
1 ml of 0.02 M perchloric acid is equivalent to 0.006529 g of
light at 365 nm. The principal spot in the chromatogram
C19H22N2OS.
obtained with the test solution corresponds to the spot in the
chromatogram obtained with the reference solution. A Labelling. The label states the strength in terms of the
secondary spot due to maleic acid is also observed in both equivalent amount of acepromazine.
chromatograms. Spray the plate with ethanolic sulphuric acid
(10 per cent v/v). The spot in the chromatogram obtained
with the test solution corresponds to the spot in the
chromatogram obtained with the reference solution. Amitraz
D. Dissolve a quantity of the powdered tablets containing 25
CH3 H3C
mg of acepromazine as completely as possible in a mixture of
CH3
3 ml of water and 2 ml of 5 M sodium hydroxide and shake
with three quantities, each of 3 ml, of ether. Discard the ether H3C N C N C N CH3
H H
extracts. Add 2 ml of bromine solution to the aqueous solution,
warm in a water-bath for 10 minutes, heat to boiling, cool and C19H23N3 Mol. Wt. 293.41
add 0.25 ml of a solution of 10 mg of resorcinol in 3 ml of
sulphuric acid; a bluish-black colour is produced on heating Amitraz is N,N-di-(2,4-xylyliminomethyl)methylamine.
for 15 minutes in a water-bath. Description. A white to buff powder.

Tests Amitraz contains not less than 95.0 per cent and not more
than 101.5 per cent of C19H23N3, calculated on the anhydrous
Related substances. Determine by thin-layer chromatography basis.
(2.4.17), coating the plate with silica gel GF254.
Solvent mixture. A mixture of 95 volumes of methanol and 5 Identification
volumes of diethylamine. A. Determine by infrared absorption spectrophotometry (2.4.6).
Mobile phase. A mixture of 75 volumes of hexane, 17 volumes Compare the spectrum with that obtained with amitraz RS or
of 2-butanol and 8 volumes of diethylamine. with the reference spectrum of amitraz.

1509
LIQUID AMITRAZ DIP CONCENTRATE IP 2007

B. In the test for Related substances, the principal spot in the Test solution (a). Dissolve 0.8 g of the substance under
chromatogram obtained with test solution (b) corresponds to examination in 100 ml of methyl acetate.
that in the chromatogram obtained with reference solution (a).
Test solution (b). Dissolve 0.8 g of the substance under
C. In the Assay, the principal peak in the chromatogram examination in 100 ml of the internal standard solution.
obtained with the test solution (b) corresponds to the peak in
Reference solution. A 0.8 per cent w/v solution of amitraz RS
the chromatogram obtained with the reference solution.
in the internal standard solution.
Tests Chromatographic system
– a glass column 1.5 m x 4 mm, packed with acid-washed,
Related substances. Determine by thin-layer chromatography silanised diatomaceous support (80 to 100 mesh) coated
(2.4.17), coating the plate with silica gel HF254. with 3 per cent w/w methyl silicone gum or fluid
Mobile phase. A mixture of 50 volumes of cyclohexane, 30 (such as OV-1 or OV-101),
volumes of ethyl acetate and 20 volumes of triethylamine. – temperature:
column. 250°,
Test solution (a). Dissolve 1 g of the substance under
inlet port and detector. 280°,
examination in 10 ml of toluene.
– flow rate. 30 ml per minute of the carrier gas.
Test solution (b). Dissolve 20 mg of the substance under Calculate the content of C19H23N3.
examination in 10 ml of toluene.
Storage. Store in containers which may contain
Reference solution (a). A 0.20 per cent w/v solution of amitraz paraformaldehyde packed in separate sachets as stabiliser.
RS in toluene.
Reference solution (b). A 0.030 per cent w/v of
2,4-dimethylaniline in toluene.
Impregnate the plate to a depth of about 3.5 cm with a solution
Liquid Amitraz Dip Concentrate
prepared by dissolving 35 g of acetamide in 100 ml of methanol, Liquid Amitraz Dip Concentrate contains Amitraz in a suitable
adding 100 ml of triethylamine and diluting to 250 ml with emulsifiable vehicle. It may contain a suitable stabilising agent.
methanol, before standing it in a stream of cold air for about
Liquid Amitraz Dip Concentrate contains not less than 90.0
30 seconds. Immediately apply to the plate, at a level 1 cm
per cent and not more than 110.0 per cent of the stated amount
below the top of the impregnated zone, 2 µl of each solution.
of amitraz, C19H23N3.
After development, dry the plate in air and examine in
ultraviolet light at 254 nm. Any secondary spot in the
Identification
chromatogram obtained with test solution (a) is not more
intense than the spot in the chromatogram obtained with A. In the test for Related substances, the principal spot in the
reference solution (a). Expose the plate to the vapours of chromatogram obtained with test solution (b) corresponds to
hydrochloric acid until the plate smells strongly of acid. that in the chromatogram obtained with reference solution (a).
Expose to the vapours of nitrogen dioxide (prepared by the
B. In the Assay, the principal peak in the chromatogram
action of nitric acid on granulated zinc) for 10 minutes, remove
obtained with the test solution (b) corresponds to the peak in
the excess of nitrogen dioxide with air and spray with a 0.5 per
the chromatogram obtained with reference solution (b).
cent w/v solution of N-(1-naphthyl)ethylenediamine
dihydrochloride in a 50 per cent v/v solution of methanol. Tests
Any secondary spot corresponding to 2,4-dimethylaniline in
the chromatogram obtained with test solution (a) is not more Related substances. Determine by thin-layer chromatography
intense than the corresponding spot in the chromatogram (2.4.17), coating the plate with silica gel HF254.
obtained with reference solution (b).
Mobile phase. A mixture of 50 volumes of cyclohexane, 30
Water (2.3.43). Not more than 0.1 per cent , determined on 5 g volumes of ethyl acetate and 20 volumes of triethylamine.
and using anhydrous pyridine in place of anhydrous Test solution (a). Dilute the dip concentrate with toluene to
methanol. obtain 5.0 per cent w/v of Amitraz.
Sulphated ash (2.3.18). Not more than 0.2 per cent.
Test solution (b). Dilute the dip concentrate with toluene to
Assay. Determine by gas chromatography (2.4.13). obtain 0.2 per cent w/v of Amitraz.
Internal Standard Solution. A 1.0 per cent v/v solution of Reference solution (a). A 0.20 per cent w/v solution of amitraz
squalane in methyl acetate. RS in toluene.

1510
IP 2007 AMITRAZ DIP CONCENTRATE POWDER

Reference solution (b). A 0.030 per cent w/v of Amitraz Dip Cconcentrate Powder
2,4-dimethylaniline in toluene.
Amitraz Dip Concentrate Powder consists of Amitraz mixed
Impregnate the plate to a depth of about 3.5 cm in a solution
with suitable wetting, dispersing and suspending agents. It
prepared by dissolving 35 g of acetamide in 100 ml of methanol,
may contain a suitable stabilising agent.
adding 100 ml of triethylamine and diluting to 250 ml with
methanol, before standing it in a stream of cold air for about Amitraz Dip Concentrate Powder contains not less than 92.0
30 seconds. Immediately apply to the plate, at a level 1 cm per cent and not more than 108.0 per cent of the stated amount
below the top of the impregnated zone, 2 µl of each solution. of amitraz, C19H23N3.
After development, dry the plate in air and examine in
ultraviolet light at 254 nm. Any secondary spot in the Identification
chromatogram obtained with the test solution (a) is not more A. Shake a quantity of the powder containing 0.1 g of Amitraz
intense than the spot in the chromatogram obtained with the with 10 ml of acetone for 5 minutes, filter and evaporate the
reference solution (a). Expose the plate to the vapours of filtrate to dryness. The residue complies with the following
hydrochloric acid until the plate smells strongly of acid. test.
Expose to the vapours of nitrogen dioxide (prepared by the
action of nitric acid on granulated zinc) for 10 minutes, remove Determine by infrared absorption spectrophotometry (2.4.6).
the excess of nitrogen dioxide with air and spray with a 0.5 per Compare the spectrum with that obtained with amitraz RS or
cent w/v solution of N-(1-naphthyl)ethylenediamine with the reference spectrum of amitraz.
dihydrochloride in a 50 per cent v/v solution of methanol. B. In the test for Related substances, the principal spot in the
Any secondary spot corresponding to 2,4-dimethylaniline in chromatogram obtained with test solution (b) corresponds to
the chromatogram obtained with test solution (a) is not more that in the chromatogram obtained with reference solution (a).
intense than the corresponding spot in the chromatogram
C. In the Assay, the principal peak in the chromatogram
obtained with reference solution (b).
obtained with the test solution (b) corresponds to the peak in
Water (2.3.43). Not more than 0.15 per cent w/v, determined in the chromatogram obtained with the reference solution.
5 ml of the dip concentrate and using anhydrous pyridine in
place of anhydrous methanol. Tests
Other tests. Complies with the tests stated under Dip
Related substances. Determine by thin-layer chromatography
Concentrates.
(2.4.17), coating the plate with silica gel HF254.
Assay. Determine by gas chromatography (2.4.13).
Mobile phase. A mixture of 50 volumes of cyclohexane, 30
Internal Standard Solution. A 1.0 per cent v/v solution of volumes of ethyl acetate and 20 volumes of triethylamine.
squalane in methyl acetate. Test solution (a). The supernatant liquid obtained by shaking
Test solution (a). Dissolve an accurately measured volume of a quantity of the powder containing 0.5 g of Amitraz with 10 ml
the dip concentrate containing 80 mg of Amitraz in 10 ml of of toluene for 5 minutes and centrifuging the suspension.
methyl acetate. Test solution (b). The supernatant liquid obtained by shaking
Test solution (b). Dissolve an accurately measured volume of a quantity of the powder containing 20 mg of Amitraz with 10
the dip concentrate containing 80 mg of Amitraz in 10 ml of the ml of toluene for 5 minutes and centrifuging the suspension.
internal standard solution. Reference solution (a). A 0.20 per cent w/v solution of amitraz
Reference solution. A 0.8 per cent w/v solution of amitraz RS RS in toluene.
in the internal standard solution.
Reference solution (b). A 0.030 per cent w/v of
Chromatographic system 2,4-dimethylaniline in toluene.
– a glass column 1.5 m x 4 mm, packed with acid-washed,
Impregnate the plate to a depth of about 3.5 cm in a solution
silanised diatomaceous support (80 to 100 mesh) coated
prepared by dissolving 35 g of acetamide in 100 ml of methanol,
with 3 per cent w/w methyl silicone gum or fluid (such
adding 100 ml of triethylamine and diluting to 250 ml with
as OV-1 or OV-101),
methanol, before standing it in a stream of cold air for about
– temperature:
30 seconds. Immediately apply to the plate, at a level 1 cm
column. 250°,
below the top of the impregnated zone, 2 µl of each solution.
inlet port and detector. 280°,
After development, dry the plate in air and examine in
– flow rate. 30 ml per minute of the carrier gas.
ultraviolet light at 254 nm. Any secondary spot in the
Calculate the content of C19H23N3. chromatogram obtained with the test solution (a) is not more

1511
AMPICILLIN AND CLOXACILLIN INTRAMAMMARY INFUSION (LACTATING COW/BUFFALO) IP 2007

intense than the spot in the chromatogram obtained with the Ampicillin and Cloxacillin Intramammary Infusion (Lactating
reference solution (a). Expose the plate to the vapours of Cow/Buffalo) contains not less than 90.0 per cent and not
hydrochloric acid until the plate smells strongly of acid. more than 110.0 per cent of the stated amount of each of
Expose to the vapours of nitrogen dioxide (prepared by the ampicillin, C16H19N3O4S, and cloxacillin, C19H18ClN3O5S.
action of nitric acid on granulated zinc) for 10 minutes, remove
the excess of nitrogen dioxide with air and spray with a 0.5 per Identification
cent w/v solution of N-(1-naphthyl)ethylenediamine
A. Determine by thin-layer chromatography (2.4.17), coating
dihydrochloride in a 50 per cent v/v solution of methanol.
the plate with silica gel G.
Any secondary spot corresponding to 2,4-dimethylaniline in
the chromatogram obtained with test solution (a) is not more Mobile phase. A mixture of 10 volumes of butyl acetate, 6
intense than the corresponding spot in the chromatogram volumes of glacial acetic acid, 1 volume of 1-butanol and 2
obtained with reference solution (b). volumes of solution A (see below).
Other tests. Complies with the tests stated under Dip Test solution. Extract a quantity of the infusion containing 50
Concentrates. mg of ampicillin with three successive quantities, each of 15
ml, of light petroleum ( 120° to 160°). Discard the extracts,
Assay. Determine by gas chromatography (2.4.13).
wash the residue with 10 ml of ether and dry in a current of air.
Internal Standard Solution. A 1.0 per cent v/v solution of Dissolve the residue in 50 ml of phosphate buffer pH 7.0,
squalane in methyl acetate. shake well, filter and use the filtrate.

Test solution (a). Shake a quantity of the powder containing Reference solution. A 0.12 per cent w/v solution of ampicillin
80 mg of Amitraz with 10 ml of methyl acetate, centrifuge and trihydrate RS in phosphate buffer pH 7.0.
use the supernatant liquid. Impregnate the plate by spraying it with a 0.1 per cent w/v
Test solution (b). Shake a quantity of the powder containing solution of disodium edetate in a 5 per cent w/v solution of
80 mg of Amitraz with 10 ml of the internal standard solution, sodium dihydrogen phosphate (solution A), allow the plate
centrifuge and use the supernatant liquid. to dry in air and heat it at 105° for 1 hour. Apply to the plate
1 µl of each solution. After development, dry the plate in air
Reference solution. A 0.8 per cent w/v solution of amitraz RS and heat at 150° for 10 to 15 minutes and spray with a mixture
in the internal standard solution. of 100 volumes of starch mucilage, 6 volumes of glacial acetic
Chromatographic system acid and 2 volumes of a 1 per cent w/v solution of iodine in a
– a glass column 1.5 m x 4 mm, packed with acid-washed, 4 per cent w/v solution of potassium iodide. The principal
silanised diatomaceous support (80 to 100 mesh) coated spot in the chromatogram obtained with the test solution
with 3 per cent w/w methyl silicone gum or fluid corresponds to the spot in the chromatogram obtained with
(such as OV-1 or OV-101), the reference solution.
– temperature: B. Determine by thin-layer chromatography (2.4.17), coating
column. 250°, the plate with silanised silica gel GF254.
inlet port and detector. 280°,
– flow rate. 30 ml per minute of the carrier gas. Mobile phase. A mixture of 70 volumes of 0.05 M potassium
hydrogen phthalate, 30 volumes of acetone and 1 volume of
Calculate the content of C19H23N3. formic acid that has been adjusted first to pH 6.0 with 5 M
sodium hydroxide and then to pH 9.0 with 0.1 M sodium
hydroxide.
Test solution. Extract a quantity of the infusion containing
Ampicillin and Cloxacillin 130 mg of cloxacillin with three successive quantities, each of
Intramammary Infusion (Lactating 15 ml, of light petroleum ( 120° to 160°). Discard the extracts,
wash the residue with 10 ml of ether and dry in a current of air.
Cow/Buffalo) Dissolve the residue in 50 ml of phosphate buffer pH 7.0,
Ampicillin Sodium and Cloxacillin Sodium Intramammary shake well, filter and use the filtrate.
Infusion (LC/B) Reference solution. A 0.28 per cent w/v solution of cloxacillin
Ampicillin and Cloxacillin Intramammary Infusion (Lactating sodium RS in phosphate buffer pH 7.0.
Cow/Buffalo) is a sterile suspension of Ampicillin Sodium and Apply to the plate 1 µl of each solution. After development,
Cloxacillin Sodium in a suitable vehicle containing suitable dry the plate in air and heat at 150° for 10 to 15 minutes and
suspending agents. spray with a mixture of 100 volumes of starch mucilage, 6

1512
IP 2007 AMPICILLIN AND CLOXACILLIN BENZATHINE INTRAMAMMARY INFUSION (DRY COW/BUFFALO)

volumes of glacial acetic acid and 2 volumes of a 1 per cent out the procedure simultaneously using 2.0 ml of a solution
w/v solution of iodine in a 4 per cent w/v solution of prepared by dissolving 0.14 g of cloxacillin sodium RS in
potassium iodide. The principal spot in the chromatogram 100.0 ml of water.
obtained with the test solution corresponds to the spot in the
Labelling. The label states the quantity of Ampicillin Sodium
chromatogram obtained with the reference solution.
in terms of the equivalent amount of ampicillin and the quantity
C. Extract a quantity containing 50 mg of ampicillin with three of Cloxacillin Sodium in terms of the equivalent amount of
successive quantities, each of 15 ml, of light petroleum ( cloxacillin.
120° to 160°). Discard the extracts, wash the residue with 10
ml of ether and dry the residue at 55°. The residue produces
an intense, persistent yellowish orange colour when
introduced into a non-luminous flame on a platinum wire Ampicillin and Cloxacillin Benzathine
moistened with hydrochloric acid.
Intramammary Infusion (Dry Cow/
Tests Buffalo)
Water (2.3.43). Not more than 1.0 per cent , determined on 1.5 Ampicillin and Cloxacillin Benzathine Intramammary Infusion
g using a mixture of 70 volumes of dichloromethane and 30 (Dry Cow/Buffalo) is a sterile suspension of Ampicillin
volumes of anhydrous methanol as the solvent. Trihydrate and Cloxacillin Benzathine in a suitable vehicle
Other tests. Complies with the tests stated under containing suitable suspending agents.
Intramammary Infusions. Ampicillin and Cloxacillin Benzathine Intramammary Infusion
Assay. Weigh and mix the contents of 10 containers. Weigh (Dry Cow/Buffalo) contains not less than 90.0 per cent and
accurately a quantity of the mixed contents containing 50 mg not more than 110.0 per cent of the stated amounts of ampicillin,
of ampicillin and extract with three successive quantities, each C16H19N3O4S, and cloxacillin, C19H18ClN3O5S.
of 15 ml, of light petroleum ( 120° to 160°) previously
Identification
saturated with ampicillin sodium and cloxacillin sodium.
Discard the extracts, wash the residue with ether previously A. Extract a quantity containing 250 mg of ampicillin with
saturated with ampicillin sodium and cloxacillin sodium, dry in three quantities, each of 15 ml, of light petroleum ( 120° to
a current of air, dissolve in water and dilute to 100.0 ml with 160°). Discard the extracts, wash the residue with 10 ml of
water. Centrifuge and use the clear supernatant liquid (solution ether and dry in a current of air. Shake with 10 ml of
B). dichloromethane and filter. Keep both the residue and the
For ampicillin — Dilute 2.0 ml of solution B to 50.0 ml with filtrate.
buffered cupric sulphate solution pH 5.2, transfer 10.0 ml of Wash the residue with two quantities, each of 5 ml, of
the resulting solution to a stoppered test-tube and heat in a dichloromethane and dry in a vaccum desiccator.
water-bath at 75° for 30 minutes. Cool to room temperature
On the residue determine by infrared absorption
rapidly, dilute to 20.0 ml with buffered cupric sulphate solution
spectrophotometry (2.4.6). Compare the spectrum with that
pH 5.2 and measure the absorbance of the resulting solution
obtained with ampicillin trihydrate RS or with the reference
at the maximum at about 320 nm (2.4.7), using as the blank a
spectrum of ampicillin trihydrate.
solution prepared by diluting 2.0 ml of solution B to 100.0 ml
with buffered cupric sulphate solution pH 5.2. B. Wash the filtrate with two quantities, each of 5 ml, of water,
dry the dichloromethane layer with anhydrous sodium
Calculate the content of C16H19N3O4S in a container of average
sulphate, filter and dilute the filtrate to 20 ml with
content from the absorbance obtained by carrying out the
dichloromethane.
procedure simultaneously using 2.0 ml of a solution prepared
by dissolving 60 mg of ampicillin trihydrate RS in 100.0 ml of On the filtrate determine by infrared absorption
water, diluting to 50.0 ml with buffered cupric sulphate solution spectrophotometry (2.4.6). Compare the spectrum with that
pH 5.2 and beginning at the words “transfer 10.0 ml.....” . obtained with cloxacillin benzathine RS or with the reference
spectrum of cloxacillin benzathine.
For cloxacillin — Dilute 2.0 ml of solution B to 100.0 ml with
1 M hydrochloric acid. Measure the absorbance of the
Tests
resulting solution at 20° after exactly 12 minutes at the maximum
at about 350 nm (2.4.7), using 1 M hydrochloric acid as the Water (2.3.43). Not more than 3.0 per cent, determined on 1.5
blank. Calculate the content of C19H18ClN3O5S in a container g using a mixture of 70 volumes of dichloromethane and 30
of average content from the absorbance obtained by carrying volumes of anhydrous methanol as the solvent.

1513
AMPICILLIN VETERINORY ORAL POWDER IP 2007

Other tests. Complies with the tests stated under Description. A fine granular powder.
Intramammary Infusions.
Assay. Weigh and mix the contents of 10 containers. Weigh Identification
accurately a quantity of the mixed contents containing 60 mg A. Determine by thin-layer chromatography (2.4.17), coating
of ampicillin and extract with three quantities, each of 15 ml, of the plate with silica gel G.
light petroleum ( 120° to 160°) previously saturated with
ampicillin trihydrate and cloxacillin benzathine. Discard the NOTE — Prepare the solutions immediately before use.
extracts, wash the residue with ether previously saturated Mobile phase. A mixture of 10 volumes of butyl acetate, 6
with ampicillin trihydrate and cloxacillin benzathine, dry in volumes of glacial acetic acid, 2 volumes of a 0.1 per cent w/
a current of air, dissolve in 50 ml of methanol and dilute to v solution of disodium edetate in mixed phosphate buffer pH
100.0 ml with water. Centrifuge and use the clear supernatant 4.0 and 1 volume of 1-butanol.
liquid (solution A).
Test solution. Shake a quantity of the powder containing 0.1 g
For ampicillin — Dilute 2.0 ml of solution A to 50.0 ml with of ampicillin with 50 ml of phosphate buffer pH 7.0 for 15
buffered cupric sulphate solution pH 5.2, transfer 10.0 ml to a minutes, filter and use the filtrate.
stoppered test-tube and heat in a water-bath at 75° for 30
minutes. Cool to room temperature rapidly, dilute to 20.0 ml Reference solution. A 0.2 per cent w/v solution of ampicillin
with buffered cupric sulphate solution pH 5.2 and measure trihydrate RS in phosphate buffer pH 7.0.
the absorbance of the resulting solution at the maximum at Impregnate the dry plate by placing it in a tank containing a
about 320 nm (2.4.7), using as the blank the unheated buffered shallow layer of a 0.1 per cent w/v solution of disodium edetate
solution of the infusion. in mixed phosphate buffer pH 4.0, allowing the solvent to
Calculate the content of C16H19N3O4S in a container of average ascend to the top, removing the plate from the tank and
content from the absorbance obtained by carrying out the allowing the solvent to evaporate. Use the plate with the flow
procedure simultaneously using 2.0 ml of a solution prepared of the mobile phase in the direction in which impregnation
by dissolving 70 mg of ampicillin trihydrate RS in 100.0 ml of was carried out. Before use heat the plate at 100° for 1 hour
a 50 per cent v/v solution of methanol, diluting to 50.0 ml with and allow to cool. Apply to the plate 1 µl of each solution.
buffered cupric sulphate solution pH 5.2, and beginning at After development, dry the plate in air and spray with a mixture
the words “transfer 10.0 ml.....” . of 100 volumes of a 1 per cent w/v solution of starch, 6 volumes
of glacial acetic acid and 2 volumes of a 1 per cent w/v
For cloxacillin — Dilute 2.0 ml of solution A to 100.0 ml with solution of iodine in a 4 per cent w/v solution of potassium
1 M hydrochloric acid and measure the absorbance of the iodide. The principal spot in the chromatogram obtained with
resulting solution at 20° after exactly 12 minutes at the maximum the test solution corresponds to the spot in the chromatogram
at about 350 nm, (2.4.7), using 1 M hydrochloric acid as the obtained with the reference solution.
blank. Calculate the content of C19H18ClN3O5S in a container
of average content from the absorbance obtained by carrying B. To a quantity of the powder containing 10 mg of ampicillin
out the procedure simultaneously using 2.0 ml of a solution add sufficient water to produce 10 ml, shake for 15 minutes
prepared by dissolving 0.165 g of cloxacillin benzathine RS and filter. Place 0.1 ml of a 0.1 per cent w/v solution of ninhydrin
in 100.0 ml of a 50 per cent v/v solution of methanol. on a filter paper, dry at 105°, superimpose 0.1 ml of the solution
of the preparation under examination, heat for 5 minutes at
Labelling. The label states the strength of Ampicillin 105° and allow to cool; a mauve colour is produced.
Trihydrate in terms of the equivalent amount of ampicillin and
that of Cloxacillin Benzathine in terms of the equivalent amount C. Suspend a quantity of the powder containing 10 mg of
of cloxacillin. ampicillin in 1 ml of water and add 2 ml of a mixture of 2 ml of
potassium cupri-tartrate solution and 6 ml of water; a
magenta-violet colour is immediately produced.
Ampicillin Veterinary Oral Powder Tests
Ampicillin TrihydrateVeterinary Oral Powder
Uniformity of weight. When supplied in containers intended
Ampicillin Veterinary Oral Powder is a mixture of Ampicillin for use on one occasion, complies with the test for Uniformity
Trihydrate and Lactose or other suitable diluent. of weight described under Parenteral Preparations (Powders
Ampicillin Veterinary Oral Powder contains not less than 90.0 for Injection).
per cent and not more than 110.0 per cent of the stated amount Other tests. Complies with the tests stated under Veterinary
of ampicillin, C16H19N3O4S. Oral Powders.

1514
IP 2007 AMPROLIUM HYDROCHLORIDE AND ETHOPABATE PREMIX

Assay. Weigh accurately a quantity of the powder containing C. To 1 mg add 5 ml of naphthalenediol reagent; a deep violet
0.1 g of ampicillin, add sufficient water to produce 100.0 ml, colour is produced.
shake for 15 minutes and filter. Dilute 2.0 ml to 100.0 ml with
D. Gives the reactions of chlorides (2.3.1).
buffered cupric sulphate solution pH 5.2, transfer 10.0 ml of
the resulting solution to a stoppered test-tube and heat in a Tests
water-bath at 75° for 30 minutes. Cool to room temperature
rapidly, adjust the volume if necessary to 10.0 ml with water Picoline. Dissolve 1.5 g in 30 ml of water in a distillation flask,
and measure the absorbance at the maximum at about 320 nm add 20 ml of a saturated solution of potassium carbonate
(2.4.7), using as the blank the unheated buffered solution of sesquihydrate, connect the flask to a ground-glass aerator
the powder under examination. Calculate the content of extending to the bottom of a l00-ml graduated cylinder
C16H19N3O4S from the absorbance obtained by carrying out containing 50 ml of 0.05 M hydrochloric acid and pass air,
the procedure simultaneously using 0.12 g of ampicillin which has previously been passed through sulphuric acid
trihydrate RS. When the powder is supplied in containers and glass wool, through the system for 60 minutes. To 5 ml of
intended for use on more than one occasion, take the quantity the hydrochloric acid solution add sufficient 0.05 M
of powder without previous mixing. hydrochloric acid to produce 200 ml. Absorbance of the
resulting solution at about 262 nm (2.4.7), not more than 0.52.
Storage. Store protected from moisture, at a temperature not
exceeding 30°. Sulphated ash (2.3.18). Not more than 0.1 per cent.
Labelling. The label states the strength in terms of the Loss on drying (2.4.19). Not more than 1.0 per cent, determined
equivalent concentration of ampicillin. on 1.0 g by drying to constant weight at 100° at a pressure not
exceeding 0.7 kPa.
Assay. Weigh accurately about 0.3 g, dissolve in 20 ml of
anhydrous glacial acetic acid, add 10 ml of mercuric acetate
Amprolium Hydrochloride solution. Titrate with 0.1 M perchloric acid, using
1-naphtholbenzein solution as indicator. Carry out a blank
N CH3 titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.01577 g of
N
Cl , HCl C14H19ClN4,HCl.
H3C N NH2

Amprolium Hydrochloride and


C14H19ClN4,HCl Mol. Wt. 315.2
Ethopabate Premix
Amprolium Hydrochloride is hydrochloride salt of 1-[(4- Amprolium Hydrochloride and Ethopabate Premix contains
amino-2-propyl-5-pyrimidinyl)methyl]-2-methylpyridinium Amprolium Hydrochloride and Ethopabate.
chloride. Amprolium Hydrochloride and Ethopabate Premix contains
Amprolium Hydrochloride contains not less than 97.5 per cent not less than 90.0 per cent and not more than 110.0 per cent of
and not more than 101.0 per cent of C14H19ClN4,HCl, calculated the stated amounts of amprolium hydrochloride,
on the dried basis. C14H19ClN4,HCl and of ethopabate, C12H15NO4.

Description. A white or almost white powder. Identification


Identification A. Shake a quantity containing 20 mg of Amprolium
Hydrochloride with 90 ml of methanol and filter. Add 5 ml of
A. Determine by infrared absorption spectrophotometry (2.4.6). the filtrate to 5 ml of naphthalenediol reagent; a deep violet
Compare the spectrum with that obtained with amprolium colour is produced.
hydrochloride RS or with the reference spectrum of amprolium
hydrochloride. B. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel GF254.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
0.002 per cent w/v solution in 0.1 M hydrochloric acid exhibits NOTE — Prepare the solution immediately before use.
maxima at about 246 nm and 262 nm; absorbance at about 246 Mobile phase. A mixture of 90 volumes of dichloromethane
nm, about 0.84 and at about 262 nm, about 0.80. and 10 volumes of methanol.

1515
AMPROLIUM, ETHOPABATE AND SULPHAQUINOXALINE PREMIX IP 2007

Test solution. Shake continuously for 10 minutes a quantity at the words “add 2.5 ml of 2 M hydrochloric acid......”.
containing 10 mg of Ethopabate with 25 ml of acetone that has Calculate the content of C12H15NO4 from the absorbance
been warmed to 50°, filter and use the filtrate. obtained by carrying out the procedure simultaneously, using
Reference solution. A 0.04 per cent w/v solution of ethopabate 10.0 ml of a 0.006 per cent w/v solution of ethopabate RS in
RS in acetone. methanol and beginning at the words “add 10 ml of 1 M sodium
hydroxide and evaporate to dryness.....”.
Apply to the plate 2 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with the test
solution corresponds to the spot in the chromatogram Amprolium, Ethopabate and
obtained with the reference solution. Sulphaquinoxaline Premix
Tests Amprolium Hydrochloride, Ethopabate and
pH (2.4.24). 2.5 to 4.0, determined in a 25 per cent w/v slurry in Sulphaquinoxaline Premix
carbon dioxide-free water. Amprolium, Ethopabate and Sulphaquinoxaline Premix
Other tests. Complies with the tests stated under Premixes. contains Amprolium Hydrochloride, Ethopabate and
Sulphaquinoxaline.
Assay. For amprolium hydrochloride — Weigh accurately a
quantity containing 50 mg of Amprolium Hydrochloride, shake Amprolium, Ethopabate and Sulphaquinoxaline Premix
continuously for 20 minutes with 100.0 ml of a mixture of 2 contains not less than 90.0 per cent and not more than 110.0
volumes of methanol and 1 volume of water and filter. Dilute per cent of the stated amounts of amprolium hydrochloride,
5.0 ml of the filtrate to 100.0 ml with the methanol-water mixture. C 14 H 19 ClN 4 ,HCl, of ethopabate, C 12 H 15 NO 4 , and of
To 4.0 ml of the resulting solution add 10.0 ml of sulphaquinoxaline, C14H12N4O2S.
naphthalenediol reagent, allow to stand for 20 minutes and
Identification
measure the absorbance of the resulting solution at the
maximum at about 520 nm (2.4.7), using as the blank a solution A. Shake a quantity containing 20 mg of Amprolium
obtained by mixing 4.0 ml of a mixture of 2 volumes of methanol Hydrochloride with 90 ml of methanol and filter. Add 5 ml of
and 1 volume of water with 10.0 ml of naphthalenediol reagent the filtrate to 5 ml of naphthalenediol reagent; a deep violet
and allowing to stand for 20 minutes. Calculate the content of colour is produced.
C14H19ClN4,HCl from the absorbance obtained by carrying out
the procedure simultaneously, using 4.0 ml of a 0.0025 per B. Determine by thin-layer chromatography (2.4.17), coating
cent w/v solution of amprolium hydrochloride RS in a mixture the plate with silica gel GF254.
of 2 volumes of methanol and 1 volume of water and beginning NOTE — Prepare the solution immediately before use.
at the words, “To 4.0 ml of the resulting solution add 10.0 ml
Mobile phase. A mixture of 90 volumes of dichloromethane
of.....” .
and 10 volumes of methanol.
For ethopabate — Weigh accurately a quantity containing 6
Test solution. Shake continuously for 10 minutes a quantity
mg of Ethopabate, add 75 ml of methanol, shake continuously
containing 10 mg of Ethopabate with 25 ml of acetone that has
for 20 minutes, dilute to 100.0 ml with methanol and filter. To
been warmed to 50°, filter and use the filtrate.
10.0 ml of the filtrate add 10 ml of 1 M sodium hydroxide and
evaporate to dryness. Dissolve the residue in 10.0 ml of water, Reference solution (a). A 0.04 per cent w/v solution of
heat on a water-bath for 15 minutes, add 10 ml of 2 M ethopabate RS in acetone.
hydrochloric acid, dilute to 100.0 ml with water and filter. To Reference solution (b). A 0.4 per cent w/v solution of
25.0 ml of the filtrate, add 2.5 ml of 2 M hydrochloric acid and sulphaquinoxaline RS in acetone.
5 ml of a 0.1 per cent w/v solution of sodium nitrite prepared
immediately before use. Allow to stand for 3 minutes and add Apply to the plate 2 µl of each solution. After development,
2.0 ml of a freshly prepared 0.5 per cent w/v solution of dry the plate in air and examine in ultraviolet light at 254 nm.
ammonium sulphamate. Allow to stand for 2 minutes, add 5.0 The principal spot in the chromatogram obtained with the test
ml of a freshly prepared 0.1 per cent w/v solution of solution corresponds to the spot in the chromatogram
N-(1-naphthyl)ethylenediamine dihydrochloride, allow to obtained with reference solutions (a) and (b).
stand for 10 minutes and dilute to 50.0 ml with water. Measure
Tests
the absorbance of the resulting solution at the maximum at
about 540 nm (2.4.7), using as the blank a solution obtained pH (2.4.24). 2.5 to 4.0, determined in a 25 per cent w/v slurry in
by repeating the procedure with 25 ml of water and beginning carbon dioxide-free water.

1516
IP 2007 CALCIUM BOROGLUCONATE INJECTION

Other tests. Complies with the tests stated under Premixes. a freshly prepared 0.5 per cent w/v solution of ammonium
sulphamate and allow to stand for 2 minutes. Add 5.0 ml of a
Assay. For amprolium hydrochloride — Weigh accurately a
freshly prepared 0.1 per cent w/v solution of
quantity containing 50 mg of Amprolium Hydrochloride, shake
N-(1-naphthyl)ethylenediamine dihydrochloride, allow to
continuously for 20 minutes with 100.0 ml of a mixture of 2
stand for 10 minutes and dilute to 50.0 ml with water. Measure
volumes of methanol and 1 volume of water and filter. Dilute
the absorbance of the resulting solution at the maximum at
5.0 ml of the filtrate to 100.0 ml with the methanol-water mixture.
about 540 nm (2.4.7), using as the blank the solution obtained
To 4.0 ml of the resulting solution add 10.0 ml of
by repeating the procedure with 10 ml of water and beginning
naphthalenediol reagent, allow to stand for 20 minutes and
at the words “add 2.5 ml of 2 M hydrochloric acid......”.
measure the absorbance of the resulting solution at the
Calculate the content of C14H12N4O2S from the absorbance
maximum at about 520 nm (2.4.7), using as the blank a solution
obtained by carrying out the procedure simultaneously, using
obtained by mixing 4.0 ml of a mixture of 2 volumes of methanol
10.0 ml of a 0.0008 per cent w/v solution of sulphaquinoxaline
and 1 volume of water with 10.0 ml of naphthalenediol reagent
RS in 0.001 M sodium hydroxide and beginning at the words
and allowing to stand for 20 minutes. Calculate the content of
“To 10.0 ml of the diluted solution add 2.5 ml of 2 M
C14H19ClN4,HCl from the absorbance obtained by carrying out
hydrochloric acid.....”.
the procedure simultaneously, using 4.0 ml of a 0.0025 per
cent w/v solution of amprolium hydrochloride RS in a mixture
of 2 volumes of methanol and 1 volume of water and beginning
at the words, “To 4.0 ml of the resulting solution add 10.0 ml Calcium Borogluconate Injection
of.....” . Calcium Borogluconate Injection is a sterile solution of Calcium
For ethopabate — Weigh accurately a quantity containing 6 Gluconate and Boric Acid in Water for Injections. The solution
mg of Ethopabate, add 75 ml of methanol, shake continuously may contain up to 0.2 per cent w/v of Chlorocresol.
for 20 minutes, dilute to 100.0 ml with methanol and filter. To Calcium Borogluconate Injection contains not less than 95.0
10.0 ml of the filtrate add 10 ml of 1 M sodium hydroxide and per cent and not more than 105.0 per cent of the stated amount
evaporate to dryness. Dissolve the residue in 10.0 ml of water, of calcium, Ca, and boric acid, H3BO3 equiivalent to not more
heat on a water-bath for 15 minutes, add 10 ml of 2 M than 2.3 times the stated content of calcium.
hydrochloric acid, dilute to 100.0 ml with water and filter. To
25.0 ml of the filtrate, add 2.5 ml of 2 M hydrochloric acid and Identification
5 ml of a 0.1 per cent w/v solution of sodium nitrite prepared
immediately before use. Allow to stand for 3 minutes and add A. Dilute 1 ml with sufficient water to produce a solution
2.0 ml of a freshly prepared 0.5 per cent w/v solution of containing about 0.75 per cent w/v of calcium and add 0.05 ml
ammonium sulphamate. Allow to stand for 2 minutes, add 5.0 of ferric chloride test solution; an intense yellow or yellowish
ml of a freshly prepared 0.1 per cent w/v solution of green colour is produced.
N-(1-naphthyl)ethylenediamine dihydrochloride, allow to B. Gives the reactions of calcium salts (2.3.1).
stand for 10 minutes and dilute to 50.0 ml with water. Measure
the absorbance of the resulting solution at the maximum at C. To 1 ml add 0.15 ml of sulphuric acid and 5 ml of methanol
about 540 nm (2.4.7), using as the blank a solution obtained and ignite; the mixture burns with a flame tinged with green.
by repeating the procedure with 25 ml of water and beginning
Tests
at the words “add 2.5 ml of 2 M hydrochloric acid......”.
Calculate the content of C12H15NO4 from the absorbance pH (2.4.24). 3.0 to 4.0, determined in a solution diluted if
obtained by carrying out the procedure simultaneously, using necessary with carbon dioxide-free water to produce a
10.0 ml of a 0.006 per cent w/v solution of ethopabate RS in solution containing 1.5 per cent w/v of calcium.
methanol and beginning at the words “add 10 ml of 1 M sodium
Other tests. Complies with the tests stated under Parenteral
hydroxide and evaporate to dryness.....”.
Preparations (Injections).
For sulphaquinoxaline — Weigh accurately a quantity
containing 40 mg of Sulphaquinoxaline, shake continuously Assay. For calcium — Dilute an accurately measured volume
for 10 minutes with a mixture of 75 ml of water and 4 ml of 2 M containing 45 mg of calcium to about 50 ml with water. Titrate
sodium hydroxide, dilute to 250.0 ml with water and centrifuge. with 0.05 M disodium edetate to within a few ml of the expected
To 10.0 ml of the supernatant liquid add 5 ml of 2 M end-point, add 4 ml of a 40 per cent w/v solution of sodium
hydrochloric acid and dilute to 200.0 ml with water. To 10.0 ml hydroxide and 10 mg of calcon mixture and continue the
of the diluted solution add 2.5 ml of 2 M hydrochloric acid titration until the colour changes from pink to blue.
and 5 ml of a freshly prepared 0.1 per cent w/v solution of 1 ml of 0.05 M disodium edetate is equivalent to 0.002004 g of
sodium nitrite and allow to stand for 3 minutes. Add 5.0 ml of Ca.

1517
CALCIUM MAGNESIUM BOROGLUCONATE INJECTION IP 2007

For boric acid — Dilute an accurately measured volume Other tests. Complies with the tests stated under Parenteral
containing 0.1 g of Boric Acid to 50 ml with water, add 3 g of Preparations (Injections).
mannitol and titrate with 0.1 M sodium hydroxide using Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
phenolphthalein solution as indicator. per ml.
1 ml of 0.1 M sodium hydroxide is equivalent to 0.006183 g of Sterility (2.2.11). Complies with the test for sterility.
H3BO3.
Assay. For calcium — Dilute a volume containing 45 mg of
Storage. Store protected from light, at a temperature not calcium to about 50 ml with water. Add 1 ml of 1 M sodium
exceeding 30°. hydroxide solution. Titrate with 0.05 M disodium edetate to
Labelling. The label states (1) the strength in terms of the within a few ml of the expected end-point, add 5 ml of strong
equivalent amount of calcium in a suitable dose-volume; (2) ammonia-ammonium chloride solution and 10 mg of calcon
the proportion of boric acid present; (3) the proportion of mixture as indicator and continue the titration until the colour
chlorocresol, if present. changes from pink to blue. Calculate the volume of 0.05 M
disodium edetate consumed by substracting the volume of
0.05 M disodium edetate consumed in the assay for
Calcium Magnesium Borogluconate magnesium.
Injection 1 ml of 0.05 M disodium edetate is equivalent to 0.002004 g of
Calcium Magnesium Borogluconate Injection is a sterile Ca.
solution of Calcium Gluconate, Boric Acid, Magnesium For magnesium — Dilute a volume containing 10 mg of
Hypophosphite and Dextrose in Water for Injections. It may magnesium to about 50 ml with water. Add 1 g of ammonium
contain upto 0.2 per cent w/v of Chlorocresol. chloride and 1 g of ammonium oxalate. Neutralise to litmus
Calcium Magnesium Borogluconate Injection contains not paper with dilute ammonia solution and add 5 ml in excess.
less than 95.0 per cent and not more than 105.0 per cent of the Boil for 5 minutes and allow to stand for 1 hour. Filter and
stated amounts of calcium, Ca, of magnesium, calculated as wash the residue with hot water. Collect the filtrate and
magnesium hypophosphite, Mg(H2 PO2 )2 ,6H2O, and of washings and add 5 ml of strong ammonia-ammonium
dextrose, C6H12O6, and the content of boric acid, H3BO3, is not chloride solution. Titrate with 0.05 M disodium edetate using
more than 2.3 times the stated content of calcium. eriochrome black T mixture as indicator.
1 ml of disodium edetate is equivalent to 0.001216 g of
Identification magnesium or 0.0107894 g of magnesium hypophosphite,
A. Dilute 1 ml with sufficient water to produce a solution Mg(H2PO2)2,6H2O.
containing about 0.75 per cent w/v of calcium and add 0.05 ml For boric acid — Dilute a volume containing 0.1 g of boric
of ferric chloride test solution; an intense yellow or yellowish acid to 50 ml with water, add 3 g of mannitol and titrate with
green colour is produced. 0.1 M sodium hydroxide using phenolphthalein solution as
B. Gives the reactions of calcium salts (2.3.1). indicator.
C. Gives the reactions of magnesium salts (2.3.1). 1 ml of 0.1 M sodium hydroxide is equivalent to 0.006183 g of
H3BO3.
D. To 1 ml add 5 ml of water, neutralise to pH 7.0 with dilute
ammonia solution and add 5 ml of silver nitrate solution. A For dextrose — Dilute a volume containing 200 mg of dextrose
yellow precipitate is produced which does not change colour to 50 ml with water; add 30 ml of 0.1 M iodine solution and 10
on boiling but dissolves on addition of dilute ammonia ml of a 5 per cent w/v solution of sodium carbonate and allow
solution. to stand for 20 minutes. Add 15 ml of 1 M hydrochloric acid
and titrate the excess of iodine with 0.1 M sodium thiosulphate
E. To 1 ml add 0.15 ml of sulphuric acid and 5 ml of methanol solution using starch solution as indicator. Carry out a blank
and ignite; the mixture burns with a flame tinged with green. titration.
F. To 1 ml add 2 ml of 2 M sodium hydroxide solution and 0.05 1 ml of 0.1 M iodine solution is equivalent to 0.009008 g of
ml of copper sulphate solution. The solution is blue and clear. dextrose, C6H12O6.
Heat to boiling. A copious red precipitate is produced.
Storage. Store protected from light.
Tests Labelling. The label states (1) the strength in terms of the
pH (2.4.24). 3.0 to 4.0, determined in a solution diluted, if equivalent amount of calcium and magnesium in a suitable
necessary, with carbon dioxide-free water so as to contain dose-volume; (2) the proportion of boric acid to calcium; (3)
1.5 per cent w/v of calcium. the percentage of any added stabilising agent.

1518
IP 2007 CHLORAMPHENICOL INJECTION

Chloral Hydrate Chloramphenicol Injection


Chloramphenicol Injection is a sterile suspension of
HO Cl Chloramphenicol in Water for Injections containing suitable
Cl suspending and stabilising agents.
HO Cl
Chloramphenicol Injection contains not less than 95.0 per cent
and not more than 105.0 per cent of the stated amount of
C2H3Cl3O2 Mol. Wt. 165.4 chloramphenicol, C11H12Cl2N2O5.
Chloral Hydrate is 2,2,2-trichloro-1,1-ethane-diol.
Identification
Description. A colourless, transparent crystals; odour,
Centrifuge a volume containing 0.15 g of Chloramphenicol;
pungent.
wash the residue with water and dry over self-indicating silica
Chloral Hydrate contains not less than 98.5 per cent and not gel and then for 1 hour at 105°. The dried residue complies
more than 101.0 per cent of the stated amount of chloral with the following tests.
hydrate, C2H3Cl302.
A. Wash 75 mg of the residue with two quantities, each of 10
ml, of light petroleum ( 60° to 80°) and allow to dry. The
Identification residue complies with the following test.
A. To 10 ml of a 10 per cent w/v solution in carbon dioxide-free Determine by infrared absorption spectrophotometry (2.4.6).
water (solution A) add 2 ml of 2 M sodium hydroxide. The Compare the spectrum with that obtained with chloramphenicol
mixture becomes cloudy and when heated gives an odour of RS or with the reference spectrum of chloramphenicol.
chloroform.
B. In the test for Related substances, the principal spot in the
B. To 1 ml of solution A add 2 ml of sodium sulphide solution; chromatogram obtained with 1 µl of test solution corresponds
a yellow colour develops which quickly turns reddish brown to that in the chromatogram obtained with reference solution
and may yield a red precipitate on standing. (a).
C. To 5 ml of a 0.1 per cent w/v solution add a few drops of
Tests silver nitrate solution; no precipitate is produced. Heat about
pH (2.4.24). 3.5 to 5.5, determined in solution A. 50 mg with 3 ml of ethanolic potassium hydroxide solution
on a water-bath for 15 minutes, add 15 mg of decolorising
Appearance of solution. Solution A is clear (2.4.1) and charcoal, shake and filter. The filtrate gives the reactions of
colourless (2.4.1). chlorides (2.3.1).
Heavy metals (2.3.13). 12 ml of a 5 per cent w/v solution in
Tests
water, complies with the limit test for heavy metals, Method D
(20 ppm). pH (2.4.24). 3.5 to 6.5.
Chlorides (2.3.12). Dissolve 2.5 g in 15 ml with water; the Consistence. Chloramphenicol Injection containing 150 mg
resulting solution complies with the limit test for chlorides per ml passes readily through a 23G hypodermic needle.
(100 ppm). 2-Amino-1-(4-nitrophenyl)propane-1,3-diol Determine by
Chloral alcoholate. Warm 1 g with 10 ml of 2 M sodium liquid chromatography (2.4.14).
hydroxide, filter the upper layer and add 0.05 M iodine Test solution. Dilute the injection with sufficient of the mobile
dropwise until a yellow colour is produced. Set aside for 1 phase to produce a solution containing 0.030 per cent w/v of
hour; no yellow precipitate is produced and no smell of Chloramphenicol.
iodoform is perceptible. Reference solution. A 0.00225 per cent w/v of
Assay. Weigh accurately about 4 g, dissolve in 10 ml of water 2-amino-1-(4-nitrophenyl) propane-1,3-diol RS in the mobile
and add 40 ml of 1 M sodium hydroxide. Allow the mixture to phase.
stand for exactly 2 minutes and titrate the residual alkali Chromatographic system
immediately with 0.5 M sulphuric acid using phenolphthalein – a stainless steel column 10 cm x 4.6 mm, packed with
solution as indicator. octadecylsilane bonded to porous silica (5 µm),
1 ml of 1 M sodium hydroxide is equivalent to 0.1654 g of – mobile phase: a filtered and degassed mixture of 85
C2H3Cl3O2. volumes of 0.012 M sodium pentane- sulphonate, 15
volumes of acetonitrile and 1 volume of glacial acetic
Storage. Store protected from moisture. acid,

1519
CHLORTETRACYCLINE HYDROCHLORIDE IP 2007

– flow rate. 2 ml per minute, Chlortetracycline Hydrochloride is [4S-


– spectrophotometer set at 272 nm, (4α,4aα,5aα,6β,12aα)]-7-chloro-4- dimethylamino-
– a 20 µl loop injector. 1,4,4a,5,5a,6,11,12a-octahydro-3,6,10,12,12a-pentahydroxy-6-
Inject alternatively the test solution and the reference solution. methyl-1,11-dioxo-2-naphthacenecarboxamide
In the chromatogram obtained with test solution the area of hydrochloride.
any peak corresponding to 2-amino-1-(4- nitrophenyl)- Chlortetracycline Hydrochloride contains not less than 89.5
propane-1,3-diol is not more than the area of the peak obtained per cent of chlortetracycline hydrochloride and the sum of the
with the reference solution. contents of chlortetracycline hydrochloride and tetracycline
Related substances. Determine by thin-layer chromatography hydrochloride is not less than 94.5 per cent and not more than
(2.4.17), coating the plate with silica gel GF254. 100.5 per cent, calculated on the anhydrous basis.
Mobile phase. A mixture of 90 volumes of dichloromethane, Description. Yellow powder.
10 volumes of methanol and 1 volume of water.
Test solution. A 1.0 per cent w/v solution of the dried residue
Identification
obtained in the test for Identification in acetone. A. Determine by thin-layer chromatography (2.4.17), coating
Reference solution (a). A 1.0 per cent w/v solution of the plate with silica gel H.
chloramphenicol RS in acetone. NOTE — Use freshly prepared solution.
Reference solution (b). Dilute 1 ml of reference solution (a) to
Mobile phase. A mixture of 59 volumes of dichloromethane,
200 ml with acetone.
35 volumes of methanol and 6 volumes of water.
Apply to the plate 1 µl and 20 µl of the test solution, 1 µl of
Test solution. Dissolve 50 mg of the substance under
reference solution (a) and 20 µl of reference solution (b). After
examination in 100 ml of methanol.
development, dry the plate in air and examine in ultraviolet
light at 254 nm. Any secondary spot in the chromatogram Reference solution (a). A 0.05 per cent w/v of
obtained with the test solution is not more intense than the chlortetracycline hydrochloride RS in methanol.
spot in the chromatogram obtained with reference solution (b).
Reference solution (b). A solution containing 0.05 per cent w/
Other tests. Complies with the tests stated under Parenteral v each of chlortetracycline hydrochloride RS, tetracycline
Preparations (Injections). hydrochloride RS and metacycline hydrochloride RS in
Assay. To an accurately measured volume containing 0.75 g methanol.
of Chloramphenicol add sufficient water to produce 1000.0 ml Adjust the pH of a 10 per cent w/v solution of disodium edetate
and shake until a clear solution is obtained. Dilute 5.0 ml of to 8.0 with 10 M sodium hydroxide and spray this solution
this solution to 200.0 ml with water and measure the absorbance evenly on the plate (about 10 ml for a plate of 100 mm x 200 mm
of the resulting solution at the maximum at about 278 nm (2.4.7). size). Allow the plate to dry in a horizontal position for at least
Calculate the content of C11H12Cl2N2O5 in the injection taking 1 hour. Dry the plate in an oven at 100° for 1 hour before use.
297 as the specific absorbance at 278 nm. Apply to the plate 1 µl of each solution. After development,
Storage. Store protected from light. Do not freeze. dry the plate in air and examine in ultraviolet light at 365 nm.
Labelling. The label states (1) the name of any added The principal spot in the chromatogram obtained with the test
suspending agent; (2) that the injection is for intramuscular solution corresponds to that in the chromatogram obtained
injection only; (3) the date after which the contents are not with reference solution (a). The test is not valid unless the
intended to be used. chromatogram obtained with reference solution (b) shows
three clearly separated spots.
B. To about 2 mg add 5 ml of sulphuric acid; a deep blue
Chlortetracyline Hydrochloride colour develops which becomes bluish green. Add the
solution to 2.5 ml of water; the colour changes to brownish.
OH O OH O O
HO C. Gives reaction A of chlorides (2.3.1).
NH2 , HCl
Tests
H OH
H pH (2.4.24). 2.3 to 3.3, determined in a 1 per cent w/v solution
Cl H3C OH N(CH3)2
in carbon dioxide-free water prepared by slight heating, if
C22H23ClN2O8,HCl Mol. Wt. 515.3 necessary.

1520
IP 2007 CHLORTETRACYCLINE HYDROCHLORIDE

Specific optical rotation (2.4.22). –235° to –250°, determined Reference solution (e). Mix 5 ml of reference solution (b) and
at 20° in a 0.25 per cent w/v solution in water, calculated on 5 ml of reference solution (c) and dilute to 50 ml with 0.01 M
the anhydrous basis. hydrochloric acid.
Light absorption. When examined in the range 430 nm to 460 Reference solution (F). Dilute 1 ml of reference solution (c) to
nm of a 0.5 per cent w/v solution in water is not greater than 20 ml with 0.01 M hydrochloric acid and dilute 2.5 ml of this
0.40. solution to 100 ml with 0.01 M hydrochloric acid.

Related substances. Carry out the method described under Chromatographic system
Assay injecting test solution, reference solutions (e) and (f). – a stainless steel column 25 cm x 4.6 mm, packed with
The test is not valid unless the peak in the chromatogram octylsilyl groups (5 µm),
obtained with reference solution (f) is properly integrated. In – column temperature 35°,
the chromatogram obtained with test solution the area of the – mobile phase: a filtered and degassed mixture of 450 ml
peak corresponding to 4-epichlortetracycline is not more than of dimethyl sulphoxide, 50 ml of 1 M perchloric acid
the area of the peak corresponding to 4-epichlortetracycline and 500 ml of water,
– flow rate. 1 ml per minute,
in the chromatogram obtained with reference solution (e) (4
– spectrophotometer set at 280 nm,
per cent) and the total area of any secondary peaks, other
– a 20 µl loop injector.
than the peaks due to tetracycline and 4-epichlortetracycline,
is not more than 25 per cent of the area of the peak Inject reference solution (d) and adjust the instrument so that
corresponding to 4-epichlortetracycline in the chromatogram the peak heights correspond to at least 50 per cent of the full
obtained with reference solution (e) (1 per cent). Ignore any scale deflection of the recorder. If necessary, adjust the
peak with an area smaller than that of the principal peak in the dimethyl sulphoxide content in the mobile phase. The test is
chromatogram obtained with reference solution (f) (0.1 per not valid unless the resolution factor between the first peak
cent). (4-epichlor- tetracycline) and the second (chlortetracycline)
is not less than 2.0 and the symmetry factor for the second
Tetracycline hydrochloride. Not more than 8.0 per cent, peak is not more than 1.3.
calculated on the anhydrous basis and determined as described
under the Assay, injecting separately test solution and Inject reference solution (a). The test is not valid unless the
reference solution (e). relative standard deviation of the peak area for chlortetracycline
hydrochloride is not more than 1.0 per cent. If necessary, adjust
Heavy metals (2.3.13). 0.5 g complies with the limit test for the integrator parameters.
heavy metals, Method D (50 ppm), using 2.5 ml of lead standard
Inject alternately the test solution and reference solution (a).
solution (10 ppm Pb) as the standard.
Calculate the content of C22H23ClN2O8,HCl.
Sulphated ash (2.3.18). Not more than 0.5 per cent.
Chlortetracycline Hydrochloride intended for use in the
Water (2.3.43). Not more than 2.0 per cent , determined on manufacture of Parenteral Preparations without a further
0.3 g. appropriate procedure for removal of bacterial endotoxins
Assay. Determine by liquid chromatography (2.4.14). complies with the following additional requirement.

Test solution. Dissolve 100 mg of the substance under Bacterial endotoxins (2.2.3). Not more than 1.1 EU per mg.
examination in 0.01 M hydrochloric acid. Chlortetracycline Hydrochloride intended for use in the
manufacture of Parenteral Preparations without a further
Reference solution (a). A 0.1 per cent w/v solution of
appropriate sterilisation procedure complies with the
chlortetracycline hydrochloride RS in 0.01 M hydrochloric
following additional requirement.
acid.
Sterility (2.2.11). Complies with the test for sterility.
Reference solution (b). A 0.04 per cent w/v of
4-epichlortetracycline hydrochloride RS in 0.01 M Storage. Store protected from light. If it is intended for use in
hydrochloric acid. the manufacture of Parenteral Preparations, the container
should be sterile, tamper-evident and sealed so as to exclude
Reference solution (c). A 0.08 per cent w/v of tetracycline micro-organisms.
hydrochloride RS in 0.01 M hydrochloric acid.
Labelling. The label states (1) the date after which the material
Reference solution (d). Mix 5 ml of reference solution (a) and is not intended to be used; (2) the storage conditions; (3)
10 ml of reference solution (b) and dilute to 25 ml with 0.01 M where applicable, that the material is sterile and free from
hydrochloric acid. Bacterial endotoxins.

1521
CHLORTETRACYCLINE VETERINORY ORAL POWDER IP 2007

Chlortetracycline Veterinary Oral Tests


Powder Other tests. Complies with the tests stated under Veterinary
Oral Powders.
Chlortetracycline Hydrochloride Veterinary Oral Powder;
Chlortetracycline Soluble Powder Assay. Weigh accurately a quantity containing 0.25 g of
Chlortetracycline Hydrochloride, add 500 ml of water, mix
Chlortetracycline Veterinary Oral Powder is a mixture of thoroughly and carry out the microbiological assay of
Chlortetracycline Hydrochloride and Lactose or other suitable antibiotics (2.2.10).
diluent.
Calculate the content of chlortetracycline hydrochloride taking
Chlortetracycline Veterinary Oral Powder contains not less each 1000 Units found to be equivalent to 1 mg of
than 90.0 per cent and not more than 110.0 per cent of the chlortetracycline hydrochloride.
stated amount of chlortetracycline hydrochloride,
C22H23ClN2O8,HCl Storage. Store protected from moisture, at a temperature not
exceeding 30°.
Identification Labelling. The label states the strength in terms of the
concentration of Chlortetracycline Hydrochloride.
A. Determine by thin-layer chromatography (2.4.17), coating
the plate with silica gel H.
NOTE — Use freshly prepared solutions. Cloxacillin Benzathine
Mobile phase. A mixture of 59 volumes of dichloromethane,
35 volumes of methanol and 6 volumes of water. COOH H
O
O N CH3 N
Test solution. The supernatant liquid obtained by extracting a , N
N CH3 H
Cl S
quantity containing 5 mg of Chlortetracycline Hydrochloride H H H
N
with 10 ml of methanol and centrifuging. O 2

Reference solution (a). A 0.05 per cent w/v of


chlortetracycline hydrochloride RS in methanol. C19H18ClN3O5S)2,C16H20N2 Mol. Wt. 1112.1
Cloxacillin Benzathine is N,N′-dibenzylethylene-
Reference solution (b). A solution containing 0.05 per cent w/
diammonium bis-[(6R)-6-{3-(2-chlorophenyl)-5-methyl-
v each of chlortetracycline hydrochloride RS, tetracycline
isoxazole-4-carboxamido}penicillanate].
hydrochloride RS and metacycline hydrochloride RS in
methanol. Cloxacillin Benzathine contains not less than 92.0 per cent of
(C19H18ClN3O5S)2,C16H20N2 and not less than 20.0 per cent and
Adjust the pH of a 10 per cent w/v solution of disodium edetate
not more than 22.0 per cent of benzathine, C16H20N2, both
to 8.0 with 10 M sodium hydroxide and spray this solution calculated on the anhydrous basis.
evenly on the plate (about 10 ml for a plate of 100 mm x 200 mm
size). Allow the plate to dry in a horizontal position for at least Description. A white or almost white powder.
1 hour. Dry the plate in an oven at 100° for 1 hour before use.
Identification
Apply to the plate 1 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 365 nm. A. Determine by infrared absorption spectrophotometry
The principal spot in the chromatogram obtained with the test (2.4.6). Compare the spectrum with that obtained with
solution corresponds to that in the chromatogram obtained cloxacillin benzathine RS or with the reference spectrum of
with reference solution (a). The test is not valid unless the cloxacillin benzathine.
chromatogram obtained with reference solution (b) shows
B. Shake 0.1 g with 1 ml of 1 M sodium hydroxide for 2
three clearly separated spots.
minutes, add 2 ml of ether, shake for 1 minute and allow to
B. To a quantity containing 10 mg of Chlortetracycline separate. Evaporate 1 ml of the ether layer to dryness, dissolve
Hydrochloride, add 20 ml of warm ethanol (95 per cent), allow the residue in 2 ml of glacial acetic acid and add 1 ml of dilute
to stand for 20 minutes, filter and evaporate to dryness on a potassium dichromate solution; a golden yellow precipitate
water-bath. Dissolve the residue in sufficient phosphate buffer is produced.
pH 7.6 to produce a 0.1 per cent w/v solution and heat at 100° C. Shake 50 mg with 10 ml of water and filter. To 5 ml of the
for 1 minute; it exhibits a strong blue fluorescence in filtrate add a few drops of silver nitrate solution; no precipitate
ultra-violet light. is produced. Heat 50 mg with 2 ml of ethanolic potassium

1522
IP 2007 CLOXACILLIN BENZATHINE INTRAMAMMARY INFUSION (DRY COW/BUFFALO)

hydroxide solution on a water-bath for 15 minutes, add 15 mg Cloxacillin Benzathine Intramammary


of decolorising charcoal, shake and filter. Acidify the filtrate
with 2 M nitric acid; the solution gives reaction A of chlorides Infusion (Dry Cow/ Buffalo)
(2.3.1). Cloxacillin Benzathine Intramammary Injection; Cloxacillin
Intramammary Infusion (Dry Cow/Buffalo); Cloxacillin
Tests Intramammary Infusion (DC/B)
Water (2.3.43). Not more than 5.0 per cent w/w, determined on Cloxacillin Benzathine Intramammary Infusion (Dry Cow/
0.5 g. Buffalo) is a sterile suspension of Cloxacillin Benzathine in a
suitable non-aqueous vehicle containing suitable suspending
Assay. For cloxacillin benzathine — Weigh accurately about agents.
60 mg, add 40 ml of methanol, shake to dissolve, add 25 ml of
1 M sodium hydroxide and allow to stand for 30 minutes. Add Cloxacillin Benzathine Intramammary Infusion (Dry Cow/
27.5 ml of 1 M hydrochloric acid and sufficient water to Buffalo) contains not less than 90.0 per cent and not more
produce 100.0 ml, mix, transfer 20.0 ml of the solution to a than 110.0 per cent of the stated amount of cloxacillin,
stoppered conical flask, add 30.0 ml of 0.01 M iodine, close C19H18ClN3O5S.
the flask with a wet stopper and allow to stand for 15 minutes
Identification
protected from light. Titrate the excess of iodine with 0.02 M
sodium thiosulphate, using starch mucilage, added towards Extract a quantity containing 75 mg of cloxacillin with three
the end of the titration, as indicator. Add a further 12 mg of the quantities, each of 15 ml, of light petroleum ( 120° to 160°).
substance under examination to 10 ml of water, swirl to Discard the extracts, wash the residue with 10 ml of ether and
disperse, add 30 ml of 0.01 M iodine and titrate immediately dry in a current of air. The residue complies with the following
with 0.02 M sodium thiosulphate, using starch mucilage, tests.
added towards the end of the titration, as indicator. The
A. Determine by infrared absorption spectrophotometry (2.4.6).
difference between the titrations represents the volume of
Compare the spectrum with that obtained with cloxacillin
0.01 M iodine equivalent to the total penicillins present.
benzathine RS or with the reference spectrum of cloxacillin
Calculate the content of (C19H18ClN3O5S)2,C16H20N2 from the benzathine.
difference obtained by carrying out the procedure
B. Shake 50 mg with 1 ml of 1 M sodium hydroxide for 2
simultaneously using cloxacillin benzathine RS.
minutes, add 2 ml of ether, shake for 1 minute and allow to
For benzathine — Weigh accurately about 1 g, add 30 ml of a separate. Evaporate 1 ml of the ether layer to dryness, dissolve
saturated solution of sodium chloride and 10 ml of 5 M sodium the residue in 2 ml of glacial acetic acid and add 1 ml of dilute
hydroxide, shake well and extract with four quantities, each of potassium dichromate solution; a golden yellow precipitate
50 ml, of ether. Wash the combined extracts with three is produced.
quantities, each of 10 ml, of water, extract the combined
washings with 25 ml of ether and add the extract to the main Tests
ether solution. Evaporate the ether solution to low volume, Water (2.3.43). Not more than 2.0 per cent , determined on 3 g
add 2 ml of ethanol and evaporate to dryness. To the residue and using a mixture of 70 volumes of dichloromethane and 30
add 50 ml of anhydrous glacial acetic acid. Titrate with 0.1 M volumes of anhydrous methanol as the solvent.
perchloric acid, using 0.1 ml of 1-naphtholbenzein solution
Other tests. Complies with the tests stated under
as indicator. Carry out a blank titration.
Intramammary Infusions.
1 ml of 0.1 M perchloric acid is equivalent to 0.01202 g of
Assay. Weigh and mix the contents of 10 containers. Weigh
C16H20N2.
accurately a quantity of the mixed contents containing 80 mg
Cloxacillin Benzathine intended for use in the manufacture of cloxacillin and extract with three quantities, each of 15 ml, of
of either parenteral preparations or intramammary infusions light petroleum ( 120° to 160°) previously saturated with
without a further appropriate sterilisation procedure cloxacillin benzathine. Discard the extracts, wash the residue
complies with the following additional requirement. with ether previously saturated with cloxacillin benzathine.
Dry in a current of air, dissolve in 25 ml of methanol and dilute
Sterility (2.2.11). Complies with the test for sterility.
to 50.0 ml with water. Dilute 2.0 ml to 100.0 ml with buffered
Storage. Store protected from moisture. If it is intended for cupric sulphate solution pH 2.0, transfer 10.0 ml to a stoppered
use in the manufacture of parenteral preparations or test-tube and heat in a water-bath at 70° for 20 minutes. Cool
intramammary infusions, the container should be sterile, to room temperature rapidly, dilute to 20.0 ml with ethanol and
tamper-evident and sealed so as to exclude micro-organisms. measure the absorbance of the resulting solution at the

1523
CLOXACILLIN SODIUM INTRAMAMMARY INFUSION (LACTATING COW/BUFFALO) IP 2007

maximum at about 338 nm (2.4.7), using as the blank 10.0 ml of cloxacillin sodium. Discard the extracts, wash the residue with
the unheated buffered solution of the substance under ether previously saturated with cloxacillin sodium. Dry in a
examination after dilution to 20.0 ml with ethanol. current of air, dissolve in water and dilute to 50.0 ml with the
same solvent. Dilute 2.0 ml to 100.0 ml with buffered cupric
Calculate the content of C19H18ClN3O5S in a container of
sulphate solution pH 2.0, transfer 10.0 ml to a stoppered
average weight from the absorbance obtained by carrying
test-tube and heat in a water-bath at 70° for 20 minutes. Cool
out the procedure simultaneously using 2.0 ml of a solution
to room temperature rapidly, dilute to 20.0 ml with ethanol and
prepared by dissolving 105 mg of cloxacillin benzathine RS
measure the absorbance of the resulting solution at the
in 50.0 ml of a mixture of equal volumes of methanol and
maximum at about 338 nm (2.4.7), using as the blank 10.0 ml of
water.
the unheated buffered solution of the substance under
Labelling. The label states the strength in terms of the examination after dilution to 20.0 ml with ethanol.
equivalent amount of cloxacillin in the sealed container.
Calculate the content of C19H18ClN3O5S in a container of
average content from the absorbance obtained by carrying
out the procedure simultaneously using 2.0 ml of a solution
Cloxacillin Sodium Intramammary prepared by dissolving 85 mg of cloxacillin sodium RS in 50.0
Infusion (Lactating Cow/Buffalo) ml of water, diluting to 100 ml with buffered cupric sulphate
solution pH 2.0, and beginning at the words “transfer 10.0
Cloxacillin Intramammary Injection; Cloxacillin ml.....”.
Intramammary Infusion (Lactating Cow/Buffalo);
Labelling. The label states the strength in terms of the
Cloxacillin Intramammary Infusion (LC/B) equivalent amount of cloxacillin.
Cloxacillin Sodium Intramammary Infusion (Lactating Cow/
Buffalo) is a sterile suspension of Cloxacillin Sodium in a
suitable non-aqueous vehicle containing suitable suspending Dichlofenthion
and dispersing agents.
Cl
Cloxacillin Sodium Intramammary Infusion (Lactating Cow/ H3 C S
Buffalo) contains not less than 90.0 per cent and not more O
P
than 110.0 per cent of the stated amount of cloxacillin, O O
C19H18ClN3O5S. H3 C Cl
Identification C10H13Cl2O3 Mol. Wt. 315.2
Extract a quantity containing 75 mg of cloxacillin with three Dichlofenthion is O-2,4-dichlorophenyl-O,O-diethyl
quantities, each of 15 ml, of light petroleum ( 120° to 160°). phosphorothioate.
Discard the extracts, wash the residue with 10 ml of ether and Dichlofenthion contains not less than 95.0 per cent and not
dry in a current of air. The residue complies with the following more than 100.5 per cent of C10H13Cl2O3PS.
tests.
Description. A colourless or pale yellow, oily substance.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with cloxacillin Identification
sodium RS or with the reference spectrum of cloxacillin sodium.
A. Determine by infrared absorption spectrophotometry
B. Gives reaction A of sodium salts (2.3.1). (2.4.6). Compare the spectrum with that obtained with
dichlofenthion RS or with the reference spectrum of
Tests
dichlofenthion.
Water (2.3.43). Not more than 1.0 per cent, determined on 3 g
B. Determine by thin-layer chromatography (2.4.17), coating
using a mixture of 70 volumes of dichloromethane and 30
the plate with silica gel G.
volumes of anhydrous methanol as the solvent.
Mobile phase. A mixture of 95 volumes of hexane and 5
Other tests. Complies with the tests stated under
volumes of 2-butanone.
Intramammary Infusions.
Assay. Weigh and mix the contents of 10 containers. Weigh Test solution. Dissolve 0.5 g of the substance under
accurately a quantity of the mixed contents containing 80 mg examination in 100 ml of methanol.
of cloxacillin and extract with three quantities, each of 15 ml, of Reference solution. A 0.5 per cent w/v solution of
light petroleum ( 120° to 160°) previously saturated with dichlofenthion RS in methanol.

1524
IP 2007 DICHLOROPHEN

Apply to the plate 2 µl of each solution. After development, Dichlorophen contains not less than 97.0 per cent and not
dry the plate in air and spray with a 2 per cent w/v solution of more than 101.0 per cent of C13H10Cl2O2, calculated on the
4-(4-nitrobenzyl)pyridine in ethyl acetate. Heat the plate at dried basis.
130° for 10 minutes, allow to cool and spray with a 2 per cent
Description. A white or almost white powder; odour, slightly
w/v solution of lithium hydroxide in a mixture of 8 volumes of
phenolic.
methanol, 1 volume of diethylene glycol and 1 volume of
water. The principal spot in the chromatogram obtained with Identification
the test solution corresponds to that in the chromatogram
obtained with the reference solution. A. When examined in the range 220 nm to 360 nm (2.4.7), a
0.002 per cent w/v solution in 0.1 M sodium hydroxide shows
C. Burn 50 mg by the oxygen-flask method (2.3.34), using 20
absorption maxima at about 245 nm and 304 nm. The
ml of 1 M sodium hydroxide as the absorbing liquid. The
absorbances of the solution after further dilution with an equal
solution obtained, after acidification with 2 M nitric acid
volume of 0.1 M sodium hydroxide at these maxima are about
gives reaction A of chlorides and reaction C of phosphates
0.65 and 0.27 respectively.
(2.3.1).
B. Dissolve 0.2 g in 10 ml of 2.5 M sodium hydroxide, cool in
Tests ice and add a solution prepared by mixing 1 ml of sodium
nitrite solution with a cold solution containing 0.15 ml of
Refractive index (2.4.27). 1.530 to 1.533.
aniline in a mixture of 4 ml of water and 1 ml of hydrochloric
Weight per ml (2.4.29). 1.296 to 1.316 g. acid; a reddish-brown precipitate is produced.
Assay. Determine by gas chromatography (2.4.13). C. Fuse 0.5 g with 2 g of anhydrous sodium carbonate, cool,
Test solution (a). Dissolve 0.3 g of the substance under extract the residue with water and filter. The filtrate gives
examination in 100 ml of dichloromethane. reaction A of chlorides (2.3.1).
Test solution (b). A solution containing 0.3 per cent w/v of D. Melting point (2.4.21). about 175°.
the substance under examination and 0.2 per cent w/v of
methyl stearate (internal standard) in dichloromethane. Tests
Reference solution. A solution containing 0.3 per cent w/w of Chlorides (2.3.12). Shake 3.0 g with 6 ml of ethanol (95 per
dichlofenthion RS and 0.2 per cent w/v of methyl stearate cent), dilute with water to 100 ml, allow to stand for 5 minutes
(internal standard) in dichloromethane. and filter. 25 ml of the filtrate complies with the limit test for
Chromatographic system chlorides (330 ppm).
– a glass column 1.5 m x 4 mm, packed with 3 per cent w/w Sulphates (2.3.17). Shake 1.0 g with 20 ml of water for 2 minutes
of phenyl methyl silicone fluid (50 per cent phenyl) on and filter. 5 ml of the filtrate complies with the limit test for
acid-washed, silanised diatomaceous support (80 to 100 sulphates (600 ppm).
mesh) (such as OV-17),
Related substances. Determine by liquid chromatography
– temperature:
(2.4.14).
column 190°,
inlet port and detector. 280°, Test solution. Dissolve 100 mg of the substance under
– flow rate. 30 ml per minute of the carrier gas. examination in 10 ml of the mobile phase.
Calculate the content of C10H13Cl2O3PS. Reference solution (a). A 1.0 per cent w/v solution of
dichlorophen impurity standard RS in the mobile phase.
Reference solution (b). A 0.0010 per cent w/v solution of
Dichlorophen 4-chlorophenol in the mobile phase.
Chromatographic system
OH OH
– a stainless steel column 20 cm x 5 mm, packed with
octadecylsilane bonded to porous silica (10 µm) (such
as Spherisorb ODS 1),
– mobile phase: a filtered and degassed mixture of 75
Cl Cl volumes of methanol, 25 volumes of water and 1 volume
of glacial acetic acid ,
C13H10Cl2O2 Mol. Wt. 269.1 – flow rate. 1.5 ml per minute,
Dichlorophen is 2,2’-methylenebis(4-chlorophenol). – spectrophotometer set at 280 nm,

1525
DICHLOROPHEN VETERINARY AEROSOL IP 2007

– a 20 µl loop injector. cent w/v of potassium ferricyanide. The principal spot in the
chromatogram obtained with the test solution corresponds
Inject the test solution and reference solution (b). In the
to that in the chromatogram obtained with the reference
chromatogram obtained with the test solution the area of the
solution.
peak corresponding to 4-dichlorophenol is not more than the
area of the principal peak in the chromatogram obtained with Tests
reeference solution (b). The content of 4,4´-di- chloro-2,2´-
(2-hydroxy-4-chloro-m-xylene-a,a¢-diyl) diphenol in the Other tests. Complies with the tests stated under Veterinary
substance under examination does not exceed 8.0 per cent w/ Aerosols.
w and the sum of the contents of any other impurities,
Assay. Weigh the intact container. Place the container in an
excluding 4-chlorophenol, is not more than 2.0 per cent w/w.
ice-bath for 15 minutes. Make a small hole about 1 cm from the
Sulphated ash (2.3.18). Not more than 0.1 per cent. top of the body of the container and let the propellant escape.
Loss on drying (2.4.19). Not more than 3 per cent, determined When the flow of propellant stops, enlarge the hole. Transfer
on 1.0 g by drying in an oven at 105°. the contents of the container to a tared vessel capable of
being fitted with a reflux condenser. Remove the top of the
Assay. Weigh accurately about 0.5 g, dissolve in 20 ml of container carefully retaining all fragments. Wash the container
2-propanol. Titrate with 0.1 M tetrabutylammonium with suitable solvents and add the washings to the tared vessel.
hydroxide, determining the end-point potentiometrically Dry the container and reweigh to obtain the net weight of the
(2.4.25). Carry out a blank titration. contents. Heat the contents of the tared vessel under reflux
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to for 30 minutes, cool and weigh (solution A). Dilute an accurately
0.02691 g of C13H10Cl2O2. measured volume of the resulting solution containing about
0.25 g of Dichlorophen to 100.0 ml with acetone. Dilute 2.0 ml
Labelling. The label states that the substance is intended for of this solution to 200.0 ml with ammonia buffer pH 10.9 and
animal treatment only. mix. To 10.0 ml of the resulting solution add 20 ml of ammonia
buffer pH 10.9 and 2 ml of a freshly prepared 2 per cent w/v
solution of 4-aminophenazone, mix, and add 2 ml of a freshly
prepared 8 per cent w/v solution of potassium ferricyanide.
Dichlorophen Veterinary Aerosol Dilute to 50.0 ml with ammonia buffer pH 10.9 and allow to
Dichlorophen Veterinary Aerosol Spray; Dichlorophen stand for 15 minutes. Measure the absorbance of the resulting
Veterinary Spray solution at the maximum at about 510 nm (2.4.7), using as the
blank a solution obtained in a similar manner by carrying out
Dichlorophen Veterinary Aerosol is a solution of Dichloro- the procedure simultaneously, beginning at the words “To
phen in a suitable solvent to which suitable propellants have 10.0 ml of the resulting solution.....” but omitting the
been added. It may contain a suitable dye as a marker. 4-aminophenazone solution. Calculate the weight of
Dichlorophen Veterinary Aerosol contains not less than 90.0 C13H10Cl2O2 in the container from the absorbance obtained by
per cent and not more than 110.0 per cent of the stated amount repeating the operation using a 0.25 per cent w/v solution of
of dichlorophen, C13H10Cl2O2. dichlorophen in acetone beginning at the words “Dilute 2.0
ml.....”.
Identification
Labelling. The label states (1) the weight of Dichlorophen
Determine by thin-layer chromatography (2.4.17), coating the present in the container; (2) the total weight of contents; (3)
plate with silica gel GF254. the name and proportion of any added dye.
Mobile phase. A mixture of 60 volumes of hexane and 40
volumes of acetone.
Test solution. Solution A obtained in the Assay diluted with Dichlorophen Tablets
methanol to contain the equivalent of 1 per cent w/v of Dichlorophen Tablets contain not less than 95.0 per cent and
Dichlorophen. not more than 105.0 per cent of the stated amount of
Reference solution. A 1 per cent w/v solution of dichlorophen dichlorophen, C13H10Cl2O2.
RS in methanol.
Identification
Apply to the plate 5 µl of each solution. After development,
dry the plate in air and spray with a freshly prepared solution A. Shake a quantity of the powdered tablets containing 0.1 g
containing 3.5 per cent w/v of ferric chloride and 0.25 per of Dichlorophen with 50 ml of 0.1 M sodium hydroxide for 15

1526
IP 2007 DIETHYLCARBAMAZINE INJECTION

minutes, add sufficient 0.1 M sodium hydroxide to produce Other tests. Comply with the tests stated under Tablets.
100 ml, centrifuge and dilute a suitable volume of the
Assay. Weigh and powder 20 tablets. Weigh accurately a
supernatant liquid with 0.1 M sodium hydroxide to produce a
quantity of the powder containing 0.1 g of Dichlorophen,
solution containing 0.002 per cent w/v of Dichlorophen.
shake with 50 ml of 0.1 M sodium hydroxide for 15 minutes
When examined in the range 220 to 360 nm (2.4.7), the resulting and add sufficient 0.1 M sodium hydroxide to produce 100.0
solution shows absorption maxima at about 245 nm and 304 ml. Centrifuge and dilute 10.0 ml of the clear supernatant liquid
nm; absorbances at about 245 nm and 304 nm, about 1.3 and to 100.0 ml with 0.1 M sodium hydroxide. Dilute 20.0 ml of this
0.54 respectively. solution to 100.0 ml with 0.1 M sodium hydroxide and measure
B. Shake a quantity of the powdered tablets containing 0.2 g the absorbance of the resulting solution at the maximum at
of Dichlorophen with a mixture of 5 ml of water and 5 ml of 5 about 304 nm (2.4.7). Calculate the content of C13H10Cl2O2
M sodium hydroxide, filter, cool in ice and add a solution taking 275 as the specific absorbance at 304 nm.
prepared by mixing 1 ml of sodium nitrite solution with a cold
solution containing 0.15 ml of aniline in a mixture of 4 ml of
water and 1 ml of hydrochloric acid; a reddish-brown Diethylcarbamazine Injection
precipitate is produced.
Diethylcarbamazine Citrate Injection.
C. Fuse a quantity of the powdered tablets containing 0.5 g of
Dichlorophen with 2 g of anhydrous sodium carbonate, cool, Diethylcarbamazine Injection is a sterile solution of
extract the residue with water and filter. The filtrate gives Diethylcarbamazine Citrate in Water for Injections.
reaction A of chlorides (2.3.1). Diethylcarbamazine Injection contains not less than 95.0 per
cent and not more than 105.0 per cent of the stated amount of
Tests
diethylcarbamazine citrate, C10H21N3O,C6H8O7.
Related substances. Determine by liquid chromatography
(2.4.14). Identification
Test solution. Shake a quantity of the powdered tablets A. To a volume containing 0.5 g of Diethylcarbamazine Citrate
containing 0.50 g of Dichlorophen with 20 ml of methanol for add 2 ml of water and make alkaline with 5 M sodium
10 minutes, filter, add 7 ml of water and dilute to 50 ml with the hydroxide. Extract with four quantities, each of 5 ml, of
mobile phase. dichloromethane, reserve the aqueous solution for test B,
Reference solution (a). A 1.0 per cent w/v solution of wash the combined dichloromethane extracts with water and
dichlorophen impurity standard RS in the mobile phase. remove the dichloromethane by evaporation. Add 0.5 ml of
iodoethane to the residue and heat gently under a reflux
Reference solution (b). A 0.0010 per cent w/v solution of condenser for 5 minutes. Remove the excess iodoethane with
4-chlorophenol in the mobile phase. a current of air, dissolve the viscous yellow oil in 2 ml of
Chromatographic system ethanol (95 per cent) and add, with continuous stirring,
– a stainless steel column 20 cm x 5 mm, packed with sufficient ether to precipitate the quaternary ammonium salt.
octadecylsilane bonded to porous silica (10 µm) (such Decant off the ether, dissolve the residue in 2 ml of ethanol
as Spherisorb ODS 1), (95 per cent), reprecipitate with ether and dry at 105°; the
– mobile phase: a filtered and degassed mixture of 75 residue melts at about 152° (2.4.21).
volumes of methanol, 25 volumes of water and 1 volume
B. Neutralise the aqueous solution obtained in test A with 1 M
of glacial acetic acid ,
sulphuric acid, add an excess of mercuric sulphate solution,
– flow rate. 1.5 ml per minute,
boil and add a few drops of potassium permanganate solution;
– spectrophotometer set at 280 nm,
a white precipitate is produced.
– a 20 µl loop injector.
Inject the test solution and reference solution (b). In the Tests
chromatogram obtained with the test solution the area of the pH (2.4.24). 6.0 to 7.0.
peak corresponding to 4-dichlorophenol is not more than the
area of the principal peak in the chromatogram obtained with N,N’-Dimethylpiperazine and N-methylpiperazine. Determine
reeference solution (b). The content of 4,4´-di- by thin-layer chromatography (2.4.17), coating the plate with
chloro-2,2´-(2-hydroxy-4-chloro-m-xylene-a,a′-diyl) diphenol silica gel G.
in the substance under examination does not exceed 8.0 per Mobile phase. A mixture of 65 volumes of methanol, 30
cent w/w and the sum of the contents of any other impurities, volumes of 2-butanone and 5 volumes of strong ammonia
excluding 4-chlorophenol, is not more than 2.0 per cent w/w. solution.

1527
DIHYDROSTREPTOMYCIN SULPHATE IP 2007

Test solution. Dilute a volume of the injection with sufficient Dihydrostreptomycin Sulphate
methanol to produce a solution containing the equivalent of
5.0 per cent w/v of Diethylcarbamazine Citrate.
Reference solution (a). A 5.0 per cent w/v solution of NH
diethylcarbamazine citrate RS in methanol.
NH
Reference solution (b). A 0.010 per cent w/v solution of HN NH2
N,N´-dimethylpiperazine in methanol. H2N NH
OH
Reference solution (c). A 0.010 per cent w/v solution OH
N-methylpiperazine in methanol. OH
O
Apply to the plate 10 µl of each solution. Allow the mobile O , 3H2SO4
phase to rise 12 cm. Dry the plate at 105° and expose it to R'
iodine vapours for 30 minutes. Any spots corresponding to R
N,N’-dimethylpiperazine and N- methylpiperazine in the OH R = CH3
chromatogram obtained with the test solution are not more O
R = CH2OH
intense than the spots in the chromatograms obtained with OH O
reference solutions (b) and (c) respectively. R'
RHN
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections). OH
2
Assay. To an accurately measured volume containing 8 g of
Diethylcarbamazine Citrate add sufficient water to produce
100.0 ml. To 10.0 ml of this solution add 2 ml of 5 M sodium C21H41N7O12)2,3H2SO4 Mol. Wt. 1461.4
hydroxide and extract with four quantities, each of 25 ml, of Dihydrostreptomycin sulphate is O-2-deoxy-2-methylamino-
dichloromethane. Wash each extract with the same two d-L-lyxofuranosyl-(1→4)-N1,N3-diamidino-D-streptamine
quantities, each of 20 ml, of water and with a third quantity if sulphate.
the second becomes alkaline to phenolphthalein solution.
Dihydrostreptomycin Sulphate has a potency equivalent to
Extract the combined dichloromethane extracts in succession
not less than 730 Units per mg, calculated on the dried basis.
with 25.0 ml of 0.05 M sulphuric acid and 15 ml and 10 ml of
water. Combine the acid and water extracts, remove the Description. A white or almost white powder; may be
dichloromethane, by warming, cool and titrate the excess of hygroscopic.
acid with 0.1 M sodium hydroxide using bromocresol green
solution as indicator. Identification
1 ml of 0.05 M sulphuric acid is equivalent to 0.03914 g of A. Determine by thin-layer chromatography (2.4.17), coating
C10H21N3O,C6H8O7. the plate in the following manner. Mix 0.3 g of carbomer with
240 ml of water, allow to stand with moderate stirring for 1
Storage. Store protected from light.
hour, adjust to pH 7.0 by the gradual addition with constant
shaking of 2 M sodium hydroxide and add 30 g of silica gel H.
Spread a uniform layer of the resulting suspension 0.75 mm
thick. Heat the plate at 110° for 1 hour, allow to cool and use
immediately.
Mobile phase. A 7 per cent w/v solution of potassium
dihydrogen phosphate.
Test solution. Dissolve 100 mg of the substance under
examination in 100 ml of water.
Reference solution (a). A 0.10 per cent w/v of
dihydrostreptomycin sulphate RS in water.
Reference solution (b). A solution containing 0.10 per cent
w/v of dihydrostreptomycin sulphate RS, 0.10 per cent w/v
of neomycin sulphate RS and 0.10 per cent w/v of kanamycin
sulphate RS in water.

1528
IP 2007 DIHYDROSTREPTOMYCIN SULPHATE

Apply to the plate 10 µl of each solution. Allow the mobile 20 minutes after the addition of the ferric ammonium sulphate
phase to rise 12 cm. Dry the plate in a current of warm air, solution, measure the absorbance of a 2-cm layer at the
spray it with a mixture of equal volumes of a 0.2 per cent w/v maximum at about 525 nm (2.4.7), using as the blank a solution
solution of naphthalene-1,3-diol in ethanol (95 per cent) prepared in the same manner, omitting the substance under
and a 46 per cent w/v solution of sulphuric acid and heat at examination. The absorbance is not more than that obtained
150° for 5 to 10 minutes. The principal spot in the chromatogram by carrying out the procedure simultaneously using 5.0 ml of
obtained with the test solution corresponds to that in the a solution prepared by dissolving 10 mg, accurately weighed,
chromatogram obtained with reference solution (a). The test of streptomycin sulphate RS in sufficient water to produce 50
is not valid unless the chromatogram obtained with reference ml and beginning at the words “Add 5.0 ml....”, both
solution (b) shows three clearly separated spots. absorbances being calculated on the dried basis.
B. Dissolve 0.1 g in 2 ml of water and add 1 ml of dilute Methanol. Determine by gas chromatography (2.4.13).
1-naphthol solution and 2 ml of a mixture of equal volumes of
Test solution. Dissolve 4.0 g of the substance under
sodium hypochlorite solution (3 per cent Cl) and water; a
examination in 100 ml of water.
red colour is produced.
Reference solution. A 0.008 per cent w/v of methanol.
C. Dissolve 10 mg in 5 ml of water and add 1 ml of 1 M
hydrochloric acid. Heat in a water-bath for 2 minutes. Add 2 Chromatographic system
ml of a 0.5 per cent w/v solution of 1-naphthol in 1 M sodium – a glass column 1.5 to 2.0 m x 2 to 4 mm, packed with
hydroxide and heat in a water-bath for 1 minute; a violet-pink ethylvinylbenzene-divinylbenzene copolymer (150 to
colour is produced (distinction from streptomycin). 180 mm) porous polymer beads (such as Porapak Q),
– temperature:
D. Gives the reactions of sulphates (2.3.1).
column 50°,
Tests inlet port and detector. 280°,
– flow rate. 30 to 40 ml per minute of the carrier gas.
Appearance of solution. A 25 per cent w/v solution in carbon The area of any peak corresponding to methanol in the
dioxide-free water is not more intensely coloured than degree chromatogram obtained with test solution is not more than
4 of the appropriate range of reference solutions (2.4.1). The that of the peak in the chromatogram obtained with reference
solution, after standing protected from light at a temperature solution (0.2 per cent).
of about 20° for 24 hours, is not more opalescent than
opalescence standard OS2 (2.4.1). Sulphated ash (2.3.18). Not more than 1.0 per cent.
pH (2.4.24). 5.0 to 7.0, determined in a 25 per cent w/v solution. Loss on drying (2.4.19). Not more than 5 per cent, determined
on 1 g by drying over phosphorus pentoxide at 60° at a
Specific optical rotation (2.4.22). –83° to –91°, calculated on pressure not exceeding 0.1 kPa for 4 hours.
the dried basis, determined in a 2 per cent w/v solution in
water. Assay. Carry out the microbiological assay of antibiotics,
Method A or B (2.2.10), and express the result in Units of
Sulphate. 18.0 per cent to 21.5 per cent, calculated on the dihydrostreptomycin per mg.
dried basis.
Dihydrostreptomycin Sulphate intended for use in the
Dissolve 0.25 g in 100 ml of water, adjust the pH to 11 with manufacture of parenteral preparations without a further
strong ammonia solution and add 10.0 ml of 0.1 M barium appropriate procedure for the removal of bacterial
chloride and 0.5 mg of metalphthalein. Titrate the excess of endotoxins complies with the following additional
barium chloride with 0.1 M disodium edetate, adding 50 ml of requirement.
ethanol (95 per cent) when the colour of the solution begins
to change and continuing the titration until the violet-blue Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit
colour disappears. per mg of dihydrostreptomycin sulphate.

1 ml of 0.1 M barium chloride is equivalent to 0.009606 g of Dihydrostreptomycin Sulphate intended for use in the
sulphate, SO4. manufacture of parenteral preparations without a further
appropriate sterilization procedure complies with the
Streptomycin. Weigh accurately about 0.10 g and dissolve in following additional requirement.
sufficient water to produce 5.0 ml. Add 5.0 ml of 0.2 M sodium
hydroxide and heat for exactly 10 minutes in a water-bath. Sterility (2.2.11). Complies with the test for sterility.
Cool in ice for exactly 5 minutes, add 3 ml of a 1.5 per cent w/ Storage. Store protected from light, at a temperature not
v solution of ferric ammonium sulphate in 0.25 M sulphuric exceeding 30°. If it is intended for use in the manufacture of
acid and sufficient water to produce 25.0 ml, and mix. Exactly parenteral preparations or intramammary infusions, the

1529
DIHYDROSTREPTOMYCIN INJECTION IP 2007

container should be sterile and sealed so as to exclude neomycin sulphate RS and 0.10 per cent w/v of kanamycin
micro-organisms. sulphate RS in water.
Labelling. The label states (1) the number of Units per mg; (2) Apply to the plate 10 µl of each solution. Allow the mobile
the name and quantity of any added stabiliser; (3) whether or phase to rise 12 cm. Dry the plate in a current of warm air,
not the contents are intended for use in the manufacture of spray it with a mixture of equal volumes of a 0.2 per cent w/v
Parenteral Preparations or intramammary infusions; (4) that solution of naphthalene-1,3-diol in ethanol (95 per cent)
the substance is meant for veterinary use only; (5) the storage and a 46 per cent w/v solution of sulphuric acid and heat at
conditions; (6) the date after which the contents are not 150° for 5 to 10 minutes. The principal spot in the chromatogram
intended to be used. obtained with test solution corresponds to that in the
chromatogram obtained with reference solution (a). The test
is not valid unless the chromatogram obtained with reference
solution (b) shows three clearly separated spots.
Dihydrostreptomycin Injection B. Dissolve 0.1 g in 2 ml of water and add 1 ml of dilute
Dihydrostreptomycin Sulphate Injection. 1-naphthol solution and 2 ml of a mixture of equal volumes of
sodium hypochlorite solution (3 per cent Cl) and water; a
Dihydrostreptomycin Injection is a sterile solution of red colour is produced.
Dihydrostreptomycin Sulphate in Water for Injections. It is
prepared by dissolving the contents of a sealed container in C. Dissolve 10 mg in 5 ml of water and add 1 ml of 1 M
the requisite amount of Water for Injections immediately hydrochloric acid. Heat in a water-bath for 2 minutes. Add 2
before use. ml of a 0.5 per cent w/v solution of 1-naphthol in 1 M sodium
hydroxide and heat in a water-bath for 1 minute; a violet-pink
Dihydrostreptomycin Injection contains not less than 90.0 colour is produced (distinction from streptomycin).
per cent and not more than 115.0 per cent of the stated amount
of dihydrostreptomycin, C21H41N7O12, calculated on the dried D. Gives the reactions of sulphates (2.3.1).
basis.
Tests
Description. A white or almost white powder which yields a
clear, colourless or faintly yellow solution when dissolved in Appearance of solution. A 25 per cent w/v solution in carbon
water. dioxide-free water is not more intensely coloured than degree
4 of the appropriate range of reference solutions (2.4.1). The
The injection complies with the tests stated under Parenteral solution, after standing protected from light at a temperature
Preparations (Powders for Injection). of about 20° for 24 hours, is not more opalescent than
The contents of the sealed container comply with the opalescence standard OS2 (2.4.1).
following requirements. pH (2.4.24). 5.0 to 7.0, determined in a 25 per cent w/v solution.

Identification Specific optical rotation (2.4.22). –83° to –91°, calculated on


the dried basis, determined in a 2 per cent w/v solution in
A. Determine by thin-layer chromatography (2.4.17), prepared water.
by mixing 0.3 g of carbomer with 240 ml of water, allow to
Sulphate. 18.0 per cent to 21.5 per cent, calculated on the
stand with moderate stirring for 1 hour, adjust to pH 7.0 by the
dried basis.
gradual addition with constant shaking of 2 M sodium
hydroxide and add 30 g of silica gel H. Spread a uniform layer Dissolve 0.25 g in 100 ml of water, adjust the pH to 11 with
of the resulting suspension 0.75 mm thick. Heat the plate at strong ammonia solution and add 10.0 ml of 0.1 M barium
110° for 1 hour, allow to cool and use immediately. chloride and 0.5 mg of metalphthalein. Titrate the excess of
barium chloride with 0.1 M disodium edetate, adding 50 ml of
Mobile phase. A 7 per cent w/v solution of potassium
ethanol (95 per cent) when the colour of the solution begins
dihydrogen phosphate.
to change and continuing the titration until the violet-blue
Test solution. Dissolve 100 mg of the sunbstance under colour disappears.
examination in 100 ml of water.
1 ml of 0.1 M barium chloride is equivalent to 0.009606 g of
Reference solution (a). A 0.10 per cent w/v of sulphate, SO4.
dihydrostreptomycin sulphate RS in water.
Streptomycin. Weigh accurately about 0.10 g and dissolve in
Reference solution (b). A solution containing 0.10 per cent w/ sufficient water to produce 5.0 ml. Add 5.0 ml of 0.2 M sodium
v of dihydrostreptomycin sulphate RS, 0.10 per cent w/v of hydroxide and heat for exactly 10 minutes in a water-bath.

1530
IP 2007 DIMETRIDAZOLE

Cool in ice for exactly 5 minutes, add 3 ml of a 1.5 per cent w/ Storage. Store protected from light. Use the injection within 7
v solution of ferric ammonium sulphate in 0.25 M sulphuric days of the preparation of the solution when stored in a cool
acid and sufficient water to produce 25.0 ml, and mix. Exactly place or within 1 month when stored in a cold place.
20 minutes after the addition of the ferric ammonium sulphate
Labelling. The label states (1) the strength in terms of the
solution, measure the absorbance of a 2-cm layer at the
equivalent amount of dihydrostreptomycin in a suitable
maximum at about 525 nm (2.4.7), using as the blank a solution
dose-volume; (2) that the contents are meant for veterinary
prepared in the same manner, omitting the substance under
use only; (3) the storage conditions; (4) the date after which
examination. The absorbance is not more than that obtained
the contents are not intended to be used.
by carrying out the procedure simultaneously using 5.0 ml of
a solution prepared by dissolving 10 mg, accurately weighed,
of streptomycin sulphate RS in sufficient water to produce 50
ml and beginning at the words “Add 5.0 ml....”, both
absorbances being calculated on the dried basis. Dimetridazole
Methanol. Determine by gas chromatography (2.4.13).
CH3
Test solution. Dissolve 4.0 g of the substance under N
examination in 100 ml of water. O2N CH3
Reference solution. A 0.008 per cent w/v of methanol. N
Chromatographic system
C5H7N3O2 Mol. Wt. 141.1
– a glass column 1.5 to 2.0 m x 2 to 4 mm, packed with
ethylvinylbenzene-divinylbenzene copolymer (150 to Dimetridazole is 1,2-dimethyl-5-nitro-1H-imidazole.
180 mm) porous polymer beads (such as Porapak Q), Dimetridazole contains not less than 98.0 per cent and not
– temperature: more than 101.0 per cent of the stated amount of dimetridazole,
column 50°, C5H7N3O2, calculated on the anhydrous basis.
inlet port and detector. 280°,
– flow rate. 30 to 40 ml per minute of the carrier gas. Description. An almost white to brownish yellow powder;
darkens on exposure to light, odourless or almost odourless.
The area of any peak corresponding to methanol in the
chromatogram obtained with test solution is not more than Identification
that of the peak in the chromatogram obtained with reference
solution (0.2 per cent). A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with dimetridazole
Sulphated ash (2.3.18). Not more than 1.0 per cent. RS or with the reference spectrum of dimetridazole.
Loss on drying (2.4.19). Not more than 5 per cent, determined B. When examined in the range of 230 to 360 nm (2.4.7), a 0.002
on 1 g by drying over phosphorus pentoxide at 60° at a per cent w/v solution in methanol shows a well-defined
pressure not exceeding 0.1 kPa for 4 hours. absorption maximum only at about 309 nm; absorbance at
Assay. On the mixed contents of ten containers carry out the about 309 nm, about 1.3.
microbiological assay, Method A or B (2.2.10), and express the C. Dissolve 0.1 g in 20 ml of ether, add 10 ml of a 1 per cent w/
result in Units of dihydrostreptomycin per mg. v solution of picric acid in ether, induce crystallisation by
Dihydrostreptomycin Sulphate intended for use in the scratching the sides of the vessel and allow to stand. Wash
manufacture of Parenteral Preparations without a further the precipitate obtained with ether and dry at 105°; the residue
appropriate procedure for the removal of bacterial melts at about 160° (2.4.21).
endotoxins complies with the following additional
requirement. Tests
Bacterial endotoxins (2.2.3). Not more than 0.5 Endotoxin Unit 2-Methyl-5-nitroimidazole. Determine by thin-layer
per mg of dihydrostreptomycin sulphate. chromatography (2.4.17), coating the plate with silica gel
Dihydrostreptomycin Sulphate intended for use in the GF254.
manufacture of Parenteral Preparations without a further Mobile phase. A mixture of 90 volumes of dichloromethane
appropriate sterilization procedure complies with the and 10 volumes of 2-propanol.
following additional requirement.
Test solution. Dissolve 2 g of the substance under examination
Sterility (2.2.11). Complies with the test for sterility. in 100 ml of dichloromethane.

1531
DIMETRIDAZOLE PREMIX IP 2007

Reference solution. A 0.010 per cent w/v of 2-methyl- Dimetridazole Veterinary Oral Powder
5-nitroimidazole RS in dichloromethane.
Dimetridazole Veterinary Oral Powder is a mixture of
Apply to the plate 5 µl of each solution. After development, Dimetridazole and a suitable water-soluble diluent.
dry the plate in air and examine in ultraviolet light at 254 nm.
Any spot corresponding to 2-methyl-5-nitroimidazole in the Dimetridazole Veterinary Oral Powder contains not less than
chromatogram obtained with the test solution is not more 95.0 per cent and not more than 105.0 per cent of the stated
intense than the spot in the chromatogram obtained with the amount of dimetridazole, C5H7N3O2.
reference solution.
Identification
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Water (2.3.43). Not more than 1.0 per cent, determined on 1 g. Mix a quantity containing 0.1 g of Dimetridazole with 20 ml of
ether, shake and filter. To the filtrate add 10 ml of a 1 per cent
Assay. Weigh accurately about 0.3 g, dissolve in 30 ml of w/v solution of picric acid in ether, stir to induce
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric crystallisation and allow to stand. Wash the precipitate
acid, using crystal violet solution as indicator. Carry out a obtained with ether and dry at 105°; the residue melts at about
blank titration. 160° (2.4.21).
1 ml of 0.1 M perchloric acid is equivalent to 0.01411 g of
C5H7N3O2. Tests
Storage. Store protected from light. Other tests. Complies with the tests stated under Veterinary
Oral Powders.
Assay. Weigh accurately a quantity containing 0.4 g of
Dimetridazole Premix Dimetridazole, transfer to a sintered glass funnel (porosity
No. 4), add 10 ml of dichloromethane, stir for 1 minute, and
Dimetridazole Premix contains not less than 95.0 per cent and apply gentle suction. Repeat the extraction with four further
not more than 105.0 per cent of the stated amount of quantities, each of 10 ml, of dichloromethane. To the combined
dimetridazole, C5H7N3O2. dichloromethane extracts add 50 ml of anhydrous glacial acetic
acid previously neutralised to crystal violet solution by the
Identification
dropwise addition of 0.1 M perchloric acid. Titrate with 0.1
Mix a quantity containing 0.1 g of Dimetridazole with 20 ml of M perchloric acid, using crystal violet solution as indicator.
ether, shake and filter. To the filtrate add 10 ml of a 1 per cent Carry out a blank titration.
w/v solution of picric acid in ether, stir to induce 1 ml of 0.1 M perchloric acid is equivalent to 0.01411 g of
crystallisation and allow to stand. Wash the precipitate C5H7N3O2.
obtained with ether and dry at 105°; the residue melts at about
160° (2.4.21). Storage. Store protected from light.

Tests
Other tests. Complies with the tests stated under Premixes. Dinitolmide
Assay. Weigh accurately a quantity containing 0.45 g of
Dimetridazole, transfer to a sintered glass funnel (porosity O NH2
No. 4), add 10 ml of dichloromethane, stir for 1 minute, and
apply gentle suction. Repeat the extraction with four further CH3
quantities, each of 10 ml, of dichloromethane. To the combined
dichloromethane extracts add 50 ml of anhydrous glacial acetic O2N NO2
acid previously neutralised to crystal violet solution by the
dropwise addition of 0.1 M perchloric acid. Titrate with 0.1
M perchloric acid, using crystal violet solution as indicator. C8H7N3O5 Mol. Wt. 225.2
Carry out a blank titration. Dinitolmide is 3,5-dinitro-2-methylbenzamide.
1 ml of 0.1 M perchloric acid is equivalent to 0.01411 g of Dinitolmide contains not less than 98.0 per cent and not more
C5H7N3O2. than 100.5 per cent of C8H7N3O5, calculated on the dried basis.
Storage. Store protected from light. Description. A cream to light tan powder.

1532
IP 2007 ETHOPABATE

Identification 1 ml of 0.1 M titanium trichloride is equivalent to 0.001876 g


of C8H7N3O5.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with dinitolmide
RS or with the reference spectrum of dinitolmide.
B. Heat 1 g with 20 ml of 9 M sulphuric acid under a reflux
Ethopabate
condenser for 1 hour, cool, add 50 ml of water and filter. The
residue after washing with water and drying at 105° melts at O
about 205° (2.4.21).
O OCH3
Tests
H3C N O CH3
Acid value (2.3.23). Not more than 5.0, determined on 0.5 g and H
using 50 ml of ethanol (95 per cent) as the solvent. C12H15NO4 Mol. Wt. 237.3
Related substances. Determine by thin-layer chromatography Ethopabate is methyl 4-acetamide-2-ethoxybenzoate.
(2.4.17), coating the plate with silica gel GF254.
Ethopabate contains not less than 96.0 per cent and not more
Mobile phase. A mixture of 85 volumes of dichloromethane, than 104.0 per cent of ethopabate, C12H15NO4, calculated on
10 volumes of methanol and 5 volumes of glacial acetic acid. the dried basis.
Test solution. Dissolve 2.5 g of the substance under Description. A white or pinkish white powder, odourless or
examination in 100 ml of acetone. almost odourless.
Reference solution (a). A 0.0125 per cent w/v of the substance
under examination in acetone. Identification
Reference solution (b). A 0.0125 per cent w/v of o-toluic acid A. Determine by infrared absorption spectrophotometry (2.4.6).
in acetone. Compare the spectrum with that obtained with ethopabate RS
or with the reference spectrum of ethopabate.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. B. When examined in the range 230 to 360 nm (2.4.7), a 0.0016
Spray with titanium trichloride solution, diluted 5 times with per cent w/v solution in methanol shows absorption maxima
water, heat at 100° for 5 minutes and spray with ethanolic at about 268 nm and at about 299 nm; absorbance at about 268
dimethylaminobenzaldehyde solution. When viewed under nm, about 1.3 and at about 299 nm, about 0.58.
ultraviolet light at 354 nm the spot in the chromatogram C. Melts at about 148° (2.4.21).
obtained with reference solution (b) is more intense than any
corresponding spot in the chromatogram obtained with the Tests
test solution. By both methods of visualisation any secondary
Diazotisable substances. Dissolve 0.2 g in 10 ml of
spot in the chromatogram obtained with the test solution is
dichloromethane and extract in succession with 100 ml and
not more intense than the spot in the chromatogram obtained
90 ml of 0.1 M hydrochloric acid, combine the acid extracts,
with reference solution (a).
wash with 5 ml of dichloromethane, dilute to 200 ml with 0.1
Loss on drying (2.4.19). Not more than 1.0 per cent, determined M hydrochloric acid and filter. To 5 ml, add 6 ml of 1 M
on 1.0 g by drying in an oven at 105°. hydrochloric acid and 1 ml of a 0.1 per cent w/v solution of
Assay. Weigh accurately about 0.15 g, dissolve in acetone sodium nitrite, mix, and allow to stand for 4 minutes. Add 1 ml
and dilute to 50.0 ml. To 10.0 ml of the solution add 10 ml of of a 0.5 per cent w/v solution of ammonium sulphamate, mix
glacial acetic acid and 15 ml of a 40 per cent w/v solution of and allow to stand for 3 minutes. Add 1.0 ml of a 0.1 per cent
sodium acetate. Maintain a stream of carbon dioxide through w/v solution of N-(1-naphthyl)ethylenediamine dihydro-
the flask throughout the determination. Add 25.0 ml of 0.1 M chloride, mix, and allow to stand for 30 minutes. Absorbance
titanium trichloride and allow to stand for 5 minutes. Add 10 of the resulting solution at about 545 nm (2.4.7), not more than
ml of hydrochloric acid, 10 ml of water and 1 ml of potassium 0.70.
thiocyanate solution. Titrate with 0.1 M ferric ammonium Phenolic substances. Dissolve 0.25 g in 15 ml of methanol
sulphate until the solution becomes first colourless and then and add sufficient methanol to produce 25 ml. To 5 ml add 5
orange. Repeat the operation without the substance under ml of a 3 per cent w/v solution of anhydrous ferric chloride,
examination. The difference between the titrations represents mix and allow to stand for 10 minutes. Absorbance of the
the amount of titanium trichloride required. resulting solution at about 525 nm (2.4.7), not more than 0.70,

1533
FURAZOLIDONE VETERINARY ORAL SUSPENSION IP 2007

using as the blank a solution prepared by adding 5 ml of a 3 Test solution. Shake a quantity of the suspension containing
per cent w/v solution of anhydrous ferric chloride to 5 ml of 5 mg of Furazolidone with 1 ml of acetone, allow to stand, and
methanol. use the supernatant liquid.
Sulphated ash (2.3.18). Not more than 0.5 per cent. Reference solution. A 0.5 per cent w/v solution of furazolidone
RS in acetone.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g, by drying in an oven at 105° at a pressure not Apply to the plate 10 µl of each solution. After development,
exceeding 0.7 kPa. dry the plate in air and examine in ultraviolet light at 254 nm.
The principal spot in the chromatogram obtained with the test
Assay. Weigh accurately about 75 mg and dissolve in sufficient
solution corresponds to that in the chromatogram obtained
methanol to produce 250.0 ml. Dilute 10.0 ml to 100.0 ml with
with the reference solution.
water, transfer 10.0 ml of this solution to an evaporating dish,
add 10 ml of 1 M sodium hydroxide and evaporate to dryness
on a water-bath. Moisten the dish with 10 ml of water, evaporate
Tests
to dryness, again moisten with 10 ml of water and heat on a Other tests. Complies with the tests stated under Veterinary
water-bath for 15 minutes. Add 20 ml of 1 M hydrochloric Oral Liquids.
acid, transfer to a flask and add sufficient water to produce
100.0 ml. To 25.0 ml add 5.0 ml of 1 M hydrochloric acid and Assay. Protect the solutions from light throughout the assay.
5.0 ml of a 0.1 per cent w/v solution of sodium nitrite, mix and Weigh accurately a quantity of the well-shaken suspension
allow to stand for 2 minutes. Add 5.0 ml of 0.5 per cent w/v containing 35 mg of Furazolidone, add slowly and with stirring,
solution of ammonium sulphamate, mix and allow to stand for 50 ml of dimethylformamide. Warm on a water-bath, with
2 minutes. Add 5.0 ml of a 0.1 per cent w/v solution of occasional stirring, until most of the solid is dissolved. Decant
N-(1-naphthyl)ethylenediamine dihydrochloride, mix and the supernatant liquid and extract the residue further with two
allow to stand for 10 minutes. Add sufficient water to produce quantities, each of 50 ml, of dimethylformamide, decanting
50.0 ml and measure the absorbance of the resulting solution the supernatant solution. No yellow colour should be visible
at the maximum at about 545 nm (2.4.7). Calculate the content in the third extract. Cool the combined dimethylformamide
of C12H15NO4 from the absorbance obtained by carrying out extracts, add sufficient water to produce 500.0 ml and filter. To
the procedure simultaneously, using ethopabate RS in place 10.0 ml of the filtrate add sufficient water to produce 100.0 ml
of the substance under examination. and measure the absorbance of the resulting solution at the
maximum at about 367 nm (2.4.7). Calculate the content of
C8H7N3O5 taking 754 as the specific absorbance at 367 nm.
Determine the weight per ml of the suspension (2.4.29), and
Furazolidone Veterinary Oral calculate the content of furazolidone, weight in volume.
Suspension Storage. Store protected from light.
Furazolidone Veterinary Mixture; Furazolidone Mixture; Labelling. The label states that the oral suspension should
Furazolidone Drench be administered undiluted.
Furazolidone Veterinary Oral Suspension is an aqueous
suspension of Furazolidone.
Furazolidone Veterinary Oral Suspension contains not less
than 90.0 per cent and not more than 110.0 per cent of the Furazolidone Premix
stated amount of furazolidone, C8H7N3O5. Furazolidone Premix contains not less than 95.0 per cent and
not more than 105.0 per cent of the stated amount of
Identification furazolidone, C8H7N3O5.
A. Add 0.2 ml to a mixture of 15 ml of dimethylformamide and
1 ml of 0.5 M ethanolic potassium hydroxide; a blue colour is Identification
produced. A. To a mixture of 15 ml of dimethylformamide and 1 ml of 0.5
B. Determine by thin-layer chromatography (2.4.17), coating M ethanolic potassium hydroxide add 5 mg of the premix; a
the plate with silica gel G. blue colour is produced.
Mobile phase. A mixture of 50 volumes of dichloromethane B. Determine by thin-layer chromatography (2.4.17), coating
and 40 volumes of nitromethane and 10 volumes of methanol. the plate with silica gel G.

1534
IP 2007 HALOXON

Mobile phase. A mixture of 50 volumes of dichloromethane When examined in the range 230 to 360 nm (2.4.7), the resulting
and 40 volumes of nitromethane and 10 volumes of methanol. solution exhibits a maximum at about 290 nm and a less well
defined maximum at about 312 nm. Ratio of the absorbance at
Test solution. The supernatant liquid obtained by shaking a
about 312 nm to that at about 290 nm, about 1.08.
quantity of the premix containing 5 mg of furazolidone with 1
ml of acetone. B. Dissolve 0.1 g in 5 ml of 5 M sodium hydroxide with the aid
of warming, cool, acidify 1 ml of the solution by the addition
Reference solution. A 0.5 per cent w/v solution of furazolidone
of 2 M nitiric acid and add 1 ml of silver nitrate solution, a
RS in acetone.
white precipitate is formed. The precipitate is soluble in 5 M
Apply to the plate 10 µl of each solution. After development, ammonia giving a brown solution which exhibits a green
dry the plate in air and examine in ultraviolet light at 254 nm. fluorescence when viewed under screened ultraviolet light.
The principal spot in the chromatogram obtained with the test
C. Melting range (2.4.21). 88° to 93°.
solution corresponds to that in the chromatogram obtained
with the reference solution. Tests
Tests Acidity. Dissolve 0.1 g in 10 ml of ethanol (95 per cent)
previously neutralised to methyl red solution; the solution
Other tests. Complies with the tests stated under Premixes.
requires for neutralisation not more than 0.1 ml of 0.1 M sodium
Assay. Protect the solutions from light throughout the assay. hydroxide.
Weigh accurately a quantity of the premix containing 35 mg of 3-Chloro-4-methylumbelliferone. Not more than 2.0 per cent.
Furazolidone, add 50 ml of dimethylformamide and shake for
NOTE – Prepare the solutions immediately before use and
20 minutes. Add sufficient water to produce 500.0 ml and
protected from light.
filter. To 10.0 ml of the filtrate add sufficient water to produce
100.0 ml and measure the absorbance of the resulting solution Dissolve 0.20 g in 50 ml of 0.01 M methanolic hydrochloric
at the maximum at about 367 nm (2.4.7). Calculate the content acid and dilute 5 ml to 100 ml with 0.01 M methanolic
of C8H7N3O5 taking 754 as the specific absorbance at 367 nm. hydrochloric acid. Measure the fluorescence of the resulting
solution (2.4.5), using an excitation wavelength of about 345
Determine the weight per ml, (2.4.29), and calculate the content
nm and an emission wavelength of about 400 nm and setting
of furazolidone, weight in volume.
the spectrofluorimeter to zero with 0.01 M methanolic
Storage. Store protected from light. hydrochloric acid and to 100 with a standard solution prepared
by dissolving 25 mg of 3-chloro-4-methyl- umbelliferone RS
in sufficient 0.01 M methanolic hydrochloric acid to produce
250 ml (solution A) and diluting 5 ml to 100 ml with 0.01 M
Haloxon methanolic hydrochloric acid. Calculate the content of
O O O O 3-chloro-4-methylumbelliferone from a calibration curve
Cl P prepared by measuring the fluorescence of suitable dilutions
Cl O O of solution A.
Cl
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
CH3
on 1.0 g by drying in an oven at 80° at a pressure not exceeding
0.7 kPa.
C14H14Cl3O6P Mol. Wt. 415.6
Assay. Weigh accurately about 0.25 g and dissolve in sufficient
Haloxone is phosphoric acid bis(2-chloroethyl) 3-chloro-4-
acetonitrile to produce 10 ml and record the infrared
methyl-2-oxo-2H-1-benzopyran-7-yl ester.
absorption of a 0.2 mm layer of the solution at the maximum at
Haloxon contains not less than 95.0 per cent and not more about 1155 cm– 1(2.4.6). Construct a base line between the
than 100.5 per cent of C14H14Cl3O6P, calculated on the dried minima at about 1125 cm– 1 and 1180 cm– 1. Calculate the content
basis. of C14H14Cl3O6P from the absorption obtained by repeating
Description. A white or almost white powder. the procedure using haloxon RS in place of the substance
under examination.
Identification Storage. Avoid contact with metals.
A. Dissolve about 20 mg in 10 ml of dioxan, add 0.5 ml of 0.1
M hydrochloric acid and dilute to 25 ml with methanol. Dilute
1 ml to 25 ml with methanol.

1535
IVERMECTION IP 2007

Ivermectin Chromatographic system


– a stainless steel column 25 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm),
OCH3
– mobile phase: a mixture of 15 volumes of water, 34
HO OCH3 volumes of methanol and 51 volumes of acetonitrile,
O – flow rate. 1 ml per minute,
H3C O CH3 CH3
H O H – spectrophotometer set at 254 nm,
O CH3
H3 C O – a 20 µl loop injector.
O
R
H3C H Inject reference solution (a). This test is not valid unless
O O resolution between the component H2B1b (first peak) and
OH component H2B1b (second peak) is not less than 3.0.
Component B1a R = CH2CH3
Inject the test solution and reference solution (b). In the
Component B1b R = CH3 O
CH3 chromatogram obtained with the test solution the impurity
H
OH with a relative retention of 1.3 to 1.5 with reference to the
principal peak is not more than 2.5 times the area of the principal
C48 H74O14, H2B1a Mol.Wt. 875.0 peak in the chromatogram obtained with reference solution
C47 H72O14, H2B1b Mol.Wt. 861.0 (b) (2.5 per cent). The area of any other peak is not more than
the area of the principal peak in the chromatogram obtained
Ivermectin contains not less than 95.0 per cent and not more with reference solution (b) (1.0 per cent) and the sum of all the
than 102.0 per cent of H2B1a + H2B1b, calculated on the dried secondary peaks is not more than 5 times the area of the
basis. principal peak in the chromatogram obtained with reference
The ratio H 2B1a/(H 2B1a + H2B1b), determined by liquid solution (b) (5.0 per cent).
chromatography is not less than 90.0 per cent. Ethanol and formamide. Determine by gas chromatography
Description. A white crystalline powder, slightly hygroscopic. (2.4.13).
Internal standard solution. Dilute 0.5 ml of propanol to 100
Identification
ml with water.
A. Determine by infrared absorption spectrophotometery Test solution. Dissolve 0.120 g of the substance under
(2.4.6). Compare the spectrum with that obtained with examination in 2.0 ml of m-xylene by heating on a water-bath
ivermectin RS. at 40 to 50°, add 2.0 ml of water, mix thoroughly and centrifuge.
B. In the Assay, the principal peak in the chromatogram Remove the upper layer and extract it with 2.0 ml of water.
obtained with the test solution corresponds to the peak in the Discard the upper layer and combine the aqueous layers. Add
chromatogram obtained with reference solution (a). 1.0 ml of the internal standard solution. Centrifuge and discard
any remaining m-xylene.
Tests Reference solution (a). Dilute 3.0 g of ethanol to 100 ml with
Appearance of solution. A 2.0 per cent w/v solution in toluene water.
is clear (2.4.1) and not more intensely colored than reference Reference solution (b). Dilute 1.0 g of formamide to 100 ml
solution BY57 (2.4.1). with water.
Specific optical rotation (2.4.22) -17.0 to -20.0, determined on Reference solution (c). Dilute 5.0 ml of reference solution (a)
a 2.5 per cent w/v solution in methanol. and 5 ml of reference solution (b) to 50.0 ml with water. Transfer
Related substances. Determine by liquid chromatography 2.0 ml of this solution to a centrifuge tube, add 2 ml of m-
(2.4.14). xylene, mix thoroughly and centrifuge. Remove the upper layer
Test solution. Dissolve 40 mg of the substance under and extract it with 2.0 ml of water. Discard the upper layer and
examination in 50 ml of methanol. combine the aqueous layers. Add 1.0 ml of the internal standard
solution. Centrifuge and discard any remaining m-xylene.
Reference solution (a). A 0.08 per cent w/v solution of
ivermectin RS in methanol. Reference solution (d). Dilute 10.0 ml of reference solution (a)
and 10.0 ml of reference solution (b) to 50.0 ml with water.
Reference solution (b). Dilute 1.0 ml of reference solution (a) Transfer 2.0 ml of this solution to a centrifuge tube, add 2 ml of
to 100 ml with methanol. m-xylene, mix thoroughly and centrifuge. Remove the upper
Reference solution (c). Dilute 5 ml of reference solution (b) to layer and extract it with 2.0 ml of water. Discard the upper
100 ml with methanol. layer and combine the aqueous layers. Add 1.0 ml of the internal

1536
IP 2007 KAOLIN VETERINARY ORAL SUSPENSION

standard solution. Centrifuge and discard any remaining m- Assay. Determine by liquid chromatography (2.4.14).
xylene.
Test solution. Dilute accurately a volume of the injection
Chromatographic system containing 5 mg of Ivermectin to 100 ml with water.
– a glass column 30 m x 0.53 mm, packed with fused silica Reference solution. A 0.005 per cent w/v solution of ivermectin
with macrogol 20,000 with film thickness 1 mm, RS in water.
– temperature
column 80° increase @ 60° per minute to 240°, Chromatographic system
injection port 220° and detector 280°, – a stainless steel column 25 cm x 4.6 mm, packed with
– flow rate. 7.5 ml per minute of helium as carrier gas. octadecylsilane bonded to porous silica (5 µm),
– mobile phase: a mixture of 9 volumes of methanol and 1
Inject 1 µl of the test solution and reference solutions (c) and volume of water,
(d). – flow rate. 1 ml per minute,
Calculate the content of ethanol is not more than 5 per cent – spectrophotometer set at 245 nm,
and formamide not more than 1 per cent. – a 20 µl loop injector.
Heavy metals (2.3.13). 1 g complies with the limit test for heavy Inject the reference solution. The test is not valid unless the
metals, Method C (20 ppm). tailing factor is not less than 2.0
Sulphated ash (2.3.18). Not more than 0.1 per cent. Inject alternatively the test solution and the reference solution.
Water (2.3.43). Not more 0.1 per cent, determined on 0.5 gm. Calculate the content of ivermectin in the injection.
Assay. Determine by liquid chromatography (2.4.14), as Storage. Store protected from light.
described under Related substances. Labelling. The label states (1) the strength in mg of Ivermectin
Inject the test solution and reference solution (a). per ml; (2) that the contents are to be used for subcutaneous
use only; (3) the names of any preservatives used.
Calculate the percentage contents of ivermectin (H2B1a + H2B1b)
and the ratio H2B1a/(H2B1a + H2B1b).
Storage. Store protected from light.
Kaolin Veterinary Oral Suspension
Kaolin Veterinary Mixture; Kaolin Mixture
Ivermectin Injection
Light Kaolin 200 g
Ivermectin Injection is a sterile solution of Ivermectin with or
Light Magnesium Carbonate 50 g
with out one or more anaesthetics, preservatives and solvents.
Sodium Bicarbonate 50 g
Ivermectin Injection contains not less than 90 per cent and
Water to produce 1000 ml
not more than 110 per cent of H2B1a, and not more than 5 per
cent of H2B1b. Kaolin Veterinary Oral Suspension should be freshly prepared,
The content of H2B1a + H2B1b is not less than 95 per cent and unless the Light Kaolin has been sterilised.
not more than 110 per cent of the stated amount of Ivermectin. Kaolin Veterinary Oral Suspension contains not less than 1.04
Description. A clear, colourless to yellow colour solution. per cent w/w and not more than 1.25 per cent w/w of the stated
amount of magnesium, Mg and not less than 4.05 per cent w/
Identification w and not more than 4.65 per cent w/w of the stated amount of
sodium bicarbonate, NaHCO3.
When examined in the range 220 nm to 360 nm (2.4.7), a 0.001
per cent w/v solution in methanol shows an absorption Tests
maximum at about 245 nm.
Acid-insoluble matter. 13.8 to 18.4 per cent w/w, determined
Tests by the following method. Weigh accurately about 3 g, add 15
pH (2.4.24). 5.5 to 7.0. ml of water and make acid to litmus paper by the cautious
addition of 2 M hydrochloric acid; boil for 5 minutes, replacing
Pyrogens (2.2.8). Complies with the test for pyrogens, by water lost by evaporation, cool and decant the supernatant
injecting 0.2 mg of Ivermectin per kg body weight of rabbit. layer through a filter. Boil the residue with 20 ml of water and
Other tests. Complies with the tests stated under Parenteral 10 ml of 2 M hydrochloric acid, cool, filter through the same
Preparations (Injections). filter, and wash the residue with water until the washings are

1537
LEVAMISOLE INJECTION IP 2007

free from chloride, reserving the filtrate and washings for the Apply to the plate 1 µl of each solution. After development,
Assay for magnesium. Dry and ignite the residue to constant dry the plate in air and spray with potassium iodoplatinate
weight at red heat. solution. The principal spot in the chromatogram obtained
with the test solution corresponds to that in the chromatogram
Other tests. Complies with the tests stated under Veterinary
obtained with the reference solution.
Oral Liquids.
B. Dilute a volume of the injection containing 0.75 g of
Assay. For magnesium — Dilute the combined filtrate and
Levamisole Hydrochloride to 20 ml with water and add 6 ml of
washings reserved in the determination of acid-insoluble matter
1 M sodium hydroxide. Extract with 20 ml of dichloromethane,
to 100.0 ml with water. To 20.0 ml add 0.1 g of ascorbic acid,
discard the aqueous layer and wash the dichloromethane layer
make slightly alkaline to litmus paper with 5 M ammonia and
with 10 ml of water. Dry by shaking with anhydrous sodium
add 10 ml of triethanolamine, 10 ml of ammonia buffer pH
sulphate, filter and evaporate the solvent at room temperature.
10.9 and 1 ml of potassium cyanide solution. Titrate with
The residue, after drying over phosphorus pentoxide at a
0.05 M disodium edetate using eriochrome black T solution
pressure of 1.5 to 2.5 kPa at a temperature not exceeding 40°,
as indicator.
melts at about 59° (2.4.21).
1 ml of 0.05 M disodium edetate is equivalent to 0.001215 g of
C. The injection is laevorotatory.
Mg.
D. Gives reaction B of chlorides (2.3.1).
For sodium bicarbonate — Weigh accurately about 10 g, boil
with 100 ml of water for 5 minutes and filter. Boil the residue Tests
with 100 ml of water for 5 minutes and filter. Cool the combined
filtrates and titrate with 0.5 M hydrochloric acid using methyl pH (2.4.24). 3.3 to 3.7.
orange-xylene cyanol FF solution as indicator. Add 10 ml of 2,3-Dihydro-6-phenylimidazo[2,1-b]thiazole hydrochloride.
ammonia buffer pH 10.9 and titrate with 0.05 M disodium Determine by thin-layer chromatography (2.4.17), coating the
edetate using eriochrome black T solution as indicator. plate with silica gel G.
1 ml of 0.5 M hydrochloric acid after subtracting one fifth of Mobile phase. A mixture of 45 volumes of toluene, 8 volumes
the volume of 0.05 M disodium edetate is equivalent to 0.0420 of methanol and 4 volumes of anhydrous glacial acetic acid.
g of NaHCO3.
Test solution. Dilute a volume of the injection with methanol
to produce a solution containing 5.0 per cent w/v of Levamisole
Hydrochloride.
Levamisole Injection Reference solution. A 0.025 per cent w/v of 2,3-dihydro-6-
phenylimidazo[2,1-b]thiazole hydrochloride RS in
Levamisole Hydrochloride Injection
methanol.
Levamisole Injection is a sterile solution of Levamisole
Apply to the plate 10 µl of each solution. After development,
Hydrochloride in Water for Injections. It may contain suitable
dry the plate in air and spray with potassium iodoplatinate
colouring agents.
solution. Any spot corresponding to 2,3-dihydro-6-
Levamisole Injection contains not less than 92.5 per cent and phenylimidazo[2,1-b]thiazole hydrochloride in the
not more than 107.5 per cent of the stated amount of levamisole chromatogram obtained with test solution is not more intense
hydrochloride, C11H12N2S,HCl. than the spot in the chromatogram obtained with reference
solution.
Identification
Other tests. Complies with the tests stated under Parenteral
A. Determine by thin-layer chromatography (2.4.17), coating Preparations (Injections).
the plate with silica gel G. Assay. To an accurately measured volume containing 0.75 g
Mobile phase. A mixture of 100 volumes of ethyl acetate, 10 of Levamisole Hydrochloride add 50 ml of water and 15 ml of
volumes of methanol and 1 volume of strong ammonia 2 M sodium hydroxide, extract with three quantities, each of
solution. 25 ml, 20 ml and 15 ml of dichloromethane, wash the combined
extracts with two quantities, each of 10 ml, of water and discard
Test solution. Dilute a volume of the injection to produce a
the washings. To the clear dichloromethane solution, after
solution containing 1.0 per cent w/v of Levamisole
drying with anhydrous sodium sulphate, add 50 ml of
Hydrochloride in methanol.
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
Reference solution. A 1.0 per cent w/v of levamisole acid, using 1-naphtholbenzein solution as indicator. Carry
hydrochloride RS in methanol. out a blank titration.

1538
IP 2007 LINCOMYCIN PREMIX

1 ml of 0.1 M perchloric acid is equivalent to 0.02408 g of Mobile phase. A mixture of 45 volumes of toluene, 8 volumes
C11H12N2S,HCl. of methanol and 4 volumes of anhydrous glacial acetic acid.
Storage. Store protected from light. Test solution. Dilute a volume of the preparation under
examination with methanol to produce a solution containing
1.0 per cent w/v of Levamisole Hydrochloride.
Levamisole Hydrochloride Veterinary Reference solution. A 0.025 per cent w/v of 2,3-dihydro-6-
Oral Solution phenylimidazo[2,1-b]thiazole hydrochloride RS in
methanol.
Levamisole Hydrochloride Veterinary Mixture;
Apply to the plate 50 µl of the test solution and 10 µl of the
Levamisole Veterinary Oral Solution; Levamisole reference solution. After development, dry the plate in air and
Veterinary Mixture spray with potassium iodoplatinate solution. Any spot
Levamisole Hydrochloride Veterinary Oral Solution is an corresponding to 2,3-dihydro-6-phenylimidazo[2,1-b]thiazole
aqueous solution of Levamisole Hydrochloride containing hydrochloride in the chromatogram obtained with the test
suitable stabilising agents. solution is not more intense than the spot in the chromatogram
obtained with the reference solution.
Levamisole Hydrochloride Veterinary Oral Solution contains
not less than 92.5 per cent and not more than 107.5 per cent of Other tests. Complies with the tests stated under Veterinary
the stated amount of levamisole hydrochloride, C11H12N2S,HCl. Oral Liquids.
Assay. Weigh accurately a quantity containing 0.75 g of
Identification
Levamisole Hydrochloride add 15 ml of 2 M sodium hydroxide,
A. Determine by thin-layer chromatography (2.4.17), coating extract with three quantities each of 25 ml, 20 ml and 15 ml of
the plate with silica gel G. dichloromethane, wash the combined extracts with two
Mobile phase. A mixture of 100 volumes of ethyl acetate, 10 quantities, each of 10 ml, of water and discard the washings.
volumes of methanol and 1 volume of strong ammonia To the clear dichloromethane solution, after drying with
solution. anhydrous sodium sulphate, add 50 ml of anhydrous glacial
acetic acid. Titrate with 0.1 M perchloric acid, using
Test solution. Dilute a volume of the preparation under
1-naphtholbenzein solution as indicator. Carry out a blank
examination with methanol to produce a solution containing
titration.
1.0 per cent w/v of Levamisole Hydrochloride.
1 ml of 0.1 M perchloric acid is equivalent to 0.02408 g of
Reference solution. A 1.0 per cent w/v of levamisole
C11H12N2S,HCl.
hydrochloride RS in methanol.
Apply to the plate 1 µl of each solution. After development,
dry the plate in air and spray with potassium iodoplatinate
solution. The principal spot in the chromatogram obtained Lincomycin Premix
with the test solution corresponds to that in the chromatogram Lincomycin Hydrochloride Premix.
obtained with the reference solution.
Lincomycin Premix contains Lincomycin Hydrochloride.
B. To a quantity containing 0.3 g of Levamisole Hydrochloride
add 10 ml of water and 6 ml of 1 M sodium hydroxide. Extract Lincomycin Premix contains not less than 90.0 per cent and
with 20 ml of dichloromethane, discard the aqueous layer and not more than 110.0 per cent of the stated amount of
wash the dichloromethane layer with 10 ml of water. Dry by lincomycin, C18H34N2O6S.
shaking with anhydrous sodium sulphate, filter and allow the
dichloromethane to evaporate at room temperature. The Identification
residue, after drying over phosphorus pentoxide at a pressure In the Assay, the chromatogram obtained with the test solution
of 1.5 to 2.5 kPa at a temperature not exceeding 40°, melts at (b) corresponds to the chromatogram obtained with the
about 59° (2.4.21). reference solution.
C. The solution is laevorotatory.
Tests
Tests Lincomycin B. Examine test solution (a) as described in the
2,3-Dihydro-6-phenylimidazo[2,1-b]thiazole hydrochloride. Assay but increasing the sensitivity by 8 to 10 times while
Determine by thin-layer chromatography (2.4.17), coating the recording the peak due to the trimethylsilyl derivative of
plate with silica gel G. lincomycin B, which is eluted immediately before the

1539
LITHIUM ANTIMONY THIOMALATE IP 2007

trimethylsilyl derivative of lincomycin. The area of the peak 5.1 per cent and not more than 5.7 per cent of Li, calculated on
due to the trimethylsilyl derivative of lincomycin B, after the dried, solvent-free basis.
correction for the sensitivity factor, is not more than 5 per cent Description. A pinkish white or creamy powder; hygroscopic.
of the area of the peak due to the trimethylsilyl derivative of
lincomycin. Identification
Other tests. Complies with the tests stated under Premixes. A. To 0.2 g dissolved in 5 ml of water add 2 ml of hydrochloric
Assay. Determine by gas chromatography (2.4.13). acid and 5 ml of sodium sulphide solution; a yellowish- orange
precipitate is produced which does not dissolve on addition
Solution A. Weigh accurately a quantity of the premix
of dilute ammonia solution.
containing about 90 mg of lincomycin, shake with 10.0 ml of
dimethylformamide and filter. B. When moistened with hydrochloric acid and introduced
on a platinum wire it imparts a red colour to a non-luminous
Test solution (a). Add 1 ml of a 1 per cent w/v solution of
flame.
tetraphenylcyclopentadienone (internal standard) in
dimethylformamide and 0.4 ml of a mixture of 9 volumes of Tests
N,O-bis(trimethylsilyl)acetamide and 1 volume of
trimethylchlorosilane to 1 ml of solution A, mix and allow to Appearance of solution. A 6 per cent w/v solution in carbon
stand for 15 minutes. dioxide-free water is clear (2.4.1), and not more intensely
coloured than reference solution RS3 (2.4.1).
Test solution (b). Add 1 ml of dimethylformamide and 0.4 ml
of a mixture of 9 volumes of N,O-bis(trimethylsilyl)acetamide pH (2.4.24). 9.0 to 10.5, determined in a 6 per cent w/v solution
and 1 volume of trimethylchlorosilane to 1 ml of solution A, in carbon dioxide-free water.
mix and allow to stand for 15 minutes. Assay. For antimony — Weigh accurately about 0.50 g , add
Reference solution. Add 1 ml of a 1 per cent w/v solution of 35 ml of water and swirl to dissolve. Add 5 g of ammonium
tetraphenylcyclopentadienone (internal standard) in persulphate, 10 ml of sodium hydroxide solution and 3 or 4
dimethylformamide and 0.4 ml of a mixture of 9 volumes of glass beads (approximately 0.5 cm diameter). Place a small
N,O-bis(trimethylsilyl)acetamide and 1 volume of funnel in the neck of the flask and boil gently for 20 minutes at
trimethylchlorosilane to 1 ml of a 1 per cent w/v solution of such a rate that the volume is not reduced appreciably. Cool,
lincomycin hydrochloride RS in dimethylformamide, mix and add through the funnel 0.25 ml of phenolphthalein solution
allow to stand for 15 minutes. and sufficient 0.1 M hydrochloric acid until the last trace of
pink colour disappears. Add 25 ml of a 10 per cent w/v solution
Chromatographic system of oxalic acid through the funnel and boil vigorously for 3
– a glass column 1.5 m x 3 mm, packed with silanised minutes. Rinse the funnel, with a small quantity of water,
diatomaceous support (100 to 120 mesh) coated with 3 remove it and add 5 ml of hydrochloric acid and 2 g of
per cent w/w of methyl silicone gum (such as SE 30), potassium iodide. Allow to stand for 10 minutes and boil until
– temperature: the solution becomes yellow and shows no further decrease
column 260°, in colour, but taking care to see that the volume is not reduced
inlet port and detector. 260° to 290°, to less than about 30 ml. Cool and remove a small drop of the
– flow rate. 30 ml per minute of the carrier gas. solution with a sealed capillary melting point tube and add to
Calculate the content of C18H34N2O6S. starch iodide paper. If a bluish colour is produced, add 1 drop
Labelling. The label states the strength in terms of the of 0.1 M sodium thiosulphate while swirling and again test
equivalent amount of lincomycin. with starch iodide paper. Repeat if necessary until a bluish
colour is no longer produced.
Add 5 g of sodium potassium tartrate, cool to about 15° to
20° and cautiously add small portions of sodium bicarbonate
Lithium Antimony Thiomalate until no further effervescence is produced. Add 2 to 4 g more
of sodium bicarbonate and titrate with 0.1 M iodine until the
LiOOC C CHCOOLi first permanent light yellow colour is produced.
3
, 9H2O
H2 1 ml of 0.1 M iodine is equivalent to 0.006088 g of Sb.
S Sb
For lithium — Weigh accurately about 0.2 g, dissolve in 50 ml
C12H9Li6O12S3Sb,9H2O Mol. Wt. 766.9
of glacial acetic acid . Titrate with 0.1 M perchloric acid,
Lithium Antimony Thiomalate contains not less than 15.5 per using 1 ml of crystal violet solution as indicator. Carry out a
cent and not more than 16.5 per cent of Sb and not less than blank titration.

1540
IP 2007 MAGNESIUM HYPOPHOSPHITE

1 ml of 0.1 M perchloric acid is equivalent to 0.000694 g of Li. loss by spurting due to effervescence) till alkaline to litmus
paper and titrate with 0.05 M iodine using 1 ml of starch
Storage. Store protected from light and moisture.
solution, added towards the end of the titration, as indicator.
1 ml of 0.05 M iodine is equivalent to 0.03834 g of
C12H9Li6O12S3Sb,9H2O.
Lithium Antimony Thiomalate Injection Storage. Store protected from light.
Lithium Antimony Thiomalate Injection is a sterile solution of
Lithium Antimony Thiomalate in Water for Injection containing
a suitable antimicrobial preservative. Magnesium Hypophosphite
Lithium Antimony Thiomalate Injection contains not less than
95.0 per cent and not more than 105.0 per cent of the stated
HO P Mg P OH , 6H2O
amount of lithium antimony thiomalate, C12H9Li6O12S3Sb,9H2O.
OH OH
Identification
Mg(H2PO2)2,6H2O Mol. Wt. 262.4
A. Dilute a volume containing 0.2 g of Lithium Antimony
Magnesium Hypophosphite contains not less than 98.5 per
Thiomalate to 5 ml with water. Add 2 ml of hydrochloric acid
cent and not more than 101.0 per cent of Mg(H2PO2)2,6H2O.
and 5 ml of sodium sulphide solution; a yellowish orange
precipitate is produced which does not dissolve on addition Description. Colourless crystals or white crystalline powder.
of dilute ammonia solution.
Identification
B. Dilute 0.2 ml of the injection under examination to 10 ml
with a 5 per cent w/v solution of sodium potassium tartrate. A. Gives the reactions of magnesium salts (2.3.1).
To 2 ml of the solution add sodium sulphide solution dropwise; B. Dissolve about 50 mg in 5 ml of water and add 0.5 ml of
a reddish orange precipitate is produced. The precipitate mercuric chloride solution; a white precipitate is produced.
dissolves on adding dilute sodium hydroxide solution.
C. Dissolve about 50 mg in 5 ml of water and acidify with
Tests sulphuric acid. Add 0.5 ml of cupric sulphate solution and
warm; a red precipitate is produced.
Appearance of solution. The solution is clear (2.4.1), and not
more intensely coloured than reference solution RS3 (2.4.1). Tests
pH (2.4.24). 9.0 to 10.5. Appearance of solution. A 5 per cent w/v solution is clear
Pyrogens. Complies with the test for pyrogens (2.2.8), using (2.4.1) and colourless (2.4.1).
per 1.5 kg of the rabbit’s weight, a volume containing 0.012 g Heavy metals (2.3.13). Dissolve 1.0 g in 20 ml of water, add 2 ml
of Lithium Antimony Thiomalate. of dilute hydrochloric acid and sufficient water to produce
Sterility (2.2.11). Complies with the test for sterility. 25 ml. The resulting solution complies with the limit test for
heavy metals, Method A (20 ppm).
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections). Chlorides (2.3.12). To 1 g add 200 ml of water and filter. 10 ml
of the filtrate complies with the limit test for chlorides (0.1 per
Assay. Dilute 10.0 ml with 25 ml of water, add 7.5 g of ammonium
cent).
persulphate and 16 ml of sodium hydroxide solution, boil
gently for 20 minutes, cool and add 0.5 ml of phenolphthalein Sulphates (2.3.17). 1 g complies with the limit test for sulphates
solution. Neutralise the solution with dilute hydrochloric acid (0.015 per cent).
and boil for 3 minutes. Add 50 ml of a 10 per cent w/v solution Assay. Weigh accurately about 0.2 g, dissolve in 50 ml of
of oxalic acid, 7.5 ml of hydrochloric acid and sufficient water, add 5 ml of strong ammonia-ammonium chloride
water to make up the volume, if necessary. Add 2 g of solution and titrate with 0.05 M disodium edetate using 0.1 g
potassium iodide to the hot solution, allow to stand for 10 of mordant black II mixture as indicator, until a blue colour is
minutes and boil until it acquires a pale yellow colour (about obtained.
10 minutes). Cool and remove the colour by adding 0.1 M
sodium thiosulphate using starch iodide solution as an 1 ml of 0.05 M disodium edetate is equivalent to 0.01312 g of
external indicator. Add 7.5 g of sodium potassium tartrate and Mg(H2PO2)2,6H2O.
dilute to 200 ml. Add sodium bicarbonate carefully (avoiding Storage. Store protected from moisture.

1541
MECLEOFENAMIC ACID IP 2007

Meclofenamic Acid Chromatographic system


– a stainless steel column 20 cm x 4 mm, packed with
particles of silica gel (10 µm) the surface of which has
been modified with chemically-bonded octadecasilyl
groups (such as Spherisorb ODS),
– mobile phase: a mixture of 75 volumes of methanol, 25
volumes of water and 1 volume of glacial acetic acid,
C14H11Cl2NO2 Mol. Wt. 296.2 – flow rate. 2 ml per minute,
– spectrophotometer set at 254 nm,
Meclofenamic acid is N-(2,6-dichloro-3-methylphenyl) – a 20 µl loop injector.
anthranilic acid.
Inject reference solution (a). The test is not valid unless the
Meclofenamic Acid contains not less than 98.5 per cent and column efficiency is not less than 4500 theoretical plates.
not more than 100.5 per cent of the stated amount of
C14H11Cl2NO2, calculated on the dried basis. Inject reference solution (b). The area of the peak immediately
preceding the peak due to meclofenamic acid is not more than
Description. A white or almost white, crystalline powder.
one-seventh of the area of the peak due to the internal standard.
Identification The area of any other peak is not more than the area of the
peak due to the internal standard.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with meclofenamic Heavy metals (2.3.13). 1.0 g complies with the limit test for
acid RS or with the reference spectrum of meclofenamic acid. heavy metals, Method D (20 ppm).

B. When examined in the range 220 nm to 360 nm (2.4.7), a Sulphated ash (2.3.18). Not more than 0.1 per cent.
0.002 per cent w/v solution in 0.1 M sodium hydroxide shows Loss on drying (2.4.19). Not more than 0.5 per cent, determined
absorption maxima at about 279 nm, and 371 nm; absorbance on 1.0 g by drying in an oven at 105°.
at about 279 nm, about 0.45, and at about 317 nm, about 0.33.
Assay. Weigh accurately about 0.6 g, dissolve in 100 ml of
C. Dissolve 25 mg in 15 ml of dichloromethane; the solution warm ethanol previously neutralised to phenol red solution
exhibits a strong blue fluorescene when examined under and titrate with 0.1 M sodium hydroxide, using phenol red
ultraviolet light. solution as indicator.
D. Dissolve 1 mg in 2 ml of sulphuric acid and add 0.05 ml of 1 ml of 0.1 M sodium hydroxide is equivalent to 0.02962 g of
0.02 M potassium dichromate; an intense purple colour is C14H11Cl2NO2.
produced, which rapidly fades to purple brown.
Storage. Store protected from moisture.
Tests
Appearance of solution. A 5.0 per cent w/v solution in 1 M
sodium hydroxide is not more opalescent than reference Mepyramine Injection
suspension OS2 (2.4.1) and is not more intensely coloured
Mepyramine Maleate Injection; Pyrilamine Maleate
than reference solution BYS5 (2.4.1).
Injection; Pyrilamine Injection
Light absorption (2.4.7). Absorbance of a 0.002 per cent w/v
solution in 0.01 M methanolic hydrochloric acid at the Mepyramine Injection is a sterile solution of Mepyramine
maximum at about 279 nm, not less than 0.400 and not more Maleate in Water for Injections.
than 0.445, and at the maximum at about 335 nm, not less than Mepyramine Injection contains not less than 95.0 per cent
0.440 and not more than 0.490. and not more than 105.0 per cent of the stated amount of
Related substances. Determine by liquid chromatography mepyramine maleate, C17H23N3O,C4H4O4.
(2.4.14). Description. Colourless or almost colourless solution.
Test solution. Dissolve 1.0 g of the substance under
examination in 100 ml of ethanol. Identification
Reference solution (a). A 0.0035 per cent w/v solution of ethyl A. To a volume containing 0.1 g of Mepyramine Maleate add
meclofenamate RS (internal standard). in ethanol. 2 ml of 5 M sodium hydroxide and shake with three quantities,
Reference solution (b). A solution containing 1.0 per cent w/ each of 3 ml, of ether. Warm the aqueous layer in a water-bath
v of the substance under examination and 0.0035 per cent w/v for 10 minutes with 2 ml of bromine solution, heat to boiling,
of ethyl meclofenamate RS (internal standard) in ethanol. cool, and add 0.2 ml to a solution of 10 mg of resorcinol in 3 ml

1542
IP 2007 MONOSULFIRAM SOAP

of sulphuric acid; a blue-black colour develops on heating C. Boil 0.1 g with 2 M hydrochloric acid; hydrogen sulphide
for 15 minutes in a water-bath. is evolved which has a characteristic odour and turns filter
paper treated with lead acetate solution, black.
B. Dilute a volume containing 20 mg of mepyramine maleate to
2 ml with water, add 1 ml of cyanogen bromide solution and 5 Tests
ml of a 2 per cent w/v solution of potassium hydrogen
phthalate, mix, allow to stand for 15 minutes and add 1 ml of a Freezing point (2.4.11). 28.5° to 32.0°.
4 per cent solution of aniline in ethanol (95 per cent); a
Related substances. Determine by thin-layer chromatography
yellow colour is produced.
(2.4.17), coating the plate with silica gel GF254.
Tests NOTE – Carry out the test in subdued light.
pH (2.4.24). 5.5 to 6.5. Mobile phase. A mixture of 70 volumes of n-hexane and 30
volumes of butyl acetate.
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections). Test solution. Dissolve 2.5 g of the substance under
examination in 100 ml of ethyl acetate.
Assay. To an accurately measured volume containing about
25 mg of mepyramine maleate add sufficient 0.01 M Reference solution (a). A 0.125 per cent w/v solution of
hydrochloric acid to produce 100.0 ml. Dilute 10.0 ml of this disulfiram RS in ethyl acetate.
solution to 100.0 ml with 0.01 M hydrochloric acid and measure Reference solution (b). A 0.050 per cent w/v solution of the
the absorbance of the resulting solution at the maximum at substance under examination in ethyl acetate.
about 316 nm (2.4.7). Calculate the content of
C17H23N3O,C4H4O4, taking 206 as the specific absorbance at Apply to the plate 5 µl of each solution. After development,
316 nm. dry the plate in air and examine in ultraviolet light at 254 nm. In
the chromatogram obtained with the test solution any
secondary spot is not more intense than the spot in the
chromatogram obtained with reference solution (a) and any
Monosulfiram other spot, apart from the principal spot, is not more intense
than the spot in the chromatogram obtained with reference
Sulfiram solution (b).
H3 C CH3 Water (2.3.43). Not more than 1.0 per cent, determined on 1 g.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
H3 C N S N CH3
Assay. Weigh accurately about 0.35 g, dissolve in 8 ml of
S S nitrogen-free sulphuric acid and carry out the method for the
C10H20N2S3 Mol. Wt. 264.5 determination of nitrogen ( 2.3.30).

Monosulfiram is bis(diethylthiocarbamoyl)sulphide. 1 ml of 0.05 M sulphuric acid is equivalent to 0.01322 g of


C10H20N2S3.
Monosulfiram contains not less than 98.0 per cent and not
more than 101.0 per cent of C10H20N2S3, calculated on the Storage. Store protected from light.
anhydrous basis.
Description. A yellow or yellowish-brown soft solid; odour,
sulphurous.
Monosulfiram Soap
Monosulfiram Soap contains not less than 5 per cent w/w of
Identification monosulfiram in a toilet soap basis which may be perfumed.
A. When examined in the range 230 nm to 360 nm (2.4.7), a Monosulfiram Soap contains not less than 90.0 per cent and
2-cm layer of a 0.001 per cent w/v solution in methanol shows not more than 110.0 per cent of the stated amount of
a well-defined absorption maximum only at about 281 nm; monosulfiram, C10H20N2S3.
absorbance at about 281 nm, about 1.3.
Identification
B. Dissolve 0.1 g in a mixture of 0.15 ml of a 1 per cent w/v
solution of cupric sulphate and 5 ml of ethanol (95 per cent), In the test for Related substances, the principal spot in the
evaporate on a water-bath and dissolve the residue in chromatogram obtained with test solution (b) corresponds to
dichloromethane; a deep yellowish brown colour is produced. that in the chromatogram obtained with reference solution (c).

1543
MONOSULFIRAM SOLUTION IP 2007

Tests Chromatographic system


– a glass column 1.5 m x 4 mm, packed with 2 per cent w/w
Related substances. Determine by thin-layer chromatography of methyl silicone gum on acid-washed, silanised
(2.4.17), coating the plate with silica gel GF254. diatomaceous support (80 to 100 mesh) (such as SE 30),
NOTE — Carry out the test in subdued light. – temperature:
column 180°,
Mobile phase. A mixture of 70 volumes of n-hexane and 30 inlet port 180° and detector 280°,
volumes of butyl acetate. – flow rate. 30 ml per minute of the carrier gas.
Test solution (a). Shake a quantity of the finely shredded Calculate the content of C10H20N2S3.
soap containing 20 mg of Monosulfiram with 10 ml of
dichloromethane, filter and wash the filtrate with Labelling. The label states (1) the proportion of Monosulfiram
dichloromethane. Evaporate the combined filtrate and in the preparation; (2) the method of use of the preparation.
washings just to dryness at room temperature in a current of
nitrogen and dissolve the residue in 1 ml of ethanol (95 per
cent).
Monosulfiram Solution
Test solution (b). Dilute 0.5 ml of test solution (a) to 10 ml with Monosulfiram Solution is a solution of Monosulfiram in
ethanol (95 per cent). Ethanol (95 per cent) containing a suitable dispersing agent.
In making Monosulfiram Solution the ethanol (95 per cent)
Reference solution (a). A 0.10 per cent w/v solution of
may be replaced by Industrial Methylated Spirit provided that
disulfiram RS in ethanol (95 per cent).
the statutory requirements governing the use of Industrial
Reference solution (b). A 0.040 per cent w/v solution of Methylated Spirit are observed.
monosulfiram RS in ethanol (95 per cent). Monosulfiram Solution contains not less than 94.0 per cent
Reference solution (c). A 0.10 per cent w/v solution of and not more than 106.0 per cent of the stated amount of
monosulfiram RS in ethanol (95 per cent). monosulfiram, C10H20N2S3.
Apply to the plate 5 µl of each solution. After development, Description. Clear, bright, deep reddish-brown liquid; crystals
dry the plate in air and examine in ultraviolet light at 254 nm. In from which may deposit slowly at low temperatures but
the chromatogram obtained with test solution any spot running dissolve on warming. Yields a pale yellow dispersion on
ahead of the principal spot and corresponding in position to dilution with water.
disulfiram is not more intense than the spot in the chromatogram
Identification
obtained with reference solution (a) and any spot running
behind the principal spot is not more intense than the spot in In the test for Related substances, the principal spot in the
the chromatogram obtained with reference solution (b). Ignore chromatogram obtained with test solution (b) corresponds to
any subsidiary spots due to the soap basis which may also be that in the chromatogram obtained with reference solution (c).
observed ahead of the principal spot in the chromatogram
obtained with test solution (a). Tests
Assay. Protect the solutions from light throughout the assay. Related substances. Determine by thin-layer chromatography
(2.4.17), coating the plate with silica gel GF254.
Determine by gas chromatography (2.4.13).
NOTE – Carry out the test in subdued light.
Test solution (a). Weigh accurately a quantity of the finely
Mobile phase. A mixture of 70 volumes of n-hexane and 30
shredded soap containing about 0.25 g of Monosulfiram, shake
volumes of butyl acetate.
for 10 minutes with 50 ml of dimethylformamide, centrifuge
and use the supernatant liquid. Test solution (a). Dilute a quantity of the solution under
examination with ethanol (95 per cent) so as to contain of 2.0
Test solution (b). Weigh accurately a quantity of the finely per cent w/v of Monosulfiram.
shredded soap containing about 0.25 g of Monosulfiram, shake
for 10 minutes with 50 ml of dimethylformamide containing Test solution (b). Dilute 0.5 ml of test solution (a) to 10 ml with
0.125 g of N-phenylcarbazole (internal standard), centrifuge ethanol (95 per cent).
and use the supernatant liquid. Reference solution (a). A 0.10 per cent w/v solution of
disulfiram RS in ethanol (95 per cent).
Reference solution. A solution containing 0.5 per cent w/v of
monosulfiram RS and 0.25 per cent w/v of N-phenylcarbazole Reference solution (b). A 0.040 per cent w/v solution of
(internal standard) in dimethylformamide. monosulfiram RS in ethanol (95 per cent).

1544
IP 2007 NANDROLONE LAURATE

Reference solution (c). A 0.10 per cent w/v solution of Nandrolone Laurate is 3-oxoestr-4-en-17β-yl-dodecanoate.
monosulfiram RS in ethanol (95 per cent).
Nandrolone Laurate contains not less than 97.0 per cent and
Apply to the plate 5 µl of each solution. After development, not more than 103.0 per cent of C30H48O3, calculated on the
dry the plate in air and examine in ultraviolet light at 254 nm. In dried basis.
the chromatogram obtained with the test solution any spot
Description. A white to creamy white, crystalline powder;
running ahead of the principal spot and corresponding in
odour, faint and characteristic.
position to disulfiram is not more intense than the spot in the
chromatogram obtained with reference solution (a) and any Identification
spot running behind the principal spot is not more intense
than the spot in the chromatogram obtained with reference A. Determine by infrared absorption spectrophotometry (2.4.6).
solution (b). Compare the spectrum with that obtained with nandrolone
Assay. Protect the solutions from light throughout the assay. laurate RS or with the reference spectrum of nandrolone
laurate.
Determine by gas chromatography (2.4.13).
B. Determine by thin-layer chromatography (2.4.17), coating
Test solution (a). Dilute the solution under examination in the plate with silica gel GF254, surface of which has been
dimethylformamide containing the equivalent of 0.5 per cent modified by chemically bonded octadecylsilyl groups (such
w/v of Monosulfiram. as Whatman KC 18F plates).
Test solution (b). Weigh accurately a quantity of the finely Mobile phase. A mixture of 60 volumes of 2-propanol, 40
shredded soap containing about 0.25 g of Monosulfiram, shake volumes of acetonitrile and 20 volumes of water.
for 10 minutes with 50 ml of dimethylformamide containing
0.125 g of N-phenylcarbazole (internal standard), centrifuge Test solution. Dissolve 0.5 g of the substance under
and use the supernatant liquid. examination in dichloromethane.

Reference solution. A solution containing 0.5 per cent w/v of Reference solution (a). A 0.5 per cent w/v of nandrolone
monosulfiram RS and 0.25 per cent w/v of N-phenylcarbazole laurate RS in dichloromethane.
(internal standard) in dimethylformamide. Reference solution (b). A mixture of equal volumes of the test
Chromatographic system solution and reference solution (a)
– a glass column 1.5 m x 4 mm, packed with 2 per cent w/w Apply to the plate 5 µl of each solution. After development,
of methyl silicone gum on acid-washed, silanised dry the plate in air until the odour of the solvent is no longer
diatomaceous support (80 to 100 mesh) (such as SE 30), detectable and heat at 100° for 10 minutes. Allow to cool and
– temperature: examine in ultraviolet light at 254 nm. The principal spot in the
column 180°, chromatogram obtained with the test solution corresponds
inlet port 180° and detector 280°, to that in the chromatogram obtained with the reference
– flow rate. 30 ml per minute of the carrier gas. solution (a). The test is not valid unless the principal spot in
Calculate the content of C10H20N2S3. the chromatogram obtained with reference solution (b) appears
as a single, compact spot.
Labelling. The label states (1) the percentage w/w of
monosulfiram; (2) the method of use of the preparation. C. Melts at about 47° (2.4.21).

Tests
Nandrolone Laurate Specific optical rotation (2.4.22). +31.0° to +35.0°, determined
in a freshly prepared 2 per cent w/v solution in dioxan.
O Nandrolone. Determine by thin-layer chromatography (2.4.17),
coating the plate with silica gel G.
H3C O CH2(CH2)9CH3
Mobile phase. A mixture of 70 volumes of n-heptane and 30
H H volumes of acetone.
Test solution. Dissolve 1.5 g of the substance under
H H examination in dichloromethane.
O
Reference solution. A 0.030 per cent w/v of nandrolone RS in
C30H48O3 Mol. Wt. 456.7 dichloromethane.

1545
NANDROLONE LAURATE INJECTION IP 2007

Apply to the plate 1 µl of each solution. After development, chromatogram obtained with the test solution corresponds
dry the plate in air until the odour of the solvent is no longer to that in the chromatogram obtained with the reference
detectable, spray with a 10 per cent v/v solution of sulphuric solution (a). The test is not valid unless the principal spot in
acid in ethanol (95 per cent), heat at 105° for 30 minutes and the chromatogram obtained with reference solution (b) appears
examine in ultraviolet light at 365 nm. Any spot in the as a single, compact spot.
chromatogram obtained with the test solution corresponding
to nandrolone is not more intense than the spot in the Tests
chromatogram obtained with the reference solution.
Other tests. Complies with the tests stated under Parenteral
Sulphated ash (2.3.18). Not more than 0.1 per cent. Preparations (Injections).
Loss on drying (2.4.19). Not more than 0.5 per cent, determined Assay. To an accurately measured volume containing about
on 1.0 g by drying over phosphorus pentoxide at a pressure 0.1 g of Nandrolone Laurate add sufficient dichloromethane
not exceeding 0.7 kPa for 24 hours. to produce 100.0 ml. Dilute 3.0 ml of the resulting solution to
Assay. Weigh accurately about 0.1 g, dissolve in sufficient 50.0 ml with dichloromethane. To 5.0 ml of this solution add
ethanol to produce 100.0 ml and dilute 10.0 ml to 100.0 ml with 10 ml of isoniazid solution and sufficient methanol to produce
ethanol. Dilute 10.0 ml of this solution to 100.0 ml with ethanol 20.0 ml. Allow to stand for 45 minutes and measure the
and measure the absorbance of the resulting solution at the absorbance of the resulting solution at the maximum at about
maximum at about 240 nm (2.4.7). Calculate the content of 380 nm (2.4.7), using as the blank 5 ml of dichloromethane
C30H48O3 taking 380 as the specific absorbance at 240 nm. treated in a similar manner. Calculate the content of C30H48O3
from the absorbance obtained by repeating the procedure
Storage. Store protected from light. using a suitable quantity of nandrolone RS.
1 mg of C18H26O2 is equivalent to 0.001664 g of C30H48O3.
Storage. Store protected from light.
Nandrolone Laurate Injection
Nandrolone Laurate Injection is a sterile solution of Nandrolone
Laurate in Ethyl Oleate or other suitable ester, in a suitable
fixed oil, or in any mixture of these. Niclosamide Veterinary Oral Powder
Nandrolone Laurate Injection contains not less than 92.5 per Niclosamide Dispersible Powder for Veterinary Use
cent and not more than 107.5 per cent of the stated amount of Niclosamide Veterinary Oral Powder contains Niclosamide with
nandrolone laurate, C30H48O3. suitable auxiliary substances.
Identification Niclosamide Veterinary Oral Powder contains not less than
97.0 per cent and not more than 103.0 per cent of the stated
Determine by thin-layer chromatography (2.4.17), coating the amount of niclosamide, C13H8Cl2N2O4.
plate with silica gel GF254, surface of which has been modified
by chemically bonded octadecylsilyl groups (such as Identification
Whatman KC 18F plates).
Heat 50 mg with 5 ml of 1 M hydrochloric acid and 0.1 g of
Mobile phase. A mixture of 60 volumes of 2-propanol, 40 zinc powder in a water-bath for 10 minutes, cool and filter. To
volumes of acetonitrile and 20 volumes of water. the filtrate add 0.5 ml of a 1 per cent w/v solution of sodium
Test solution. Dilute a suitable volume with dichloromethane nitrite and allow to stand for 10 minutes. Add 2 ml of a 2 per
to produce a solution containing 0.5 per cent w/v of cent w/v solution of ammonium sulphamate, shake, allow to
Nandrolone Laurate . stand for 10 minutes and add 2 ml of a 0.5 per cent w/v solution
of N-(1-naphthyl)ethylenediamine dihydrochloride; a deep
Reference solution (a). A 0.5 per cent w/v of nandrolone
red colour is produced.
laurate RS in dichloromethane.
Reference solution (b). A mixture of equal volumes of the test Tests
solution and reference solution (a)
2-Chloro-4-nitroaniline. Boil a quantity containing 0.10 g of
Apply to the plate 5 µl of each solution. After development, Niclosamide with 20 ml of methanol for 2 minutes, cool, add
dry the plate in air until the odour of the solvent is no longer sufficient 1 M hydrochloric acid to produce 50 ml and filter.
detectable and heat at 100° for 10 minutes. Allow to cool and To 10 ml of the filtrate add 0.5 ml of a 0.5 per cent w/v solution
examine in ultraviolet light at 254 nm. The principal spot in the of sodium nitrite and allow to stand for 10 minutes. Add 1 ml

1546
IP 2007 NITROXYNIL INJECTION

of a 2 per cent w/v solution of ammonium sulphamate, shake, C. When heated with sulphuric acid, iodine vapours are
allow to stand for 10 minutes and add 1 ml of a 0.5 per cent evolved.
w/v solution of N-(1-naphthyl)ethylenediamine dihydro-
D. Melting range (2.4.21). 136° to 139°.
chloride. The colour produced is not more than that produced
by simultaneously treating 10 mg of 2-chloro-4-nitroaniline Tests
in the same manner.
Inorganic iodide. To 0.40 g add 0.35 g of N-methylglucamine
5-Chlorosalicylic acid. Boil a quantity containing 0.50 g of
and 10 ml of water. Shake to dissolve and add sufficient water
Niclosamide with 10 ml of water for 2 minutes, cool and filter.
to produce 50 ml. To 10 ml of the resulting solution add 4 ml of
To the filtrate add a few drops of ferric chloride solution; no
1 M sulphuric acid and extract with three quantities, each of
red or violet colour is produced.
10 ml, of dichloromethane. Add to the aqueous extract 1 ml of
Loss on drying (2.4.19). Not more than 1.0 per cent, determined hydrogen peroxide solution (100 vol) and 1 ml of
on 1 g by drying in an oven at 105° for 4 hours. dichloromethane, shake for 2 minutes and allow to separate.
Other tests. Complies with the tests stated under Veterinary Any purple colour in the dichloromethane layer is not more
Oral Powders. intense than that obtained by adding 2 ml of a 0.0026 per cent
w/v solution of potassium iodide to a mixture of 4 ml of 1 M
Assay. Weigh accurately a quantity containing about 0.3 g of sulphuric acid and 8 ml of water, adding 10 ml of
Niclosamide, dissolve in 60 ml of dimethylformamide with the dichloromethane, shaking for 2 minutes, adding to the
aid of gentle heat, cool. Titrate with 0.1 M aqueous layer 1 ml of hydrogen peroxide solution (100 vol)
tetrabutylammonium hydroxide, maintaining a stream of and 1 ml of dichloromethane, shaking for 2 minutes and
nitrogen through the solution throughout the titration, and allowing to separate (500 ppm).
determining the end-point potentiometrically (2.4.25). Carry
out a blank titration. Sulphated ash (2.3.18). Not more than 0.1 per cent.

1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to Loss on drying (2.4.19). Not more than 1.0 per cent, determined
0.03271 g of C13H8Cl2N2O4. on 1 g by drying in an oven at 105° for 4 hours.
Assay. Carry out the oxygen flask method for iodine (2.3.34),
using 25 mg.
1 ml of 0.02 M sodium thiosulphate is equivalent to 0.0009667
Nitroxynil g of C7H3IN2O3.

OH Storage. Store protected from light.


I NO2

Nitroxynil Injection
CN Nitroxynil Injection is a sterile solution of the N-ethylglucamine
salt of Nitroxynil in Water for Injections.
C7H3IN2O3 Mol. Wt. 290.0
Nitroxynil Injection contains not less than 95.0 per cent and
Nitroxynil is 4-hydroxy-3-iodo-5-nitrobenzonitrile. not more than 105.0 per cent of the stated amount of nitroxynil,
Nitroxynil contains not less than 98.0 per cent and not more C7H3IN2O3.
than 101.0 per cent of C7H3IN2O3, calculated on the dried basis.
Identification
Description. A yellow to yellowish brown powder.
A. When examined in the range 230 nm to 360 nm (2.4.7), of the
Identification final solution obtained in the Assay exhibits a maximum at
about 271 nm.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with nitroxynil RS B. Heat 0.5 ml with 3 ml of sulphuric acid; iodine vapours are
or with the reference spectrum of nandrolone nitroxynil. evolved.
B. When examined in the range 220 nm to 360 nm (2.4.7), a Tests
0.002 per cent w/v solution in 0.01 M sodium hydroxide
exhibits maxima at about 225 nm and at about 271 nm; pH (2.4.24). 5.0 to 7.0, determined by using a 20 per cent w/v
absorbance at about 271 nm, about 1.3. solution of N-ethylglucamine hydrochloride instead of a

1547
OXFENDAZOLE IP 2007

saturated solution of potassium chloride as the liquid junction and dry at 105° at a pressure not exceeding 2.7 kPa. The residue
solution. complies with the following test.
Inorganic iodide. To a volume containing 0.4 g of Nitroxynil Determine by infrared absorption spectrophotometry (2.4.6).
add 0.35 g of N-methylglucamine and dilute to 100 ml with Compare the spectrum with that obtained with oxfendazole
water. To 10 ml of the diluted solution add 4 ml of 1 M sulphuric RS or with the reference spectrum of oxfendazole.
acid and extract with three quantities, each of 10 ml, of
B. When examined in the range 220 nm to 360 nm (2.4.7), a
dichloromethane. Add to the aqueous extract 1 ml of hydrogen
0.001 per cent w/v solution in methanol exhibits two maxima
peroxide solution (100 vol) and 1 ml of dichloromethane,
at about 228 nm and about 297 nm; absorbances at about 228
shake for 2 minutes and allow to separate. Any purple colour
nm, about 1.4 and at about 297 nm, about 0.55.
in the dichloromethane layer is not more intense than that
obtained by adding 2 ml of a 0.0026 per cent w/v solution of Tests
potassium iodide to a mixture of 4 ml of 1 M sulphuric acid
and 8 ml of water, adding 10 ml of dichloromethane, shaking Related substances. Determine by thin-layer chromatography
for 2 minutes, adding to the aqueous layer 1 ml of hydrogen (2.4.17), coating the plate with silica gel G.
peroxide solution (100 vol) and 1 ml of dichloromethane, Solvent mixture. 40 volumes of ethyl acetate and 10 volumes
shaking for 2 minutes and allowing to separate (500 ppm) (0.1 of glacial acetic acid.
per cent w/v of iodide).
Mobile phase. A mixture of 95 volumes of ethyl acetate and 5
Other tests. Complies with the tests stated under Parenteral volumes of glacial acetic acid.
Preparations (Injections).
Test solution. Dissolve 0.5 g of the substance under
Assay. To an accurately measured volume containing about examination in 100 ml of solvent mixture.
1.7 g of Nitroxynil add sufficient 0.01 M sodium hydroxide to
produce 500.0 ml. Dilute 20.0 ml of this solution to 500.0 ml Reference solution (a). A 0.010 per cent w/v solution of the
with 0.01 M sodium hydroxide. To 5.0 ml of this solution add substance under examination in solvent mixture.
sufficient 0.01 M sodium hydroxide to produce 100.0 ml and Reference solution (b). A 0.0050 per cent w/v solution of methyl
measure the absorbance of the resulting solution at the 5-phenylthio-1H-benzimidazol-2-yl carbamate RSin solvent
maximum at about 271 nm (2.4.7). Calculate the content of mixture.)
C7H3IN2O3 taking 660 as the specific absorbance at 271 nm.
Apply to the plate 20 µl of each solution. After development,
Storage. Store protected from light. dry the plate in air and examine in ultraviolet light at 254 nm.
Any spot corresponding to methyl 5-phenylthio-1H-
benzimidazol-2-yl carbamate in the chromatogram obtained
Oxfendazole with test solution is not more intense than the spot in the
chromatogram obtained with reference solution (b). Any other
secondary spot in the chromatogram obtained with test
solution is not more intense than the spot in the chromatogram
obtained with reference solution (a).
Sulphated ash (2.3.18). Not more than 0.2 per cent.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
C15H13N3O3S Mol. Wt. 315.4 on 1 g by drying in an oven at 105° for 2 hours at a pressure
not exceeding 0.7 kPa.
Oxfendazole is methyl 5-(phenylsulphinyl)-2-
benzimidazolecarbamate. Assay. Weigh accurately about 0.3 g, dissolve in 20 ml of
glacial acetic acid, add 3 g of potassium iodide and 1 ml of
Oxfendazole contains not less than 97.0 per cent and not more
acetyl chloride and stir for 10 minutes. Add 50 ml of 1 M
than 100.5 per cent of C15H13N3O3S, calculated on the dried
hydrochloric acid and 10 ml of dichloromethane and titrate
basis.
immediately with 0.1 M sodium thiosulphate, shaking after
Description. A white or almost white powder; odour, slight each addition, until the dichloromethane layer is colourless.
and characteristic. Repeat the operation omitting the substance under
examination; the difference between the titrations represents
Identification the amount of sodium thiosulphate required.
A. Dissolve 0.1 g in 50 ml of methanol, evaporate to a volume 1 ml of 0.1 M sodium thiosulphate is equivalent to 0.01577 g
of about 2 ml, cool, filter, wash the residue with 2 ml of water of C15H13N3O3S.

1548
IP 2007 OXYCLOZANIDE

Storage. Store protected from light. benzimidazol-2-yl carbamate in the chromatogram obtained
with test solution is not more intense than the spot in the
chromatogram obtained with reference solution (b). Any other
secondary spot in the chromatogram obtained with test
Oxfendazole Veterinary Oral solution is not more intense than the spot in the chromatogram
Suspension obtained with reference solution (a).
Oxfendazole Veterinary Mixture; Oxfendazole Mixture; Other tests. Complies with the tests stated under Veterinary
Oxfendazole Oral Suspension Oral Liquids.
Oxfendazole Veterinary Oral Suspension is an aqueous Assay. Weigh accurately a quantity of the well-mixed
suspension of Oxfendazole containing suitable suspending suspension containing about 0.1 g of Oxfendazole and disperse
or dispersing agents. in 15 ml of water. Add 200 ml of methanol and mix in an
ultrasonic bath for 15 minutes, cool, add sufficient methanol
Oxfendazole Veterinary Oral Suspension contains not less than
to produce 500.0 ml and filter. Dilute 4.0 ml of the filtrate to
90.0 per cent and not more than 110.0 per cent of the stated
100.0 ml with methanol and measure the absorbance of the
amount of oxfendazole, C15H13N3O3S.
resulting solution at the maximum at about 296 nm (2.4.7).
Identification Calculate the content of C15H13N3O3S taking 550 as the specific
absorbance at 296 nm.
Shake a quantity containing 0.1 g of Oxfendazole with 50 ml of Determine the weight per ml of the suspension (2.4.29), and
methanol for 15 minutes, centrifuge, evaporate the supernatant calculate the content of oxfendazole, weight in volume.
liquid to a volume of about 2 ml, cool, filter and wash the
residue with 2 ml of water and dry at 105° for 1 hour at a
pressure not exceeding 2.7 kPa. The residue complies with the
following tests. Oxyclozanide
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with oxfendazole Cl
RS or with the reference spectrum of oxfendazole.
OH O
B. When examined in the range 220 nm to 360 nm (2.4.7), a Cl
0.001 per cent w/v solution in 1 M hydrochloric acid exhibits N Cl
three maxima, at about 226, 284 and 291 nm. H OH
Cl
Tests Cl
pH (2.4.24). 4.3 to 5.3.
C13H6Cl5NO3 Mol. Wt. 401.5
Related substances. Determine by thin-layer chromatography
Oxyclozanide is 2,3,5-trichloro-N-(3,5-dichloro-2-
(2.4.17), coating the plate with silica gel G.
hydroxyphenyl)-6-hydroxybenzamide.
Solvent mixture. 40 volumes of ethyl acetate and 10 volumes
Oxyclozanide contains not less than 98.0 per cent and not
of glacial acetic acid.
more than 101.0 per cent of C13H6Cl5NO3, calculated on the
Mobile phase. A mixture of 95 volumes of ethyl acetate and 5 dried basis.
volumes of glacial acetic acid.
Description. A pale cream to cream-coloured powder.
Test solution. Shake a quantity containing 0.1 g of Oxfendazole
with 20 ml of solvent mixture and filter. Identification
Reference solution (a). Dilute 1 volume of test solution to 50 A. Determine by infrared absorption spectrophotometry (2.4.6).
volumes with the solvent mixture. Compare the spectrum with that obtained with oxyclozanide
Reference solution (b). A 0.0050 per cent w/v solution of methyl RS or with the reference spectrum of oxyclozanide.
5-phenylthio-1H-benzimidazol-2-yl carbamate RSin solvent B. When examined in the range 230 nm to 360 nm (2.4.7), a
mixture.) 0.003 per cent w/v solution in 1 M methanolic hydrochloric
Apply to the plate 20 µl of each solution. After development, acid exhibits a maximum only at about 300 nm; absorbance at
dry the plate in air and examine in ultraviolet light at 254 nm. about 300 nm, about 0.76.
Any spot corresponding to methyl 5-phenylthio-1H- C. Melting range (2.4.21). 208° to 211°.

1549
OXYCLOZANIDE VETERINARY ORAL SUSPENSION IP 2007

Tests Oxyclozanide Veterinary Oral


Ionisable chlorine. Dissolve 2 g in 100 ml of methanol, add 10 Suspension
ml of 1.5 M nitric acid and titrate with 0.1 M silver nitrate,
Oxyclozanide Oral Suspension; Oxyclozanide
determining the end-point potentiometrically (2.4.25). Not more
Suspension; Oxyclozanide Mixture; Oxyclozanide
than 1.4 ml is required (0.25 per cent).
Drench
Related substances. Determine by liquid chromatography
Oxyclozanide Veterinary Oral Suspension is an aqueous
(2.4.14).
suspension of Oxyclozanide containing suitable suspending
Test solution. A 0.1 per cent w/v solution of the substance or dispersing agents.
under examination prepared by dissolving it in a suitable
Oxyclozanide Veterinary Oral Suspension contains not less
volume of methanol and slowly diluting with water containing
than 95.0 per cent and not more than 105.0 per cent of the
0.1 per cent v/v of phosphoric acid to give a solution
stated amount of oxyclozanide, C13H6Cl5NO3.
containing about the same proportion of methanol to water as
in the mobile phase. Identification
Reference solution. Dilute 1 ml of test solution to 100 ml with In test A for Related substances, the principal spot in the
the mobile phase. chromatogram obtained with 10 ml of test solution
Chromatographic system corresponds to that in the chromatogram obtained with
– a stainless steel column 20 cm x 5 mm, packed with reference solution (b).
octadecylsilane bonded to porous silica (5 µm) (such as
Hypersil ODS),
Tests
– mobile phase: a filtered and degassed mixture of 62 Related substances. A. Determine by thin-layer
volumes of methanol and 38 volumes of water containing chromatography (2.4.17), coating the plate with silica gel G.
0.1 per cent v/v of phosphoric acid,
Mobile phase. A mixture of 60 volumes of light petroleum (
– flow rate. 2 ml per minute,
60° to 80°), 20 volumes of acetone and 5 volumes of glacial
– spectrophotometer set at 300 nm,
acetic acid.
– a 20 µl loop injector.
Test solution. Dilute a quantity with acetone to contain 1.0
Inject alternatively test solution and the reference solution. In per cent w/v of Oxyclozanide, centrifuge and use the
the chromatogram obtained with test solution the area of any supernatant liquid.
secondary peak with a retention time less than that of the
principal peak is not more than one-third of the area of the Reference solution (a). A 0.050 per cent w/v solution of
principal peak in the chromatogram obtained with reference 3,5,6-trichloro- 2-hydroxybenzoic acid RS in acetone.
solution and the area of any secondary peak with a retention Reference solution (b). A 1.0 per cent w/v solution of
time greater than that of the principal peak is not more than oxyclozanide RS in acetone.
the area of the principal peak in the chromatogram obtained Apply to the plate 40 µl and 10 µl of test solution, 4 µl of
with reference solution. reference solution (a) and 10 µl of reference solution (b). After
Sulphated ash (2.3.18). Not more than 0.2 per cent. development, dry the plate in air and spray with a 3 per cent w/
v solution of ferric chloride in methanol. In the chromatogram
Loss on drying (2.4.19). Not more than 1.0 per cent, determined obtained with 40 µl of test solution any spot corresponding to
on 1 g by drying in an oven at 60° at a pressure not exceeding 3,5,6-trichloro-2-hydroxybenzoic acid RS is not more intense
0.7 kPa. than that in the chromatogram obtained with reference
Assay. Weigh accurately about 0.25 g, dissolve in 75 ml of solution (a).
anhydrous pyridine and pass a stream of nitrogen through B. Determine by thin-layer chromatography (2.4.17), coating
the solution for 5 minutes. Titrate with 0.1 M the plate with silica gel G.
tetrabutylammonium hydroxide, maintaining a stream of Mobile phase. A mixture of 100 volumes of ethyl acetate, 10
nitrogen through the solution throughout the titration, volumes of methanol and 1 volume of strong ammonia
determining the end-point potentiometrically (2.4.25). Carry solution.
out a blank titration.
Test solution. Dilute a quantity with acetone to contain 1.0
1 ml of 0.1 M tetrabutylammonium hydroxide is equivalent to per cent w/v of Oxyclozanide, centrifuge and use the
0.02007 g of C13H6Cl5NO3. supernatant liquid.

1550
IP 2007 OXYCLOZANIDE PREMIX

Reference solution. A 0.040 per cent w/v of 2-amino- Reference solution (a). A 0.050 per cent w/v solution of
4,6-dichlorophenol RS in acetone. 3,5,6-trichloro- 2-hydroxybenzoic acid RS in acetone.
Apply to the plate 40 µl of test solution and 4 µl of reference Reference solution (b). A 1.0 per cent w/v solution of
solution. After development, dry the plate in air and spray oxyclozanide RS in acetone.
with lithium and sodium molybdotungstophosphate solution. Apply to the plate 40 µl and 10 µl of test solution, 4 µl of
In the chromatogram obtained with test solution any spot reference solution (a) and 10 µl of reference solution (b). After
corresponding to 2-amino-4,6-dichlorophenol is not more development, dry the plate in air and spray with a 3 per cent w/
intense than that in the chromatogram obtained with reference v solution of ferric chloride in methanol. In the chromatogram
solution. obtained with 40 µl of test solution any spot corresponding to
Other tests. Complies with the tests stated under Veterinary 3,5,6-trichloro-2-hydroxybenzoic acid RS is not more intense
Oral Liquids. than that in the chromatogram obtained with reference
solution (a).
Assay. Protect the solutions from light throughout the
procedure. Weigh accurately a quantity containing about 60 B. Determine by thin-layer chromatography (2.4.17), coating
mg of Oxyclozanide, add 60 ml of acidified methanol and boil the plate with silica gel G.
gently on a water-bath. Shake continuously for 20 minutes, Mobile phase. A mixture of 100 volumes of ethyl acetate, 10
cool to 2° and dilute to 100.0 ml with acidified methanol. volumes of methanol and 1 volume of strong ammonia
Filter, dilute 5.0 ml of the filtrate to 100.0 ml with acidified solution.
methanol and measure the absorbance of the resulting solution
Test solution. Extract the finely powdered preparation under
at the maximum at about 300 nm (2.4.7). Calculate the content
examination with sufficient acetone to produce a mixture
of C13H6Cl5NO3 taking 254 as the specific absorbance at 300
containing 1.0 per cent w/v of Oxyclozanide, centrifuge and
nm.
use the supernatant liquid.
Determine the weight per ml of the suspension (2.4.29), and
Reference solution. A 0.040 per cent w/v of
calculate the content of oxyclozanide, weight in volume.
2-amino-4,6-dichlorophenol RS in acetone.
Apply to the plate 40 µl of test solution and 4 µl of reference
solution. After development, dry the plate in air and spray
Oxyclozanide Premix with lithium and sodium molybdotungstophosphate solution.
Oxyclozanide Granules In the chromatogram obtained with test solution any spot
corresponding to 2-amino-4,6-dichlorophenol is not more
Oxyclozanide Premix contains Oxyclozanide.
intense than that in the chromatogram obtained with reference
Oxyclozanide Premix contains not less than 92.5 per cent and solution.
not more than 107.5 per cent of the stated amount of
Other tests. Complies with the tests stated under Premixes.
oxyclozanide, C13H6Cl5NO3.
Assay. Protect the solutions from light throughout the
Identification procedure. Weigh accurately a quantity of the finely powdered
preparation under examination containing 60 mg of
In test A for Related substances, the principal spot in the Oxyclozanide, add 60 ml of acidified methanol and boil gently
chromatogram obtained with 10 ml of test solution on a water-bath. Shake continuously for 20 minutes, cool to 2°
corresponds to that in the chromatogram obtained with and dilute to 100.0 ml with acidified methanol. Filter, dilute 5.0
reference solution (b). ml of the filtrate to 100.0 ml with acidified methanol and
measure the absorbance of the resulting solution at the
Tests
maximum at about 300 nm (2.4.7). Calculate the content of
Related substances. A. Determine by thin-layer C13H6Cl5NO3 taking 254 as the specific absorbance at 300 nm.
chromatography (2.4.17), coating the plate with silica gel G. Labelling. The label states (1) the proportion of oxyclozanide
Mobile phase. A mixture of 60 volumes of light petroleum ( in the premix and (2) the method of use of the preparation.
60° to 80°), 20 volumes of acetone and 5 volumes of glacial
acetic acid.
Test solution. Extract the finely powdered preparation under
examination with sufficient acetone to produce a mixture
containing 1.0 per cent w/v of Oxyclozanide, centrifuge and
use the supernatant liquid.

1551
OXYTETRACYCLINE VETERINARY ORAL POWDER IP 2007

Oxytetracycline Veterinary Oral it and filter again. The filtrate gives the reactions of chlorides
(2.3.1).
Powder
Tests
Oxytetracycline Hydrochloride Veterinary Oral Powder;
Oxytetracycline Hydrochloride Soluble Powder; Assay. To a quantity of the powder containing 0.25 g of
Oxytetracycline Soluble Powder Oxytetracylcine Hydrochloride, add 250.0 ml of water, shake,
filter and carry out the microbiological assay (2.2.10), Method
Oxytetracycline Veterinary Oral Powder is a mixture of
A or B.
Oxytetracycline Hydrochloride and Lactose or other suitable
diluent. Storage. Store at a temperature not exceeding 15°.
Oxytetracycline Veterinary Oral Powder contains not less than
90.0 per cent and not more than 110.0 per cent of the stated Pentobarbitone Injection
amount of oxytetracycline hydrochloride, C22H24N2O9,HCl.
Pentobarbitone Sodium Injection; Pentobarbital Sodium
Identification Injection
A. Determine by thin-layer chromatography (2.4.17), coating Pentobarbitone Injection is a sterile solution of Pentobarbitone
the plate with a substance prepared by mixing 25 g of silica Sodium in a suitable vehicle.
gel G with 50 ml of a mixture of 2.5 ml of glycerin and 47.5 ml of Solutions containing 20 per cent w/v of Pentobarbitone
0.1 M disodium edetate previously adjusted to pH 7.0 with Sodium in 100-ml and 500-ml containers are also available for
dilute ammonia solution. After spreading the plate, allow it use other than for injection. Such solutions may be coloured
to stand at room temperature till it is dry (70 to 90 minutes). and need not be sterile but must comply with all other
Mobile phase. The lower layer formed after shaking 200 ml of requirements of this monograph.
a mixture of 2 volumes of ethyl acetate, 2 volumes of Pentobarbitone Injection contains not less than 95.0 per cent
dichloromethane and 1 volume of acetone with 25 ml of 0.1 M and not more than 105.0 per cent of the stated amount of
disodium edetate previously adjusted to pH 7.0 with dilute pentobarbitone sodium, C11H17N2NaO3.
ammonia solution.
Description. A clear, colourless or almost colourless solution.
Test solution. Extract a quantity of the oral powder containing
10 mg of Oxytetracycline Hydrochloride with 20 ml of Identification
methanol, centrifuge and use the supernatant liquid.
A. Determine by infrared absorption spectrophotometry (2.4.6).
Reference solution (a). A 0.05 per cent w/v solution of Compare the spectrum with that obtained with pentobarbitone
oxytetracycline hydrochloride RS in methanol. RS or with the reference spectrum of pentobarbitone.
Reference solution (b). A solution containing 0.05 per cent B. The residue obtained in the Assay melts at about 128°
w/v each of demethylchlortetracycline hydrochloride RS, (2.4.21).
oxytetracycline hydrochloride RS and tetracycline
hydrochloride RS in methanol. C. When introduced on a platinum wire into a Bunsen burner
flame, a golden yellow colour is imparted to the flame.
Apply to the plate 1 µl of each solution. After development,
dry the plate in air, expose to the vapours of ammonia and Tests
examine in ultraviolet light at 365 nm. The principal spot in the
pH (2.4.24). 10.0 to 11.5.
chromatogram obtained with the test solution corresponds to
that in the chromatogram obtained with reference solution (a). Isomer. To a volume of the injection containing 0.3 g of
The test is not valid unless the chromatogram obtained with Pentobarbitone Sodium diluted, if necessary, to 5 ml with water
reference solution (b) shows three clearly separated spots. add 0.3 g of 4-nitrobenzyl bromide dissolved in 10 ml of
ethanol (95 per cent). Heat under a reflux condenser for 30
B. To a quantity of the powder containing 0.4 mg of
minutes, cool to 25°, scratch the sides of the vessel with a
Oxytetracycline Hydrochloride add 5 ml of a 1 per cent w/v
glass rod if necessary to induce crystallisation, filter and wash
solution of sodium carbonate, shake and add 2 ml of diazotised
the residue with five quantities, each of 5 ml, of water. Transfer
sulphanilic acid solution; a light brown colour is produced.
the residue as completely as possible to a small flask, add 25
C. Shake a quantity of the powder containing 100 mg of ml of ethanol (95 per cent) and heat under a reflux condenser
Oxytetracyline Hydrochloride with 10 ml of 2 M nitric acid for 10 minutes. Filter the hot solution, cool to 25° and scratch
and filter. To the filtrate add activated charcoal to decolorise the sides of the vessel with a glass rod to induce

1552
IP 2007 PROGESTERONE INJECTION

crystallisation. Filter and wash the residue with two quantities, Reference solution. A 0.1 per cent w/v of progesterone RS in
each of 5 ml, of water and dry at 105° for 30 minutes. The dried a mixturance at 241 nm.
residue melts completely between 136° and 148° (2.4.21).
Storage. Store protected from light.
Other tests. Complies with the tests stated under Parenteral
Preparations (Injections).
Assay. To an accurately measured volume containing about Progesterone Injection
0.5 g of Pentobarbitone Sodium diluted to 15 ml with water
add 5 ml of 2 M hydrochloric acid, extract with 50 ml of ether Progesterone Injection is a sterile solution of Progesterone in
and then with successive quantities, each of 25 ml, of ether Ethyl Oleate or other suitable ester, in a suitable fixed oil or in
until complete extraction is effected. Wash the combined any mixture of these. It may contain suitable alcohols.
extracts with two quantities, each of 5 ml, of water and wash Progesterone Injection contains not less than 92.5 per cent
the combined aqueous extracts with 10 ml of ether. Add the and not more than 107.5 per cent of the stated amount of
ether washings to the main ethereal extract, filter and wash the progesterone, C21H30O2.
filter with ether. Evaporate the solvent and dry the residue to
constant weight at 105°. Identification
1 g of the residue is equivalent to 1.097 g of C11H17N2NaO3. Dissolve a volume containing 50 mg of Progesterone in 8 ml of
light petroleum ( 40° to 60°) and extract with three quantities,
each of 8 ml, of a mixture of 7 volumes of glacial acetic acid
Progesterone and 3 volumes of water until the solution becomes turbid,
allow to stand in ice for 2 hours and filter. The precipitate, after
O CH3 washing with water and drying at 105°, complies with the
H3 C following tests.
A. Determine by infrared absorption spectrophotometry (2.4.6).
H3 C H Compare the spectrum with that obtained with progesterone
RS or with the reference spectrum of progesterone. If the
H H
spectra are not concordant, prepare spectra using 5 per cent
O w/v solutions in chloroform IR.
C21H30O2 Mol. Wt. 314.5 B. Determine by thin-layer chromatography (2.4.17), coating
Progesterone is pregn-4-en-3,20-dione. the plate with silica gel G.

Progesterone contains not less than 97.0 per cent and not Solvent mixture. A mixture of 90 volumes of acetone and 10
more than 103.0 per cent of C21H30O2, calculated on the dried volumes of 1,2-propanediol.
basis. Mobile phase. A mixture of equal volumes of cyclohexane
Description. Colourless crystals or a white or almost white and light petroleum (40° to 60 °).
crystalline powder. Test solution. Dissolve 25 mg of the substance under
examination in 10 ml of the solvent mixture.
Identification
Reference solution (a). A 0.25 per cent w/v solution of
A. Determine by infrared absorption spectrophotometry (2.4.6). progesterone RS in the solvent mixture.
Compare the spectrum with that obtained with progesterone
Reference solution (b). Mix equal volumes of the test solution
RS or with the reference spectrum of progesterone. If the
and reference solution (a).
spectra are not concordant, prepare spectra using 5 per cent
w/v solutions in chloroform IR. Place the dry plate in a tank containing a shallow layer of the
solvent mixture, allow the solvent mixture to ascend to the
B. Determine by thin-layer chromatography (2.4.17), coating
top, remove the plate from the tank and allow the solvent to
the plate with silica gel GF254.
evaporate. Use within 2 hours, with the flow of the mobile
Mobile phase. A mixture of 66 volumes of dichloromethane phase in the direction in which the aforementioned treatment
and 33 volumes of ethyl acetate. was done.
Test solution. Dissolve 0.1 g of the substance under Apply to the plate 2 µl of each solution. Allow the mobile
examination in 100 ml of a mixture of 9 volumes of phase to rise 12 cm. Dry the plate in a current of warm air, allow
dichloromethane and 1 volume of methanol. the solvent to evaporate, heat at 120º for 15 minutes and spray

1553
PROMAZINE HYDROCHLORIDE IP 2007

the hot plate with ethanolic sulphuric acid (20 per cent v/v). Identification
Heat at 120º for a further 10 minutes, allow to cool and examine
in daylight and in ultraviolet light at 365 nm. The principal Test A may be omitted if tests B, C, D and E are carried out.
spot in the chromatogram obtained with the test solution Tests B, C and D may be omitted if tests A and E are carried
corresponds to that in the chromatogram obtained with out.
reference solution (a). The principal spot in the chromatogram A. Determine by infrared absorption spectrophotometry (2.4.6).
obtained with reference solution (b) appears as a single, Compare the spectrum with that obtained with promazine
compact spot. hydrochloride RS or with the reference spectrum of promazine
hydrochloride.
Tests
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Other tests. Complies with the tests stated under Parenteral 0.001 per cent w/v solution in 0.01 M hydrochloric acid shows
Preparations (Injections). an absorption maximum at about 252 nm and a less well-defined
maximum at about 302 nm; absorbance at about 252 nm, about
Assay. To an accurately measured volume containing about
0.93.
50 mg of Progesterone add sufficient dichloromethane to
produce 100.0 ml. Dilute 3.0 ml to 50.0 ml with dichloromethane. C. Dissolve 5 mg in 2 ml of sulphuric acid and allow to stand
To 5.0 ml of the solution add 10 ml of isoniazid solution and for 5 minutes; an orange colour is produced.
sufficient methanol to produce 20.0 ml. Allow to stand for 45 D. Melting range (2.4.21). 177° to 181°.
minutes and measure the absorbance of the resulting solution
E. Gives reaction A of chlorides (2.3.1).
at the maximum at about 380 nm (2.4.7), using as the blank 5 ml
of dichloromethane treated in the same manner. Calculate the Tests
content of C 21H30 O2 from the absorbance obtained by
pH (2.4.24). 4.2 to 5.4, determined in a 5 per cent w/v solution.
repeating the procedure using a 0.003 per cent w/v solution of
progesterone RS in dichloromethane and beginning at the Sulphated ash (2.3.18). Not more than 0.1 per cent.
words “To 5.0 ml of the solution.....”. Loss on drying (2.4.19). Not more than 0.5 per cent, determined
Storage. Store protected from light. If solid matter separates on 1.0 g by drying in an oven at 105°.
on standing, it should be redissolved by heating before use. Assay. Weigh accurately about 0.6 g, dissolve in 100 ml of
Labelling. The label states (1) the composition of the solvent; acetone. Titrate with 0.1 M perchloric acid, using 3 ml of a
(2) that the preparation is intended for veterinary use by saturated solution of methyl orange in acetone as indicator.
subcutaneous or intramuscular injection only. Carry out a blank titration.
1 ml of 0.1 M perchloric acid is equivalent to 0.03209 g of
C17H20N2S,HCl.
Storage. Store protected from light.
Promazine Hydrochloride
CH3
Promazine Injection
N Promazine Hydrochloride Injection
CH3
Promazine Injection is a sterile solution of Promazine
N
, HCl Hydrochloride in Water for Injections free from dissolved air
and containing suitable buffering and stabilising agents. The
S solution is distributed in containers, the air in which is replaced
by nitrogen or other suitable gas.
C17H20N2S,HCl Mol. Wt. 320.9
Promazine Injection contains not less than 95.0 per cent and
Promazine Hydrochloride is 10-(3-dimethylaminopropyl) not more than 105.0 per cent of the stated amount of promazine
phenothiazine hydrochloride. hydrochloride, C17H20N2S,HCl.
Promazine Hydrochloride contains not less than 99.0 per cent Description. A colourless or almost colourless liquid.
and not more than 101.0 per cent of C17H20N2S,HCl, calculated
on the dried basis. Identification
Description. A white or almost white, crystalline powder; A. To a volume containing 0.1 g of Promazine Hydrochloride
slightly hygroscopic. add 20 ml of water and 2 ml of 10 M sodium hydroxide. Shake

1554
IP 2007 RAFOXANIDE

and extract the mixture with 25 ml of ether. Wash the ether of 0.1 M hydrochloric acid, dilute to 100.0 ml with water and
extract with two quantities, each of 5 ml, of water, dry with measure the absorbance of the resulting solution at the
anhydrous sodium sulphate and evaporate the ether. A 10 per maximum at about 251 nm (2.4.7). Calculate the content of
cent w/v solution of the oily residue in chloroform complies C17H20N2S,HCl taking 935 as the specific absorbance at
with the following test. 251 nm.
Determine by infrared absorption spectrophotometry (2.4.6). Storage. Store protected from light.
Compare the spectrum with that obtained with promazine
hydrochloride RS, treated in the same manner.
B. To a volume containing 5 mg of Promazine Hydrochloride Rafoxanide
add carefully 2 ml of sulphuric acid and allow to stand for 5
minutes; an orange colour is produced. O
O
C. To a volume containing 0.2 g of Promazine Hydrochloride
add 1 ml of 1 M sodium hydroxide and extract with four I
N Cl Cl
quantities, each of 10 ml, of ether. Wash the combined extracts H
with 10 ml of water, remove the ether and dissolve the residue OH
in 4 ml of methanol. Heat on a water-bath almost to boiling, I
immediately add 2 ml of a boiling 3.5 per cent w/v solution of
picric acid in methanol and boil for 2 minutes. Cool in ice, C19H11Cl2I2NO3 Mol. Wt. 626.0
filter, wash the crystals thrice with methanol, dissolve in 10 ml
Rafoxanide is N-[3-chloro-4-(4-chlorophenoxy)phenyl]-2-
of hot methanol and repeat the crystallisation and washing.
hydroxy-3,5-diiodobenzamide.
The rust-red crystals so obtained, after drying at 105° for 1
hour, melt at about 144° (2.4.21). Rafoxanide contains not less than 98.0 per cent and not more
than 101.0 per cent of C19H11Cl2I2NO3, calculated on the dried
Tests basis.
pH (2.4.24). 4.4 to 5.2. Description. A greyish-white to brown powder.
Related substances. Carry out the test for identification of Identification
related substances in phenothiazines (2.3.5), using mobile
phase A and applying separately to the plate 10 µl of each of A. Determine by infrared absorption spectrophotometry (2.4.6).
the following freshly-prepared solutions. Compare the spectrum with that obtained with rafoxanide RS
or with the reference spectrum of rafoxanide.
Test solution. Dilute a volume of the injection with sufficient
methanol to produce a solution containing the equivalent of B. When examined in the range 230 nm to 360 nm (2.4.7), a
1.0 per cent w/v of Promazine Hydrochloride. 0.004 per cent w/v solution in 0.1 M methanolic hydrochloric
acid shows absorption maxima at about 280 nm and at 335 nm;
Reference wsoslution (a). Dilute 1 volume of the test solution
absorbance at about 280 nm, about 0.97 and at about 335 nm,
to 40 volumes with methanol.
about 0.59.
Reference wsoslution (b). Dilute 1 volume of the test solution
C. Burn 20 mg by the oxygen-flask method (2.3.34), using 5 ml
to 200 volumes with methanol.
of 2 M sodium hydroxide as the absorbing liquid, and dilute
Any secondary spot in the chromatogram obtained with the to 25 ml with water. To 5 ml add 1 ml of silver nitrate solution;
test solution is more intense than the spot in the chromatogram a yellow precipitate is produced; add 5 ml of 5 M ammonia,
obtained with reference solution (a) and not more than one shake, filter, and acidify the filtrate with nitric acid; a white
such spot is more intense than the spot in the chromatogram precipitate is produced.
obtained with reference solution (b).
D. Shake 10 mg with 10 ml of ethanol (80 per cent) and add
Other tests. Complies with the tests stated under Parenteral 0.1 ml of ferric chloride test solution; a violet colour is
Preparations (Injections). produced.
Assay. Protect the solutions from light throughout the E. Melts at about 175° (2.4.21).
procedure.
Tests
To an accurately measured volume containing about 50 mg of
Promazine Hydrochloride, add 5 ml of 2 M hydrochloric acid Related substances. Determine by thin-layer chromatography
and sufficient water to produce 1000.0 ml. To 10.0 ml add 10 ml (2.4.17), coating the plate with silica gel GF254.

1555
RAFOXANIDE VETERINARY ORAL SUSPENSION IP 2007

NOTE – Carry out the test in subdued light and use freshly B. In addition to the absorbance at about 335 nm, measure the
prepared solution5. absorbance at about 280 nm (2.4.7), of the final solution
obtained in the Assay. The ratio of the absorbance at about
Mobile phase. A mixture of 170 volumes of dichloromethane,
280 nm to that at about 335 nm is 1.59 to 1.69.
30 volumes of methanol and 2 volumes of strong ammonia
solution. Tests
Test solution. Dissolve 2 g of the substance under examination
Other tests. Complies with requirements stated under
in 100 ml in dichloromethane.
Veterinary Oral Liquids.
Reference solution. A 0.010 per cent w/v of rafoxanide RS in
Assay. Weigh accurately a quantity of the well-mixed
dichloromethane.
suspension containing about 0.12 g of Rafoxanide in a
Apply to the plate 5 µl of each solution. After development, stoppered 50-ml test tube and add 15 ml of 0.1 M sodium
dry the plate in air and examine in ultraviolet light at 254 nm. hydroxide and 15 ml of ether. Shake for 5 minutes and
Any secondary spot in the chromatogram obtained with the centrifuge. Remove the ether layer and repeat the extraction
test solution is not more intense than the spot in the with three further quantities, each of 15 ml, of ether. Dilute the
chromatogram obtained with the reference solution. combined ether solutions to 250.0 ml with ether and mix. Dilute
5.0 ml of this solution to 100.0 ml with 0.1 M methanolic
Sulphated ash (2.3.18). Not more than 0.1 per cent.
hydrochloric acid, mix and measure the absorbance of the
Loss on drying (2.4.19). Not more than 0.5 per cent, determined resulting solution at about 335 nm (2.4.7). Calculate the content
on 1.0 g by drying in an oven at 90° at a pressure not exceeding of C19H11Cl2I2NO3 taking 149 as the specific absorbance at
0.7 kPa for 2 hours. 335 nm.
Assay. To 50 ml of dioxan add 1 ml of phenolphthalein solution, Determine the weight per ml of the suspension (2.4.29), and
replace the air in the flask with nitrogen and titrate with 0.1 M calculate the content of rafoxanide, weight in volume.
sodium hydroxide. Weigh accurately about 1.25 g, dissolve it
in the mixture and again titrate with 0.1 M sodium hydroxide.
The difference between the titrations represents the amount
of 0.1 M sodium hydroxide required.
Ronidazole
1 ml of 0.1 M sodium hydroxide is equivalent to 0.06260 g of CH3 O
C19H11Cl2I2NO3.
N
O2N
Storage. Store protected from light. O NH2
N

C6H8N4O4 Mol. Wt. 200.2


Ronidazole is 1-methyl-2-[(carbamoyloxy)methyl]-5-
Rafoxanide Veterinary Oral nitroimidazole.
Suspension
Ronidazole contains not less than 98.5 per cent and not more
Rafoxanide Suspension; Rafoxanide Veterinary Mixture; than 101.0 per cent of C6H8N4O4, calculated on the anhydrous
Rafoxanide Mixture basis.
Rafoxanide Veterinary Oral Suspension is an aqueous Description. A white to yellowish-brown powder; odourless
suspension of Rafoxanide containing suitable suspending and or almost odourless.
dispersing agents and antimicrobial preservatives.
Identification
Rafoxanide Veterinary Oral Suspension contains not less than
90.0 per cent and not more than 110.0 per cent of rafoxanide, A. Determine by infrared absorption spectrophotometry (2.4.6).
C19H11Cl2I2NO3. Compare the spectrum with that obtained with ronidazole RS
or with the reference spectrum of ronidazole.
Identification B. When examined in the range 230 nm to 360 nm (2.4.7), a
A. Evaporate a volume containing 0.2 g of Rafoxanide to 0.002 per cent w/v solution in 0.1 M methanolic hydrochloric
dryness on a water-bath and heat the residue over a Bunsen acid shows an absorption maximum only at about 270 nm;
burner flame; the vapours turn moistened starch-iodide paper absorbance at about 270 nm, about 0.64.
blue. C. Melts at about 167° (2.4.21).

1556
IP 2007 SERUM GONADOTROPHIN FOR VETERINARY USE

Tests B. When examined in the range 230 nm to 360 nm (2.4.7), the


final solution obtained in the Assay shows an absorption
Appearance of solution. A 0.5 per cent w/v solution in maximum only at about 281 nm.
methanol is not more intensely coloured than reference
solution YS6 (2.4.1). Tests
(1-Methyl-5-nitroimidazol-2-yl)methano .Determine by thin- (1-Methyl-5-nitroimidazol-2-yl)methanol .Determine by thin-
layer chromatography (2.4.17), coating the plate with silica layer chromatography (2.4.17), coating the plate with silica
gel GF254. gel GF254.
Mobile phase. A mixture of 80 volumes of toluene, 5 volumes Mobile phase. A mixture of 80 volumes of toluene, 5 volumes
of methanol and 5 volumes of glacial acetic acid. of methanol and 5 volumes of glacial acetic acid.
Test solution. Dissolve 1 g of the substance under examination Test solution. Shake a quantity of the powder containing 0.1
in 100 ml in acetone. g of Ronidazole with 10 ml of acetone for 15 minutes and filter.
Reference solution. A 0.0050 per cent w/v of Reference solution. A 0.0050 per cent w/v of
(1-methyl-5-nitroimidazol-2-yl)methanol RS in acetone. (1-methyl-5-nitroimidazol-2-yl)methanol RS in acetone.
Apply to the plate 20 µl of each solution. After development, Apply to the plate 20 µl of each solution. After development,
dry the plate in air and examine in ultraviolet light at 254 nm. dry the plate in air and examine in ultraviolet light at 254 nm.
Any secondary spot in the chromatogram obtained with the Any secondary spot in the chromatogram obtained with the
test solution corresponding to (1-methyl-5-nitroimidazol-2 test solution corresponding to
-yl)methanol RS is not more intense than the spot in the (1-methyl-5-nitroimidazol-2-yl)methanol RS is not more intense
chromatogram obtained with the reference solution. than the spot in the chromatogram obtained with the reference
solution.
Sulphated ash (2.3.18). Not more than 0.1 per cent.
Other tests. Complies with the tests stated under Veterinary
Water (2.3.43). Not more than 0.5 per cent , determined on 5 g.
Oral Powders.
Assay. Weigh accurately about 0.3 g, dissolve in 50 ml of
Assay. Weigh accurately a quantity of powder containing 2 g
anhydrous glacial acetic acid. Titrate with 0.1 M perchloric
of Ronidazole, dissolve in 450 ml of water and add sufficient
acid, determining the end-point potentiometrically (2.4.25).
water to produce 500.0 ml. Dilute 5.0 ml of this solution to
Carry out a blank titration.
100.0 ml with 0.1 M hydrochloric acid and measure the
1 ml of 0.1 M perchloric acid is equivalent to 0.02002 g of absorbance of the resulting solution at the maximum at about
C6H8N4O4. 281 nm (2.4.7). Calculate the content of C6H8N4O4 taking 279
as the specific absorbance at 281 nm.
Storage. Store proted from light.
Storage. Store proted from light.

Ronidazole Veterinary Oral Powder Serum Gonadotrophin for Veterinary


Ronidazole Veterinary Oral Powder is a mixture of Ronidazole
Use
with suitable diluents. Equine Serum Gonadotrophin for Veterinary Use
Ronidazole Veterinary Oral Powder contains not less than 92.5 Serum Gonadotrophin for Veterinary Use is a dry preparation
per cent and not more than 107.5 per cent of the stated amount of a glycoprotein fraction, obtained from the serum or plasma
of ronidazole, C6H8N4O4. of pregnant mares in their 60th to 75th day of pregnancy,
which stimulates the formation of follicles and induces
Identification leutinising activity.
A. Shake a quantity of the powder containing 0.1 g of Serum Gonadotrophin for Veterinary Use contains not less
Ronidazole with 10 ml of acetone for 15 minutes, filter and than 1000 Units per mg, calculated on the anhydrous basis.
evaporate the filtrate to dryness. The residue complies with Description. A white or pale grey, amorphous powder.
the following test.
Determine by infrared absorption spectrophotometry (2.4.6).
Identification
Compare the spectrum with that obtained with ronidazole RS Causes enlargement of the ovaries of immature female rats
or with the reference spectrum of ronidazole. when administered as directed in the Assay.

1557
SERUM GONADOTROPHIN INJECTION FOR VETERINARY USE IP 2007

Tests Calculate the result of the assay by standard statistical


methods using the combined weight of the two ovaries of
Water (2.3.43). Not more than 10.0 per cent, determined on 80 each animal as the response.
mg.
Limits of error - The estimated potency is not less than 80 per
Assay. Carry out the biological assay of serum gonadotrophin cent and not more than 125 per cent of the stated potency.
described below. The fiducial limits of error (P = 0.95) of the estimated potency
The potency of serum gonadotrophin for veterinary use is are not less than 64 per cent and not more than 156 per cent of
determined by comparing its effect in increasing the weight of the stated potency.
the ovaries of immature rats with that of the Standard Serum Gonadotrophin for Veterinary Use intended for use in
Preparation of serum gonadotrophin under the conditions of the manufacture of parenteral preparations without a further
the following method of assay. appropriate procedure for the removal of pyrogens complies
with the following additional requirement.
Standard Preparation
Pyrogens. Complies with the test for pyrogens (2.2.8), using
The Standard Preparation is the 2nd International Standard per kg of the rabbit’s weight 1 ml of a solution in sodium
for serum gonadotrophin, equine, for bioassay, established in chloride injection containing 500 Units per ml.
1966, consisting of the freeze-dried active principle from the
serum of pregnant mares, with lactose (supplied in ampoules Serum Gonadotrophin for Veterinary Use intended for use in
containing 1600 Units), or other suitable preparation the the manufacture of parenteral preparations without a further
potency of which has been determined in relation to the appropriate sterilisation procedure complies with the
International Standard. following additional requirement.
Sterility (2.2.11). Complies with the test for sterility.
Method
Storage. Store protected from moisture and light in a refrigerator
Test animals. Use immature female rats of the same strain, 21 (2 to 8). If the contents are sterile, the containers should be
to 28 days old, differing in age by not more than 3 days and of sterile, tamper-evident and sealed so as to exclude
approximately equal weights such that the difference between micro-organisms.
the heaviest and the lightest rat is not more than 10 g. Assign
the rats at random to six equal groups of not less than five Labelling. The label states (1) the number of Units per mg; (2)
animals. If sets of six litter-mates are available, allot one the total number of Units in the container; (3) the date after
litter-mate from each set at random to each group and mark which the material is not intended to be used; (4) the storage
according to the litter. conditions; (5) whether or not it is intended for use in the
manufacture of parenteral preparations.
Procedure. Choose three doses of the Standard Preparation
and three doses of the preparation under examination such
that the smallest dose is sufficient to produce a positive
response in some of the rats and the largest dose does not
produce a maximal response in all of the rats. Use doses in Serum Gonadotrophin Injection for
geometric progression. As an initial approximation total doses Veterinary Use
of 8, 12 and 18 Units may be tried although the dose will
Serum Gonadotrophin Injection for Veterinary Use is a sterile
depend on the sensitivity of the animals used, which may
material consisting of Serum Gonadotrophin for Veterinary
vary widely. Dissolve separately the total quantities of the
Use with or without buffering agents and other excipients. It
preparation under examination and of the Standard Preparation
is filled in a sealed container.
corresponding to the doses to be used in sufficient of a sterile
saline solution containing 1 mg of bovine albumin per ml The injection is constituted by dissolving the contents of the
such that each single dose may be administered by the sealed container in the requisite amount of sterile Water for
injection of 6 equally-divided portions, in the same volume of Injections, immediately before use.
about 0.2 ml. Store the solutions at a temperature 2° to 8°.
The constituted solution complies with the requirements for
Inject subcutaneously into each rat the dose allocated to its
Clarity of solution and Particulate matter stated under
group. Repeat the injections 18, 21, 24, 42 and 48 hours after
Parenteral Preparations (Injections).
the first injection. Kill the rats between 40 hours and 72 hours
after the last injection and remove the ovaries. Remove any Storage. The constituted solution should be used immediately
extraneous fluid and tissue and immediately weigh the two after preparation but, in any case, within the period
ovaries from each rat. recommended by the manufacturer.

1558
IP 2007 SPECTINOMYCIN HYDROCHLORIDE

Serum Gonadotrophin Injection for veterinary Use contains vary widely. Dissolve separately the total quantities of the
not less than 80.0 per cent and not more than 125.0 per cent of preparation under examination and of the Standard Preparation
the stated potency. corresponding to the doses to be used in sufficient of a sterile
saline solution containing 1 mg of bovine albumin per ml
The contents of the sealed container comply with the
such that each single dose may be administered by the
requirements stated under Parenteral Preparations
injection of 6 equally-divided portions, in the same volume of
(Powders for Injection) and with the following requirements.
about 0.2 ml. Store the solutions at a temperature 2° to 8°.
Identification Inject subcutaneously into each rat the dose allocated to its
group. Repeat the injections 18, 21, 24, 42 and 48 hours after
Causes enlargement of the ovaries of immature female rats the first injection. Kill the rats between 40 hours and 72 hours
when administered as directed in the Assay. after the last injection and remove the ovaries. Remove any
extraneous fluid and tissue and immediately weigh the two
Tests ovaries from each rat.
Appearance of solution. A solution containing 5000 Units per Calculate the result of the assay by standard statistical
ml (solution A) is clear (2.4.1), and colourless (2.4.1). methods using the combined weight of the two ovaries of
pH (2.4.24). 6.0 to 8.0, determined on solution A. each animal as the response.
Water (2.3.43). Not more than 10.0 per cent, determined on Limits of error - The estimated potency is not less than 80 per
80 mg. cent and not more than 125 per cent of the stated potency.
The fiducial limits of error (P = 0.95) of the estimated potency
Assay. Carry out the biological assay of serum gonadotrophin are not less than 64 per cent and not more than 156 per cent of
described below. the stated potency.
The potency of serum gonadotrophin for veterinary use is Pyrogens. Complies with the test for pyrogens (2.2.8), using
determined by comparing its effect in increasing the weight of per kg of the rabbit’s weight 1 ml of a solution in sodium
the ovaries of immature rats with that of the Standard chloride injection containing 500 Units per ml.
Preparation of serum gonadotrophin under the conditions of
the following method of assay. Storage. Store protected from light in a refrigerator (2° to 8°) .
Labelling. The label states the number of Units contained in
Standard Preparation
the sealed container.
The Standard Preparation is the 2nd International Standard
for serum gonadotrophin, equine, for bioassay, established in
1966, consisting of the freeze-dried active principle from the
serum of pregnant mares, with lactose (supplied in ampoules Spectinomycin Hydrochloride
containing 1600 Units), or other suitable preparation the
potency of which has been determined in relation to the
International Standard. H HO H H
Test animals. Use immature female rats of the same strain, 21 N O O CH3
H3C
to 28 days old, differing in age by not more than 3 days and of , 2HCl, 5H2O
approximately equal weights such that the difference between HO H OHO
the heaviest and the lightest rat is not more than 10 g. Assign NH O
the rats at random to six equal groups of not less than five H3 C
animals. If sets of six litter-mates are available, allot one
litter-mate from each set at random to each group and mark C14H24N2O7,2HCl,5H2O Mol. Wt. 495.4
according to the litter.
Spectinomycin Hydrochloride is [2R-
Procedure. Choose three doses of the Standard Preparation (2α,4aβ,5aβ,6β,7β,8β,9α,9aα,10aβ)]-decahydro-4a,7,9-
and three doses of the preparation under examination such trihydroxy-2-methyl-6,8-bis(methylamino)-4H-pyrano[2,3-
that the smallest dose is sufficient to produce a positive b][1,4]benzodioxin-4-one dihydrochloride pentahydrate.
response in some of the rats and the largest dose does not
Spectinomycin Hydrochloride contains not less than 95.0 per
produce a maximal response in all of the rats. Use doses in
cent and not more than 100.5 per cent of C14H24N2O7,2HCl,
geometric progression. As an initial approximation total doses
calculated on the anhydrous basis.
of 8, 12 and 18 Units may be tried although the dose will
depend on the sensitivity of the animals used, which may Description. A white or almost white, crystalline powder.

1559
SPECTINOMYCIN INJECTION IP 2007

Identification ml of a solution containing 0.15 per cent w/v of phenazone


(internal standard) in dimethylformamide and 2.0 ml of
A. Determine by infrared absorption spectrophotometry (2.4.6). hexamethyl- disilazane, shake intermittently for 1 hour and
Compare the spectrum with that obtained with spectinomycin dilute to 20.0 ml with dimethylformamide.
hydrochloride RS or with the reference spectrum of
spectinomycin hydrochloride. Chromatographic system
– a glass column 1.5 m x 4 mm, packed with acid-washed,
B. Gives reaction A of chlorides (2.3.1). silanised diatomaceous support (100 to 120 mesh) coated
Tests with 3 per cent w/w of phenylmethylsilicone fluid (50
per cent phenyl),
Appearance of solution. A 10 per cent w/v solution is clear – temperature:
(2.4.1), and colourless (2.4.1). column 200°,
pH (2.4.24). 3.8 to 5.6, determined in a 10 per cent w/v solution. inlet port 200° and detector 230°,
– flow rate. 45 ml per minute of the carrier gas.
Specific optical rotation (2.4.22). +15.0° to +21.0°, determined
in a 10 per cent w/v solution within 20 minutes of preparation, Inject the chosen volumes of test solutions (a) and (b). The
on the anhydrous basis. test is not valid unless the resolution factor between the peak
Related substances. Determine by thin-layer chromatography due to the internal standard and the principal peak in the
(2.4.17), coating the plate with silica gel G. chromatogram obtained with test solution (a) is not less than
8.0.
Mobile phase. A mixture of 50 volumes of 1-propanol, 40
volumes of water, 5 volumes of glacial acetic acid and 5 Inject alternately test solution (b) and the reference solution.
volumes of pyridine. Calculate the content of C14H24N2O7,2HCl.
Test solution. Dissolve 2 g of the substance under examination Spectinomycin Hydrochloride intended for use in the
in 100 ml water. manufacture of parenteral preparations without a further
Reference solution. A 0.020 per cent w/v solution of the appropriate procedure for the removal of bacterial
substance under examination in water. endotoxins complies with the following additional
requirement.
Apply to the plate 10 µl of each solution. After development,
dry the plate in air and spray with a 5 per cent w/v solution of Bacterial endotoxins (2.2.3). Not more than 0.09 Endotoxin
potassium permanganate. Allow the plate to stand for 2 to 3 Unit per mg determined in a 0.42 per cent w/v solution of
minutes. Any secondary spot in the chromatogram obtained sodium bicarbonate.
with the test solution is not more intense than the spot in the Spectinomycin Hydrochloride intended for use in the
chromatogram obtained with the reference solution (1 per manufacture of parenteral preparations without a further
cent). appropriate sterilisation procedure complies with the
Sulphated ash (2.3.18). Not more than 1.0 per cent w/v. following additional requirement.
Water (2.3.43). 16.0 to 20.0 per cent , determined on 0.2 g. Sterility (2.2.11). Complies with the test for sterility.
Assay. Determine by gas chromatography (2.4.13). Storage. Store protected from moisture, at a temperature not
NOTE – Use the solutions within 1 hour after preparation. exceeding 30°. If the substance is sterile, the container should
be sterile, tamper-evident and sealed so as to exclude
Test solution (a). Take 60 mg of the substance under
micro-organisms.
examination in a glass-stoppered conical flask, add 10.0 ml of
dimethylformamide and 2.0 ml of hexamethyl- disilazane, Labelling. The label states (1) the date after which the material
shake intermittently for 1 hour and dilute to 20.0 ml with is not intended to be used; (2) the storage conditions; (3)
dimethylformamide. whether or not it is intended to be used for manufacture of
parenteral preparations.
Test solution (b). Take 60 mg of the substance under
examination in a glass-stoppered conical flask, add 10.0 ml of
a solution containing 0.15 per cent w/v of phenazone (internal
standard) in dimethylformamide and 2.0 ml of hexamethyl-
Spectinomycin Injection
disilazane, shake intermittently for 1 hour and dilute to 20.0 Spectinomycin Hydrochloride Injection
ml with dimethylformamide.
Spectinomycin Injection is a sterile material consisting of
Reference solution. Take 60 mg of the spectinomycin Spectinomycin Hydrochloride with or without auxiliary
hydrochloride RS in a glass-stoppered conical flask, add 10.0 substances. It is filled in a sealed container.

1560
IP 2007 SPECTINOMYCIN INJECTION

The injection is constituted by suspending the contents of Assay. Determine by gas chromatography (2.4.13).
the sealed container in the requisite amount of sterile Water
NOTE – Use the solutions within 1 hour after preparation.
for Injections, immediately before use.
Test solution (a). Weigh and mix the contents of the 10
Storage. The constituted suspension should be used
containers. To an accurately weighed quantity containing
immediately after preparation but, in any case, within the period
about 60 mg of Spectinomycin Hydrochloride in a
recommended by the manufacturer.
glass-stoppered conical flask, add 10.0 ml of
Spectinomycin Injection contains not less than 90.0 per cent dimethylformamide and 2.0 ml of hexamethyl- disilazane,
and not more than 110.0 per cent the stated amount of shake intermittently for 1 hour and dilute to 20.0 ml with
spectinomycin, C14H24N2O7. dimethylformamide.
The contents of the sealed container comply with the Test solution (b). To an accurately weighed quantity containing
requirements stated under Parenteral Preparations about 60 mg of Spectinomycin Hydrochloride in a
(Powders for Injection) and with the following requirements. glass-stoppered conical flask, add 10.0 ml of a solution
containing 0.15 per cent w/v of phenazone (internal standard)
Identification in dimethylformamide and 2.0 ml of hexamethyl- disilazane,
shake intermittently for 1 hour and dilute to 20.0 ml with
A. Determine by infrared absorption spectrophotometry (2.4.6).
dimethylformamide.
Compare the spectrum with that obtained with spectinomycin
hydrochloride RS or with the reference spectrum of Reference solution. T0 about 60 mg, accurately weighed, of
spectinomycin hydrochloride. spectinomycin hydrochloride RS in a glass-stoppered conical
flask, add 10.0 ml of a solution containing 0.15 per cent w/v of
B. Gives reaction A of chlorides (2.3.1).
phenazone (internal standard) in dimethylformamide and 2.0
Tests ml of hexamethyl- disilazane, shake intermittently for 1 hour
and dilute to 20.0 ml with dimethylformamide.
pH (2.4.24). 4.0 to 7.0, determined in a suspension of the
Chromatographic system
contents of a sealed container in the volume of the liquid
– a glass column 1.5 m x 4 mm, packed with acid-washed,
stated on the label.
silanised diatomaceous support (100 to 120 mesh) coated
Related substances. Determine by thin-layer chromatography with 3 per cent w/w of phenylmethylsilicone fluid (50
(2.4.17), coating the plate with silica gel G. per cent phenyl),
Mobile phase. A mixture of 50 volumes of 1-propanol, 40 – temperature:
volumes of water, 5 volumes of glacial acetic acid and 5 column 200°,
volumes of pyridine. inlet port 200° and detector 230°,
– flow rate. 45 ml per minute of the carrier gas.
Test solution. Prepare a solution containing the equivalent of
1.4 per cent w/v of spectinomycin in water.
Inject the chosen volumes of test solutions (a) and (b). The
Reference solution. Prepare a solution containing the
test is not valid unless the resolution factor between the peak
equivalent of 0.014 per cent w/v of spectinomycin in water.
due to the internal standard and the principal peak in the
Apply to the plate 10 µl of each solution. After development, chromatogram obtained with test solution (a) is not less than
dry the plate in air and spray with a 5 per cent w/v solution of 8.0.
potassium permanganate. Allow the plate to stand for 2 to 3
Inject alternately test solution (b) and the reference solution.
minutes. Any secondary spot in the chromatogram obtained
with the test solution is not more intense than the spot in the Calculate the content of C14H24N2O7,2HCl.
chromatogram obtained with reference solution. Storage. Use the injection immediately after preparation but,
Water (2.3.43). Not more than 20.0 per cent, determined on 0.2 in any case, within the period recommended by the
g. manufacturer provided it is prepared and stored in accordance
Bacterial endotoxins (2.2.3). Not more than 0.09 Endotoxin with the manufacturer’s instructions.
Unit per ml, determined on a solution prepared by dissolving Labelling. The label states the strength in terms of the
the contents in a solution containing 0.05 M sodium equivalent amount of spectinomycin.
bicarbonate in water BET to give a solution containing the
equivalent of 1 mg of spectinomycin per ml (solution A), and
using the maximum valid dilution of solution A calculated from
the declared sensitivity of the lysate used in the test.

1561
SPIRAMYCIN IP 2007

Spiramycin Mobile phase. A mixture of 45 volumes of ethyl acetate, 40


volumes of a 15 per cent w/v of ammonium acetate previously
CH3 adjusted to pH 9.6 with 10 M sodium hydroxide and 20
N CH
O CH3 3 volumes of 2-propanol. Use the upper layer.
CH3 Spiramycin I R = H Test solution. Dissolve 0.4 g of the substance under
Spiramycin II R = COCH3
CH3 Spiramycin III R = COCH2CH3 examination in 100 ml of methanol.
CHO N CH3
H3CO
O
HO O CH3 O OH
Reference solution (a). A 0.40 per cent w/v solution of
O
H3CO O OR CH3 spiramycin RS in methanol.
O OH
CH3 Reference solution (b). A 0.040 per cent w/v solution of
spiramycin RS in methanol.
C43H74N2O14 Mol. Wt. 843.1
Reference solution (c). A 0.020 per cent w/v solution of
Spiramycin is a mixture comprised primarily of spiramycin I spiramycin RS in methanol.
produced by Streptomyces ambofacien from soil of
northern France. Reference solution (d). A 0.0080 per cent w/v solution of
spiramycin RS in methanol.
Spiramycin contains not less than 3900 Units per mg, calculated
on the dried basis. Reference solution (e). A 0.40 per cent w/v of erythromycin
RS in methanol.
Description. A white or slightly yellowish powder; odour,
slight; slightly hygroscopic. Apply to the plate 5 µl of each solution. After development,
dry the plate in air and spray with ethanolic anisaldehyde
Identification solution and heat at 110° for 5 minutes. In the chromatogram
obtained with reference solution (a) there are two spots with
A. When examined in the range 220 nm to 360 nm (2.4.7), a Rf values slightly higher than that of the principal spot; the
0.001 per cent w/v solution in methanol shows an absorption spot nearer the principal spot corresponds to the monoacetate
maximum only at about 232 nm; absorbance at about 232 nm, ester and the one farther from the principal spot corresponds
about 0.34. to the monopropionate ester. In the chromatogram obtained
B. In the test for Related substances the principal spot in the with the test solution any spot corresponding to the
chromatogram obtained with the test solution corresponds to monoacetate is not more intense than the spot in the
that in the chromatogram obtained with reference solution (a). chromatogram obtained with reference solution (c), any spot
If in the chromatogram obtained with the test solution one or corresponding to the monopropionate is not more intense
two additional spots appear with Rf values slightly higher than the spot in the chromatogram obtained with reference
than that of the principal spot, these spots correspond to the solution (b) and any other secondary spot is not more intense
secondary spots in the chromatogram obtained with reference than the spot in the chromatogram obtained with reference
solution (a) but differ from the spots in the chromatogram solution (d).
obtained with reference solution (e). Sulphated ash (2.3.18). Not more than 0.1 per cent w/v.
C. Dissolve 0.5 g in a mixture of 10 ml of 0.05 M sulphuric acid Loss on drying (2.4.19). Not more than 3.5 per cent, determined
and 25 ml of water. Adjust the pH to about 8 by addition of 0.1 on 0.5 g by drying over phosphorus pentoxide at 80° at a
M sodium hydroxide and dilute to 50 ml with water. To 5 ml of pressure not exceeding 0.7 kPa for 6 hours.
the resulting solution add 2 ml of a mixture of 1 volume of
Assay. Carry out the microbiological assay of antibiotics
water and 2 volumes of sulphuric acid; a brown colour is
(2.2.10), Method A.
produced.
Storage. Store protected from moisture.
Tests
pH (2.4.24). 8.5 to 10.5, determined in a solution prepared by Sulphadiazine and Trimethoprim
dissolving 0.5 g in 5 ml of methanol and diluting to 100 ml with
carbon dioxide-free water. Injection
Specific optical rotation (2.4.22). –80.0° to –85.0°, determined Trimethoprim and Sulphadiazine Injection; Co-trimazine
in a 2 per cent w/v solution in 0.2 M acetic acid. Injection
Heavy metals (2.3.13). 1.0 g complies with the limit test for Sulphadiazine and Trimethoprim Injection is a sterile
heavy metals, Method A (20 ppm). suspension in Water for Injections containing Sulphadiazine
Related substances. Determine by thin-layer chromatography and Trimethoprim in the proportion of five parts to one part
(2.4.17), coating the plate with silica gel G. respectively.

1562
IP 2007 SULPHADIAZINE AND TRIMETHOPRIM VETERINARY ORAL POWDER

Sulphadiazine and Trimethoprim Injection contains not less 200.0 ml with water. Dilute 10.0 ml of this solution to 100.0 ml
than 90.0 per cent and not more than 110.0 per cent of the with water. To 3.0 ml of the resulting solution add 1 ml of 2 M
stated amounts of sulphadiazine, C 10H 10N 4O 2 S and of hydrochloric acid and 1 ml of a 0.1 per cent w/v solution of
trimethoprim, C14H18N4O3. sodium nitrite and allow to stand for 2 minutes. Add 1 ml of a
0.5 per cent w/v solution of ammonium sulphamate and allow
Identification to stand for 3 minutes. Add 1 ml of a 0.1 per cent w/v solution
of N-(1-naphthyl)ethylenediamine dihydrochloride, allow to
A. When examined in the range 230 nm to 360 nm (2.4.7), the
stand for 10 minutes, add sufficient water to produce 25.0 ml
solution obtained in the Assay for trimethoprim shows an
and measure the absorbance of the resulting solution at the
absorption maximum only at about 271 nm.
maximum at about 538 nm (2.4.7). Calculate the content of
B. Determine by thin-layer chromatography (2.4.17), coating C10H10N4O2S from the absorbance obtained by carrying out
the plate with silica gel GF254. the procedure simultaneously, using 3.0 ml of a solution
Mobile phase. A mixture of 75 volumes of ethyl acetate, 15 prepared by dissolving 200 mg of sulphadiazine RS in 50 ml
volumes of dimethylformamide and 5 volumes of water. of 0.1 M sodium hydroxide, adding sufficient water to produce
200.0 ml, diluting 5.0 ml to 250.0 ml with water and beginning
Test solution. Add 4 ml of hydrochloric acid to 2.5 ml of the at the words “add 1 ml of 2 M hydrochloric acid........”.
well-mixed contents of the container and dilute to 50 ml with
1.4 M methanolic ammonia. For trimethoprim — Extract the dichloromethane solution
reserved in the Assay for sulphadiazine with three quantities,
Reference solution (a). A 2.0 per cent w/v of sulphadiazine each of 100 ml, 50 ml and 50 ml, of 1 M acetic acid and dilute
RS in 1.4 M methanolic ammonia. the combined extracts to 500.0 ml with 1 M acetic acid. To 5.0
Reference solution (b). A 0.4 per cent w/v of trimethoprim RS ml add 35 ml of 1 M acetic acid and sufficient water to produce
in 1.4 M methanolic ammonia. 200.0 ml and measure the absorbance of the resulting solution
at the maximum at about 271 nm (2.4.7). Calculate the content
Apply to the plate 1 µl of each solution. After development,
of C14H18N4O3 taking 204 as the specific absorbance at 271 nm.
dry the plate in air and examine in ultraviolet light at 254 nm.
One of the principal spots in the chromatogram obtained with Labelling. The label states the content of Sulphadiazine and
the test solution corresponds to the principal spot in the Trimethoprim in a suitable dose-volume.
chromatogram obtained with reference solution (a) and the
other corresponds to the principal spot in the chromatogram
obtained with reference solution (b). Sulphadiazine and Trimethoprim
C. To 5 ml of the filtrate obtained in the Assay for sulphadiazine Veterinary Oral Powder
add 10 ml of water and 5 ml of thiobarbituric acid-citrate
buffer. Mix and heat on a water-bath for 30 minutes; a pink Trimethoprim and Sulphadiazine Veterinary Oral Powder;
colour is produced. Sulphadiazine and Trimethoprim Dispersible Powder;
Co-trimazine Veterinary Oral Powder
Tests
Sulphadiazine and Trimethoprim Veterinary Oral Powder
pH (2.4.24). 10.0 to 10.5. consists of Sulphadiazine and Trimethoprim in the proportion
of five parts to one part respectively, mixed with suitable
Other tests. Complies with the tests stated under Parenteral
wetting, dispersing and suspending agents.
Preparations (Injections).
Sulphadiazine and Trimethoprim Veterinary Oral Powder
Assay. For sulphadiazine — Disperse the trimethoprim evenly contains not less than 92.5 per cent and not more than 107.5
throughout the injection solution by gently inverting the per cent of the stated amounts of sulphadiazine, C10H10N4O2S,
container several times without foam formation. Transfer an and of trimethoprim, C14H18N4O3.
accurately measured quantity of the injection containing 2 g
of Sulphadiazine to a separating funnel containing 50 ml of Identification
0.1 M sodium hydroxide and extract with two quantities, each
of 100 ml and 50 ml of dichloromethane, washing the extract Determine by thin-layer chromatography (2.4.17), coating the
with the same 25-ml quantity of 0.1 M sodium hydroxide. plate with silica gel GF254.
Reserve the combined dichloromethane extracts for the assay Mobile phase. A mixture of 75 volumes of ethyl acetate, 15
for trimethoprim. volumes of dimethylformamide and 5 volumes of water.
Dilute the combined aqueous solutions and washings to Test solution (a). The supernatant liquid obtained by shaking
250.0 ml with water and filter, and dilute 5.0 ml of the filtrate to a quantity of the powder containing 0.2 g of Sulphadiazine

1563
SULPHADIAZINE AND TRIMETHOPRIM VETERINARY ORAL SUSPENSION IP 2007

with sufficient 1.4 M methanolic ammonia to produce 100 ml 2.0 ml of a 0.0025 per cent w/v solution of sulphadiazine RS in
and centrifuging. 0.0005 M sodium hydroxide and beginning at the words “add
0.5 ml of 4 M hydrochloric acid.......”.
Test solution (b). The supernatant liquid obtained by shaking
a quantity of the powder containing 0.2 g of Trimethoprim For trimethoprim - Extract the combined dichloromethane
with sufficient 1.4 M methanolic ammonia to produce 100 ml extracts from the Assay for sulphadiazine with four quantities,
and centrifuging. each of 50 ml, of a 5 per cent v/v solution of 6 M acetic acid;
wash the combined aqueous extracts with 5 ml of
Reference solution (a). A 0.2 per cent w/v solution of
dichloromethane, discard the dichloromethane layer and
sulphadiazine RS in 1.4 M methanolic ammonia.
dilute to 250.0 ml with a 5 per cent v/v solution of 6 M acetic
Reference solution (b). A 0.2 per cent w/v solution of acid. Dilute 20.0 ml to 100.0 ml with water and determine the
trimethoprim RS in 1.4 M methanolic ammonia. absorbance of the resulting solution at the maximum at about
Apply to the plate 5 µl of each solution. After development, 271 nm (2.4.7). Calculate the content of C14H18N4O3 taking 204
dry the plate in air and spray with a 0.1 per cent w/v solution as the specific absorbance at 271 nm.
of 4-dimethylaminobenzaldehyde in a mixture of 1 ml of
hydrochloric acid and 100 ml of ethanol (95 per cent), allow
to dry and spray with dilute potassium iodobismuthate
solution. The spot in the chromatogram obtained with test
Sulphadiazine and Trimethoprim
solution (a) having Rf value of about 0.7 corresponds to the Veterinary Oral Suspension
principal spot in the chromatogram obtained with reference
Sulphadiazine and Trimethoprim Mixture; Trimethoprim
solution (a). The spot in the chromatogram obtained with test
and Sulphadiazine Veterinary Oral Suspension;
solution (b) having Rf value of about 0.3 corresponds to the
principal spot in the chromatogram obtained with reference Co-trimazine Oral Suspension; Co-trimazine Mixture
solution (b). Sulphadiazine and Trimethoprim Veterinary Oral Suspension
is a suspension of Sulphadiazine and Trimethoprim in the
Tests proportion of five parts to one part respectively, containing
Other tests. Complies with the tests stated under Veterinary suitable suspending and dispersing agents. It may contain
Oral Powders. suitable antimicrobial preservatives.

Assay. For sulphadiazine - Weigh accurately a quantity of Sulphadiazine and Trimethoprim Veterinary Oral Suspension
the powder containing about 0.125 g of Sulphadiazine, transfer contains not less than 90.0 per cent and not more than 110.0
into a separator containing 20 ml of 0.1 M sodium hydroxide per cent of the stated amounts of sulphadiazine, C10H10N4O2S,
and extract with four quantities, each of 50 ml, of and of trimethoprim, C14H18N4O3.
dichloromethane. Wash each dichloromethane extract with
Identification
the same two quantities, each of 10 ml, of 0.1 M sodium
hydroxide. Combine the aqueous washings and the aqueous Determine by thin-layer chromatography (2.4.17), coating the
layer from the separator and reserve the combined plate with silica gel GF254.
dichloromethane extracts for the Assay for trimethoprim.
Mobile phase. A mixture of 75 volumes of ethyl acetate, 15
Dilute the combined aqueous solutions to 250.0 ml with water, volumes of dimethylformamide and 5 volumes of water.
filter and dilute 10.0 ml of the filtrate to 200.0 ml with water. To
Test solution (a). A dilution of the oral suspension in 1.4 M
2.0 ml of the resulting solution add 0.5 ml of 4 M hydrochloric
methanolic ammonia containing the equivalent of 0.2 per cent
acid and 1 ml of a 0.1 per cent w/v solution of sodium nitrite
w/v of Sulphadiazine.
and allow to stand for 2 minutes. Add 1 ml of a 0.5 per cent w/
v solution of ammonium sulphamate and allow to stand for 3 Test solution (b). A dilution of the oral suspension in 1.4 M
minutes. Add 1 ml of a 0.1 per cent w/v solution of methanolic ammonia containing the equivalent of 0.2 per cent
N-(1-naphthyl)ethylenediamine dihydrochloride, allow to w/v of Trimethoprim.
stand for 10 minutes. Dilute the solution to 25.0 ml with water Reference solution (a). A 0.2 per cent w/v solution of
and measure the absorbance of the resulting solution at the sulphadiazine RS in 1.4 M methanolic ammonia.
maximum at about 538 nm (2.4.7), using as the blank a solution
prepared in the same manner using 2 ml of water and beginning Reference solution (b). A 0.2 per cent w/v solution of
at the words “add 0.5 ml of 4 M hydrochloric acid........”. trimethoprim RS in 1.4 M methanolic ammonia.
Calculate the content of C10H10N4O2S from the absorbance Apply to the plate 5 µl of each solution. After development,
obtained by carrying out the procedure simultaneously, with dry the plate in air and spray with a 0.1 per cent w/v solution

1564
IP 2007 SULPHAQUINOXALINE

of 4-dimethylaminobenzaldehyde in a mixture of 1 ml of Determine the weight per ml of the suspension (2.4.29), and
hydrochloric acid and 100 ml of ethanol (95 per cent), allow calculate the contents of sulphadiazine and trimethoprim,
to dry and spray with dilute potassium iodobismuthate weight in volume.
solution. The spot in the chromatogram obtained with test
Labelling. The label states the strength in terms of the amounts
solution (a) having Rf value of about 0.7 corresponds to the
of Sulphadiazine and Trimethoprim.
principal spot in the chromatogram obtained with reference
solution (a). The spot in the chromatogram obtained with test
solution (b) having Rf value of about 0.3 corresponds to the
principal spot in the chromatogram obtained with reference Sulphaquinoxaline
solution (b).
NH2
Tests H
N N
S
Other tests. Complies with the tests stated under Veterinary
O O
Oral Liquids. N
Assay. For sulphadiazine - Transfer an accurately weighed C14H12N4O2S Mol. Wt. 300.3
quantity of the oral suspension containing about 0.125 g of
Sulphadiazine, into a separator containing 20 ml of 0.1 M Sulphaquinoxaline is 4-amino-N-2-quinoxalinylbenzene-
sodium hydroxide and extract with four quantities, each of 50 sulphonamide.
ml, of dichloromethane. Wash each dichloromethane extract Sulphaquinoxaline contains not less than 98.0 per cent and
with the same two quantities, each of 10 ml, of 0.1 M sodium not more than 101.0 per cent of C14H12N4O2S, calculated on the
hydroxide. Combine the aqueous washings and the aqueous dried basis.
layer from the separator and reserve the combined
dichloromethane extracts for the Assay for trimethoprim. Description. A yellow colour powder.

Dilute the combined aqueous solutions to 250.0 ml with water, Identification


filter and dilute 10.0 ml of the filtrate to 200.0 ml with water. To
2.0 ml of the resulting solution add 0.5 ml of 4 M hydrochloric A. Determine by infrared absorption spectrophotometry (2.4.6).
acid and 1 ml of a 0.1 per cent w/v solution of sodium nitrite Compare the spectrum with that obtained with
and allow to stand for 2 minutes. Add 1 ml of a 0.5 per cent w/ sulphaquinoxaline RS or with the reference spectrum of
v solution of ammonium sulphamate and allow to stand for 3 sulphaquinoxaline.
minutes. Add 1 ml of a 0.1 per cent w/v solution of B. When examined in the range 230 nm to 360 nm (2.4.7), a
N-(1-naphthyl)ethylenediamine dihydrochloride, allow to 0.001 per cent w/v solution in 0.01 M sodium hydroxide shows
stand for 10 minutes. Dilute the solution to 25.0 ml with water an absorption maximum only at about 252 nm; absorbance at
and measure the absorbance of the resulting solution at the about 252 nm, about 1.1.
maximum at about 538 nm (2.4.7), using as the blank a solution
C. Dissolve 4 mg in 2 ml of warm 2 M hydrochloric acid. The
prepared in the same manner using 2 ml of water and beginning
solution gives the reaction of primary aromatic amines (2.3.1).
at the words “add 0.5 ml of 4 M hydrochloric acid........”.
Calculate the content of C10H10N4O2S from the absorbance Tests
obtained by carrying out the procedure simultaneously, with Acidity. To 2 g add 100 ml of water, heat at 70° for 5 minutes,
2.0 ml of a 0.0025 per cent w/v solution of sulphadiazine RS in cool to 20°, and filter. Titrate 50 ml of the filtrate to pH 7.0 with
0.0005 M sodium hydroxide and beginning at the words “add 0.1 M sodium hydroxide; not more than 0.2 ml of 0.1 M sodium
0.5 ml of 4 M hydrochloric acid.......”. hydroxide is required.
For trimethoprim - Extract the combined dichloromethane Heavy metals. Dissolve the residue obtained in the test for
extracts from the Assay for sulphadiazine with four quantities, Sulphated ash in 1 ml of 2 M hydrochloric acid and dilute to
each of 50 ml, of a 5 per cent v/v solution of 6 M acetic acid; 14 ml with water. 12 ml of the solution complies with limit test
wash the combined aqueous extracts with 5 ml of for heavy metals, Method D (2.3.13) (20 ppm).
dichloromethane, discard the dichloromethane layer and
dilute to 250.0 ml with a 5 per cent v/v solution of 6 M acetic Related substances. Determine by thin-layer chromatography
acid. Dilute 20.0 ml to 100.0 ml with water and determine the (2.4.17), coating the plate with silica gel GF254.
absorbance of the resulting solution at the maximum at about Mobile phase. A mixture of 60 volumes of dichloromethane,
271 nm (2.4.7). Calculate the content of C14H18N4O3 taking 204 40 volumes of methanol and 20 volumes of strong ammonia
as the specific absorbance at 271 nm. solution.

1565
SULPHAQUINOXALINE SODIUM SOLUTION IP 2007

Test solution. Dissolve 0.20 g of the substance under sulphaquinoxaline RS or with the reference spectrum of
examination in 2 ml of 1 M sodium hydroxide and add sufficient sulphaquinoxaline.
methanol to produce 50 ml.
B. Dissolve 4 mg of the residue obtained in test A in 2 ml of
Reference solution (a). A 0.012 per cent w/v solution of warm 2 M hydrochloric acid. The solution gives the reaction
N1,N2-diquinoxalin- 2-ylsulphanilamide RS in methanol. of primary aromatic amines (2.3.1).
Reference solution (b). A 0.0040 per cent w/v solution of C. Acidify with 6 M acetic acid, filter and evaporate the filtrate
sulphanilamide RS in methanol. to dryness. The incinerated residue, when moistened with
Apply to the plate 5 µl of each solution. After development, hydrochloric acid and introduced on a platinum wire into a
dry the plate in air until the odour of the solvent is no longer Bunsen burner flame, gives a yellow colour to the flame.
detectable and examine in ultraviolet light at 254 nm. Any spot
corresponding to N1,N2-diquinoxalin-2-ylsulphanilamide in the Tests
chromatogram obtained with the test solution not more intense pH (2.4.24). 12.2 to 12.8, determined in a 9.6 per cent w/v
than that of the spot in the chromatogram obtained with solution in carbon dioxide-free water.
reference solution (a). Any other secondary spot in the
chromatogram obtained with the test solution is not more Related substances. Determine by thin-layer chromatography
intense than that in the chromatogram obtained by reference (2.4.17), coating the plate with silica gel GF254.
solution (b). Mobile phase. A mixture of 60 volumes of dichloromethane,
Sulphated ash (2.3.18). Not more than 0.1 per cent. 40 volumes of methanol and 20 volumes of strong ammonia
solution.
Loss on drying (2.4.19). Not more than 1.0 per cent, determined
on 1.0 g by drying in an oven at 105°. Test solution. Dilute a solution containing 0.20 g of
Assay. Weigh accurately about 0.65 g and dissolve in 10 ml of Sulphaquinoxaline to 50 ml with methanol.
a mixture of equal volumes of 1 M sodium hydroxide and Reference solution (a). A 0.012 per cent w/v solution of
water. Add 20 ml of glycerin, 20 ml of 9 M sulphuric acid and N1,N2-diquinoxalin- 2-ylsulphanilamide RS in methanol.
5 g of potassium bromide, cool in ice and carry out the nitrite
titration (2.3.31). Reference solution (b). A 0.0040 per cent w/v solution of
sulphanilamide RS in methanol.
1 ml of 0.1 M sodium nitrite is equivalent to 0.03003 g of
C14H12N4O2S. Apply to the plate 5 µl of each solution. After development,
dry the plate in air until the odour of the solvent is no longer
Storage. Store protected from light. detectable and examine in ultraviolet light at 254 nm. Any spot
corresponding to N1,N2-diquinoxalin-2-ylsulphanilamide in the
chromatogram obtained with the test solution not more intense
than that in the chromatogram obtained with reference
Sulphaquinoxaline Sodium Solution solution (a). Any other secondary spot in the chromatogram
obtained with the test solution is not more intense than that
Sulphaquinoxaline Sodium Solution is an aqueous solution of of the spot in the chromatogram obtained with reference
sulphaquinoxaline sodium prepared by the interaction of solution (b).
Sulphaquinoxaline and Sodium Hydroxide.
Other tests. Complies with the tests stated under Veterinary
Sulphaquinoxaline Sodium Solution contains not less than
Oral Liquids.
95.0 per cent and not more than 105.0 per cent of the stated
amount of sulphaquinoxaline, C14H12N4O2S. Assay. To an accurately measured volume containing about
0.48 g of Sulphaquinoxaline add 30 ml water, 20 ml of glycerin,
Description. A clear, yellow to brown solution.
20 ml of 9 M sulphuric acid and 5 g of potassium bromide,
Identification cool in ice and carry out the nitrite titration (2.3.31).

A. To a volume containing 1 g of Sulphaquinoxaline add 10 ml 1 ml of 0.1 M sodium nitrite is equivalent to 0.03003 g of


of water and 3 ml of 2 M hydrochloric acid, filter, wash the C14H12N4O2S.
precipitate with water and dry for 2 hours at 105°. The residue Storage. Store protected from light.
complies with the following test.
Labelling. The label states the strength in terms of the
Determine by infrared absorption spectrophotometry (2.4.6). equivalent amount of Sulphaquinoxaline in a suitable
Compare the spectrum with that obtained with dose-volume.

1566
IP 2007 THIABENDAZOLE VETERINARY ORAL SUSPENSION

Sulphathiazole Sodium 1 ml of 0.1 M sodium nitrite is equivalent to 0.02773 g of


C9H8N3NaO2S2.
O O S Storage. Store protected from light.
S , 5H2O Labelling. The label states whether the substance is the
Na N N
sesquihydrate or the pentahydrate.
H2N

C9H8N3NaO2S2,H2O Mol. Wt. 304.3


Thiabendazole Veterinary Oral
C9H8N3NaO2S2,5H2O Mol. Wt. 367.4
Suspension
Sulphathiazole Sodium is sodium salt of 4-amino-N-2-
thiazolylbenzenesulphonamide with five or one and half Thiabendazole Oral Suspension; Thiabendazole Mixture;
molecules of water. Thiabendazole Drench
Sulphathiazole Sodium contains not less than 99.0 per cent Thiabendazole Veterinary Oral Suspension is an aqueous
and not more than 101.0 per cent of C9H8N3NaO2S2, calculated suspension of Thiabendazole containing suitable suspending
on the dried basis. agents and antimicrobial preservatives.
Description. A white or yellowish white, crystalline powder or Thiabendazole Veterinary Oral Suspension contains not less
granules. than 95.0 per cent and not more than 105.0 per cent of the
stated amount of thiabendazole, C10H7N3S.
Identification
Identification
A. Determine by infrared absorption spectrophotometry (2.4.6).
Compare the spectrum with that obtained with sulphathiazole Determine by thin-layer chromatography (2.4.17), coating the
sodium RS or with the reference spectrum of sulphathiazole plate with silica gel GF254.
sodium. Mobile phase. A mixture of 50 volumes of toluene, 20 volumes
B. Dissolve 1 g in 25 ml of water and add 2 ml of 6 M acetic of glacial acetic acid, 8 volumes of acetone and 2 volumes of
acid. Wash the precipitate formed with water and dry for 4 water.
hours at 105°. The residue melts at about 201° (2.4.21). Test solution. Add 50 ml of ethyl acetate and 2 ml of glacial
C. The precipitate obtained in test B gives the reaction of acetic acid to a volume of the well-mixed oral suspension
primary aromatic amines (2.3.1). containing about 0.25 g of Thiabendazole. Shake for 5 minutes,
heat to boiling, cool, shake for a further 15 minutes and filter.
Tests Reference solution. Dissolve 0.25 g of thiabendazole RS in 50
pH (2.4.24). 9.0 to 10.0, determined in a 1 per cent w/v solution. ml of ethyl acetate and add 2 ml of glacial acetic acid.
Heavy metals. Dissolve 2.5 g of the substance under Apply to the plate 10 µl of each solution. After development,
examination in 10 ml of water, add 15 ml of 2 M acetic acid, dry the plate in air and examine in ultraviolet light at 254 nm.
shake for 30 minutes and filter. The principal spot in the chromatogram obtained with the test
solution corresponds to that in the chromatogram obtained
12 ml of this solution complies with the limit test for heavy with reference solution.
metals, Method D (2.3.13) (20 ppm).
Related substances. Complies with test A for related Tests
substances in sulphonamides (2.3.7). Other tests. Complies with the tests stated under Veterinary
Loss on drying (2.4.19). Not less than 6.0 per cent and not Oral Liquids.
more than 10.0 per cent (sesquihydrate) or not less than 22.0 Assay. Weigh accurately a quantity of the well-mixed oral
per cent and not more than 27.0 per cent (pentahydrate), suspension containing about 1 g of Thiabendazole, add to
determined on 1.0 g by drying in an oven at 105°. 700 ml of 0.1 M hydrochloric acid, shake for 30 minutes, add
Assay. Weigh accurately about 0.5 g, dissolve in a mixture of sufficient 0.1 M hydrochloric acid to produce 1000.0 ml, mix
75 ml of water and 10 ml of hydrochloric acid, add 3 g of and filter. Dilute 10.0 ml of the filtrate to 100.0 ml with 0.1 M
potassium bromide, cool in ice and carry out the nitrite titration hydrochloric acid. Dilute 5.0 ml to 100.0 ml with 0.1 M
(2.3.31). hydrochloric acid and measure the absorbance of the resulting

1567
THIABENDAZOLE AND RAFOXANIDE VETERINARY ORAL SUSPENSION IP 2007

solution at the maximum at about 302 nm (2.4.7). Calculate the For rafoxanide - Protect the solutions from light throughout
content of C10H7N3S taking 1230 as the specific absorbance at the determination.
302 nm.
Weigh accurately a volume of the well-mixed suspension
Determine the weight per ml of the suspension (2.4.29), and containing about 0.1 g of Rafoxanide in a 500-ml stoppered
calculate the content of thiabendazole, weight in volume. flask and add sufficient water to produce 100 ml. Swirl to
disperse, add 20 ml of 1 M hydrochloric acid, mix well and
Labelling. The label states that the suspension should be
add 300 ml of ethyl acetate. Shake the mixture for 1 hour, set
administered undiluted.
aside for separation of the immiscible layers and centrifuge a
portion of the ethyl acetate layer. Transfer 15.0 ml of the clear
solution to a 50-ml centrifuge tube, add 20 ml of 0.1 M
hydrochloric acid, stopper the tube, shake for 15 minutes,
Thiabendazole and Rafoxanide and centrifuge. Remove and discard the aqueous layer. Repeat
Veterinary Oral Suspension the washing with two quantities, each of 20 ml, of 0.1 M
hydrochloric acid. Evaporate the ethyl acetate solution almost
Thiabendazole and Rafoxanide Suspension; to dryness in a warm water-bath, passing a stream of nitrogen
Thiabendazole and Rafoxanide Mixture over the surface of the liquid. Add 10 ml of water, warm on a
water-bath for 10 minutes, add 5 ml of 1 M sodium hydroxide
Thiabendazole and Rafoxanide Veterinary Oral Suspension is
and mix. Add 15 ml of ether, shake for 15 minutes, centrifuge,
an aqueous suspension of Thiabendazole and Rafoxanide
and remove the ether layer. Repeat the extraction with two
containing suitable suspending and dispersing agents.
quantities, each of 15 ml, of ether. Evaporate the combined
Thiabendazole and Rafoxanide Veterinary Oral Suspension ether extracts almost to dryness on a warm water-bath, passing
contains not less than 92.5 per cent and not more than 107.5 a stream of nitrogen over the surface of the liquid. Dissolve
per cent of the stated amount of thiabendazole, C10H7N3S, and the residue in sufficient 0.1 M methanolic hydrochloric acid
not less than 90.0 per cent and not more than 110.0 per cent of to produce 200.0 ml and measure the absorbance of the
the stated amount of rafoxanide, C19H11Cl2I2NO3. resulting solution at the maximum at about 335 nm (2.4.7).
Identification Calculate the content of C19H11Cl2I2NO3 from the absorbance
obtained by carrying out the procedure simultaneously, using
A. Mix a volume containing 20 mg of Thiabendazole with 5 ml 0.1 g of rafoxanide RS and beginning at the words, “add
of 0.1 M hydrochloric acid, add 3 mg of 4-phenylenediamine sufficient water to produce 100 ml.....”.
dihydrochloride, mix, add 0.1 g of zinc powder and allow to
Determine the weight per ml of the suspension (2.4.29), and
stand for 2 minutes. Add 10 ml of ferric ammonium sulphate
calculate the content of thiabendazole and rafoxanide, weight
solution; a deep blue or blue violet colour is produced.
in volume.
B. In addition to the absorbance at about 335 nm, measure the
absorbance at about 280 nm (2.4.7), of the final solution
obtained in the Assay. The ratio of the absorbance at about Thiabendazole Premix
280 nm to that at about 335 nm is 1.59 to 1.69.
Thiabendazole Premix contains not less than 95.0 per cent and
Tests not more than 105.0 per cent of the stated amount of
thiabendazole, C10H7N3S.
Other tests. Complies with the tests stated under Veterinary
Oral Liquids. Identification
Assay. For thiabendazole - Weigh accurately a volume of the Determine by thin-layer chromatography (2.4.17), coating the
well-mixed suspension containing about 85 mg of plate with silica gel GF254.
Thiabendazole, add 20 ml of water and 9 ml of 0.1 M
hydrochloric acid and warm on a water-bath for 30 minutes Mobile phase. A mixture of 50 volumes of toluene, 20 volumes
with occasional stirring. Transfer the suspension to a flask, of glacial acetic acid, 8 volumes of acetone and 2 volumes of
rinse the vessel with water and add the washings to the flask. water.
Cool, add sufficient water to produce 1000.0 ml and filter. Test solution. To a quantity of the premix containing 0.25 g of
Dilute 5.0 ml of the filtrate to 100.0 ml with 0.1 M hydrochloric Thiabendazole, finely powdered if necessary, add 50 ml of
acid and measure the absorbance of the resulting solution at ethyl acetate and 2 ml of glacial acetic acid, shake for 5
the maximum at about 302 nm (2.4.7). Calculate the content of minutes, heat to boiling, cool, shake for a further 15 minutes
C10H7N3S taking 1230 as the specific absorbance at 302 nm. and filter.

1568
IP 2007 TYLOSIN

Reference solution. Dissolve 0.25 g of thiabendazole RS in 50 (solution A) shows an absorption maximum only at about 290
ml of ethyl acetate and add 2 ml of glacial acetic acid. nm; absorbance at about 290 nm, about 0.94.
Apply to the plate 10 µl of each solution. After development, C. To 10 ml of solution A add 1 ml of 2 M sodium hydroxide,
dry the plate in air and examine in ultraviolet light at 254 nm. heat on a water-bath for 20 minutes and cool. When examined
The principal spot in the chromatogram obtained with the test in the range 250 nm to 430 nm (2.4.7), of the resulting solution
solution corresponds to that in the chromatogram obtained shows an absorption maximum only at about 332 nm.
with the reference solution.
D. In the test for Tylosin A and other tylosins, the retention
Tests time and size of the principal peak in the chromatogram obtained
with the test solution are approximately the same as those of
Other tests. Complies with the tests stated under Premixes. the principal peak in the chromatogram obtained with reference
solution (a).
Assay. Weigh accurately a quantity containing about 0.1 g of
Thiabendazole, add 700 ml of 0.1 M hydrochloric acid, shake E. Dissolve about 30 mg in a mixture of 0.15 ml of water, 2.5 ml
for 30 minutes, dilute to 1000.0 ml with 0.1 M hydrochloric of acetic anhydride and 7.5 ml of pyridine. Allow to stand for
acid and filter. Dilute 5.0 ml of the filtrate to 100.0 ml with 0.1 10 minutes; no green colour develops.
M hydrochloric acid and measure the absorbance of the
resulting solution at the maximum at about 302 nm (2.4.7). Tests
Calculate the content of C10H7N3S, taking 1230 as the specific
pH (2.4.24). 8.5 to 10.5, determined in a 2.5 per cent w/v
absorbance at 302 nm.
suspension in carbon dioxide-free water.
Heavy metals. To the residue obtained in the test for Sulphated
ash add 2 ml of hydrochloric acid and evaporate slowly to
Tylosin dryness on a water-bath. Moisten the residue with 0.05 ml of
hydrochloric acid, add 10 ml of boiling water and heat for 10
O
CH3
minutes on a water-bath. Cool and dilute to 25 ml with water.
CH3
12 ml of the solution complies with the limit test for heavy
H3C
CHO N CH3 metals, Method D (2.3.13) (20 ppm).
H3C H3C
O HO
HO O O
O
O CH3 O OH Tyramine. Dissolve 50 mg in 5 ml of 0.03 M phosphoric acid
H3CO
OCH3 O OH CH3 in a 25-ml volumetric flask, add 1 ml of pyridine and 2 ml of a
O OH
CH3 CH3 saturated solution of ninhydrin in water (approximately 4 per
cent w/v). Close the flask by covering with a piece of aluminium
C46H77NO17 Mol. Wt. 916.1 foil and heat in a water-bath at 85° for at least 20 minutes. Cool
rapidly and add sufficient water to produce 25 ml. Mix and
Tylosin is a macrolide antibiotic isolatede from a strain of measure without delay the absorbance of the solution at about
Stryptomycetes fradiae found in soil from Thailand. 570 nm (2.4.7), using as the blank a solution prepared in a
Tylosin has a potency of not less than 900 Units per mg, similar manner but omitting the substance under examination.
calculated on the dried basis. The content of tylosin A is not The absorbance is not more than that obtained by carrying
less than 80.0 per cent and the sum of the contents of tylosin out the procedure simultaneously, using 5 ml of a solution in
A, tylosin B, tylosin C and tylosin D is not less than 95.0 per 0.03 M phosphoric acid containing 35 mg of tyramine per ml
cent. and beginning at the words “add 1 ml of pyridine......” (0.35
per cent).
Description. Almost white or slightly yellow powder.
Sulphated ash (2.3.18). Not more than 3.0 per cent.
Identification Loss on drying (2.4.19). Not more than 5.0 per cent, determined
Tests B and C may be omitted if tests A, D and E are carried on 1.0 g by drying at 60° at a pressure not exceeding 0.7 kPa
out. Tests D and E may be omitted if tests A, B and C are for 3 hours.
carried out. Tylosin A and other tylosins. Determine by liquid
A. Determine by infrared absorption spectrophotometry (2.4.6). chromatography (2.4.14).
Compare the spectrum with that obtained with tylosin RS or NOTE- Use freshly prepared solutions.
with the reference spectrum of tylosin.
Test solution. Dissolve 20 mg of the substance under
B. When examined in the range 230 nm to 360 nm (2.4.7), a examination in 100 ml of a mixture of equal volumes of
0.004 per cent w/v solution in 0.1 M hydrochloric acid acetonitrile and water.

1569
TYLOSIN INJECTION IP 2007

Reference solution (a). A 0.02 per cent w/v solution of tylosin Tylosin Injection
RS in a mixture of equal volumes of acetonitrile and water.
Tylosin Injection is a sterile solution of Tylosin in a mixture of
Reference solution (b). A solution containing 0.02 per cent w/
equal volumes of Propylene Glycol and Water for Injections.
v each of tylosin A RS and tylosin D RS in a mixture of equal
volumes of acetonitrile and water. Tylosin Injection contains not less than 95.0 per cent and not
Chromatographic system more than 105.0 per cent of the stated amount of tylosin. The
– a stainless steel column 20 cm x 4.6 mm, packed with content of tylosin A is not less than 80.0 per cent and the sum
octadecylsilane bonded to porous silica (5 µm) (such as of the contents of tylosin A, tylosin B, tylosin C and tylosin D
Nucleosil C18), is not less than 90.0 per cent.
– column temperature 35°, Description. A pale yellow to amber-coloured solution.
– mobile phase: a filtered and degassed mixture of 60
volumes of 0.85 M sodium perchlorate and 40 volumes Identification
of acetonitrile adjusted to pH 2.5 with 1 M hydrochloric
acid, A. To a volume containing 0.1 g of Tylosin add sufficient
– flow rate. 1 ml per minute, water to obtain a solution containing 0.02 per cent w/v of
– spectrophotometer set at 290 nm, Tylosin. To 5 ml of this solution add 10 ml of 0.1 M sodium
– a 20 µl loop injector. hydroxide and extract with 10 ml of dichloromethane. Separate
the dichloromethane layer and extract it with 25 ml of 0.1 M
Inject reference solution (b). If necessary, adjust the molarity
hydrochloric acid. Discard the dichloromethane layer, wash
of the sodium perchlorate or increase the temperature of the
the aqueous layer with 3 ml of dichloromethane, discard the
column to a maximum of 50° so as to obtain a retention time of
washings and filter. When examined in the range 230 nm to
about 12 minutes for tylosin A. The test is not valid unless the
360 nm (2.4.7), of the resulting solution exhibits a maximum
resolution between the peaks due to tylosin A and tylosin D is
only at about 290 nm; absorbance at about 290 nm, about 0.94.
at least 2.0.
Inject reference solution (a). The column efficiency, determined B. To 10 ml of the filtrate obtained in test A add 1 ml of 2 M
using the peak due to tylosin A, should be not less than sodium hydroxide, heat in a water-bath for 20 minutes and
22,000 theoretical plates per metre. cool. When examined in the range 250 nm to 430 nm (2.4.7),
exhibits a maximum only at about 332 nm.
Inject alternatively the test solution and reference solution
(a). The order of elution of the major components of the Tests
substance under examination is desmycinosyltylosin, tylosin
C, tylosin B, tylosin D, tylosin A aldol and tylosin A. Tyramine. Dilute a volume containing 100 mg of Tylosin with
5 ml of 0.03 M phosphoric acid in a 25-ml volumetric flask,
Calculate the percentage content of components from the areas add 1 ml of pyridine and 2 ml of a saturated solution of
of the peaks in the chromatogram obtained with the test ninhydrin in water (approximately 4 per cent w/v). Close the
solution by normalisation. flask by covering with a piece of aluminium foil and heat in a
Assay. Carry out the microbiological assay of antibiotics water-bath at 85° for at least 20 minutes. Cool rapidly and add
(2.2.10). sufficient water to produce 25 ml. Mix and measure without
Tylosin intended for use in the manufacture of Parenteral delay the absorbance of the resulting solution at about 570
Preparations without a further appropriate sterilisation nm (2.4.7), using as the blank a solution prepared in a similar
procedure complies with the following additional manner but omitting the preparation under examination. The
requirement. absorbance is not more than that obtained by carrying out the
procedure simultaneously, using 5 ml of a solution in 0.03 M
Sterility (2.2.11). Complies with the test for sterility. phosphoric acid containing 30 mg of tyramine per ml and
Storage. Store protected from light. If it is intended for use in beginning at the words “add 1 ml of pyridine......” (0.15 per
the manufacture of Parenteral Preparations, the container cent).
should be sterile, tamper-evident and sealed so as to exclude Tylosin A and other tylosins. Determine by liquid
micro-organisms. chromatography (2.4.14).
Labelling. The label states (1) the number of Units per mg; (2)
NOTE - Use freshly prepared solutions.
the date after which the material is not intended to be used; (3)
the storage conditions; (4) where applicable, that it is suitable Test solution. Dilute the injection with sufficient of a mixture
for use in the manufacture of Parenteral Preparations; (5) that of equal volumes of acetonitrile and water to produce a
the preparation is intended for veterinary use. solution containing 0.02 per cent w/v of Tylosin.

1570
IP 2007 TYLOSIN TABLETS

Reference solution (a). A 0.02 per cent w/v solution of tylosin Identification
RS in a mixture of equal volumes of acetonitrile and water.
A. Triturate a quantity of the powdered tablets containing 0.2
Reference solution (b). A solution containing 0.02 per cent w/ g of Tylosin with 20 ml of dichloromethane and filter. Dry the
v each of tylosin A RS and tylosin D RS in a mixture of equal dichloromethane extract by shaking with anhydrous sodium
volumes of acetonitrile and water. sulphate, filter and evaporate the filtrate to dryness. Dry the
Chromatographic system residue over phosphorus pentoxide at a pressure not
– a stainless steel column 20 cm x 4.6 mm, packed with exceeding 0.7 kPa for 1 hour.
octadecylsilane bonded to porous silica (5 µm) (such as Determine by infrared absorption spectrophotometry (2.4.6).
Nucleosil C18), Compare the spectrum with that obtained with tylosin RS or
– column temperature 35°, with the reference spectrum of tylosin.
– mobile phase: a filtered and degassed mixture of 60
volumes of 0.85 M sodium perchlorate and 40 volumes B. Triturate a quantity of the powdered tablets containing 0.2
of acetonitrile adjusted to pH 2.5 with 1 M hydrochloric g of Tylosin with two quantities, each of 10 ml, of 0.1 M
acid, hydrochloric acid, filter and dilute the filtrate to 100 ml with
– flow rate. 1 ml per minute, 0.1 M hydrochloric acid. Dilute 10 ml of the resulting solution
– spectrophotometer set at 290 nm, to 50 ml with the same solvent. Dilute 5 ml of this solution
– a 20 µl loop injector. further to 50 ml with the same solvent.
Inject reference solution (b). If necessary, adjust the molarity When examined in the range 230 nm to 360 nm (2.4.7), the
of the sodium perchlorate or increase the temperature of the resulting solution shows an absorption maximum only at about
column to a maximum of 50° so as to obtain a retention time of 290 nm; absorbance at about 290 nm, about 0.94.
about 12 minutes for tylosin A. The test is not valid unless the C. To 10 ml of the final solution obtained in test B add 1 ml of
resolution between the peaks due to tylosin A and tylosin D is 2 M sodium hydroxide, heat in a water-bath for 20 minutes
at least 2.0. and cool. When examined in the range 250 nm to 430 nm (2.4.7),
Inject reference solution (a). The column efficiency, determined exhibits a maximum only at about 332 nm.
using the peak due to tylosin A, should be not less than
22,000 theoretical plates per metre. Tests
Inject alternatively test solution and reference solution (a). Tyramine. Shake a quantity of the powdered tablets containing
The order of elution of the major components of the substance 50 mg of Tylosin with 5 ml of 0.03 M phosphoric acid. Filter
under examination is desmycinosyltylosin, tylosin C, tylosin into a 25-ml volumetric flask, add 1 ml of pyridine and 2 ml of
B, tylosin D, tylosin A aldol and tylosin A. a saturated solution of ninhydrin in water (approximately 4
per cent w/v). Close the flask by covering with a piece of
Calculate the percentage content of components from the areas
aluminium foil and heat in a water-bath at 85° for at least 20
of the peaks in the chromatogram obtained with the test
minutes. Cool rapidly and add sufficient water to produce 25
solution.
ml. Mix and measure without delay the absorbance of the
Other tests. Complies with the tests stated under Parenteral solution at about 570 nm (2.4.7), using as the blank a solution
Preparations (Injections). prepared in a similar manner but omitting the substance under
Assay. Carry out the microbiological assay of antibiotics examination. The absorbance is not more than that obtained
(2.2.10). Calculate the content of tylosin in the injection, taking by carrying out the procedure simultaneously, using 5 ml of a
each 1000 Units found to be equivalent to 1 mg of tylosin. solution in 0.03 M phosphoric acid containing 35 mg of
tyramine per ml and beginning at the words “add 1 ml of
Storage. Store protected from moisture.
pyridine......” (0.35 per cent).
Labelling. The label states that the preparation is intended
for veterinary use by intramuscular injection only. Tylosin A and other tylosins. Determine by liquid
chromatography (2.4.14).
NOTE – Use freshly prepared solutions.
Tylosin Tablets Test solution. Shake a quantity of the powdered tablets
Tylosin Tablets contain not less than 95.0 per cent and not containing 0.2 g of Tylosin with 50 ml of methanol, filter and
more than 105.0 per cent of the stated amount of tylosin. The dilute 5 ml of the filtrate to 100 ml with a mixture of equal
content of tylosin A is not less than 80.0 per cent and the sum volumes of acetonitrile and water.
of the contents of tylosin A, tylosin B, tylosin C and tylosin D Reference solution (a). A 0.02 per cent w/v solution of tylosin
is not less than 90.0 per cent. RS in a mixture of equal volumes of acetonitrile and water.

1571
TYLOSIN TARTRATE IP 2007

Reference solution (b). A solution containing 0.02 per cent w/ strains of Streptomyces fradiae or by any other means. It
v each of tylosin A RS and tylosin D RS in a mixture of equal consists largely of tylosin A tartrate but tartrates of tylosin B
volumes of acetonitrile and water. (desmycosin), tylosin C (macrocin) and tylosin D (relomycin)
may also be present.
Chromatographic system
– a stainless steel column 20 cm x 4.6 mm, packed with Tylosin Tartrate contains not less than 800 Units per mg,
octadecylsilane bonded to porous silica (5 µm) (such as calculated on the dried basis. The content of tylosin A is not
Nucleosil C18), less than 80.0 per cent and the sum of the contents of tylosin
– column temperature 35°, A, tylosin B, tylosin C and tylosin D is not less than 95.0 per
– mobile phase: a filtered and degassed mixture of 60 cent.
volumes of 0.85 M sodium perchlorate and 40 volumes
Description. An almost white or slightly yellow, hygroscopic
of acetonitrile adjusted to pH 2.5 with 1 M hydrochloric
powder.
acid,
– flow rate. 1 ml per minute, Identification
– spectrophotometer set at 290 nm,
– a 20 µl loop injector. Tests B and C may be omitted if tests A, D and E are carried
Inject reference solution (b). If necessary, adjust the molarity out. Tests D and E may be omitted if tests A, B and C are
of the sodium perchlorate or increase the temperature of the carried out.
column to a maximum of 50° so as to obtain a retention time of A. Determine by infrared absorption spectrophotometry (2.4.6).
about 12 minutes for tylosin A. The test is not valid unless the Compare the spectrum with that obtained with tylosin tartrate
resolution between the peaks due to tylosin A and tylosin D is RS or with the reference spectrum of tylosin tartrate.
at least 2.0.
B. When examined in the range 230 nm to 360 nm (2.4.7), a
Inject reference solution (a). The column efficiency, determined 0.004 per cent w/v solution in 0.1 M hydrochloric acid
using the peak due to tylosin A, should be not less than (solution A) shows an absorption maximum only at about 290
22,000 theoretical plates per metre. nm; absorbance at about 290 nm, about 0.88.
C. To 10 ml of solution A add 1 ml of 2 M sodium hydroxide,
Inject alternatively the test solution and reference solution heat on a water-bath for 20 minutes and cool.
(a). The order of elution of the major components of the
substance under examination is desmycinosyltylosin, tylosin When examined in the range 250 nm to 430 nm (2.4.7), the
C, tylosin B, tylosin D, tylosin A aldol and tylosin A. resulting solution shows an absorption maximum only at about
332 nm.
Calculate the percentage content of components from the areas
of the peaks in the chromatogram obtained with test solution. D. In the test for Tylosin A and other tylosins, the retention
time and size of the principal peak in the chromatogram obtained
Other tests. Comply with the tests stated under Tablets. with the test solution are approximately the same as those of
Assay. Carry out the microbiological assay of antibiotics the principal peak in the chromatogram obtained with the
(2.2.10). Calculate the content of tylosin in the tablets, taking reference solution.
each 1000 Units found to be equivalent to 1 mg of tylosin. E. Dissolve about 30 mg in a mixture of 0.15 ml of water, 2.5 ml
of acetic anhydride and 7.5 ml of pyridine. Allow to stand for
10 minutes; a green colour develops.
Tylosin Tartrate
Tests
O pH (2.4.24). 5.0 to 7.2, determined in a 2.5 per cent w/v solution
CH3
in carbon dioxide-free water.
H3C CH3
CHO N CH3 H OH
H3C
O
H3C HO COOH Tyramine. Dissolve 50 mg in 5 ml of 0.03 M phosphoric acid
HO O O O CH3 O OH HOOC
OCH3
O
CH3
HO H in a 25-ml volumetric flask, add 1 ml of pyridine and 2 ml of a
H3CO O OH
OH
CH3
O
CH3 saturated solution of ninhydrin in water (approximately 4 per
2
cent w/v). Close the flask by covering with a piece of aluminium
foil and heat in a water-bath at 85° for at least 20 minutes. Cool
(C46H77NO17)2, C4H6O6 Mol. Wt. 1983.3 rapidly and add sufficient water to produce 25 ml. Mix and
Tylosin Tartrate is the tartrate of Tylosin, which is a mixture of measure without delay the absorbance of the solution at about
antimicrobial macrolides produced by the growth of certain 570 nm (2.4.7), using as the blank a solution prepared in a

1572
IP 2007 TYLOSIN TARTRATE AND SULPHATHIAZOLE SODIUM VETERINARY ORAL POWDER

similar manner but omitting the substance under examination. Assay. Carry out the microbiological assay of antibiotics
The absorbance is not more than that obtained by carrying (2.2.10).
out the procedure simultaneously, using 5 ml of a solution in
Tylosin Tartrate intended for use in the manufacture of
0.03 M phosphoric acid containing 35 mg of tyramine per ml
parenteral preparations complies with the above
and beginning at the words “add 1 ml of pyridine......” (0.35
requirements with the following modification.
per cent).
Tyramine. Carry out the procedure described in the test for
Sulphated ash (2.3.18). Not more than 2.5 per cent.
Tyramine but using 100 mg in 5 ml of 0.03 M phosphoric acid.
Loss on drying (2.4.19). Not more than 4.5 per cent, determined Measure the absorbance of the solution under the conditions
on 1.0 g by drying at 60° at a pressure not exceeding 0.7 kPa described in the test. The absorbance is not more than that
for 3 hours. obtained by simultaneously carrying out the procedure using
5 ml of a solution in 0.03 M phosphoric acid containing 30 mg
Tylosin A and other tylosins. Determine by liquid
of tyramine per ml and beginning at the words “add 1 ml of
chromatography (2.4.14).
pyridine......” (0.15 per cent).
NOTE - Use freshly prepared solutions.
Tylosin Tartrate intended for use in the manufacture of
Test solution. Dissolve a quantity containing 20 mg of tylosin parenteral preparations without a further appropriate
in 100 ml of a mixture of equal volumes of acetonitrile and sterilisation procedure complies with the following
water.. additional requirement.
Reference solution (a). A 0.02 per cent w/v solution of tylosin Sterility (2.2.11). Complies with the test for sterility.
RS in a mixture of equal volumes of acetonitrile and water.
Storage. Store protected from light. If it is intended to be used
Reference solution (b). A solution containing 0.02 per cent w/ in the manufacture of parenteral preparations, the container
v each of tylosin A RS and tylosin D RS in a mixture of equal should be sterile, tamper-evident and sealed so as to exclude
volumes of acetonitrile and water. micro-organisms.
Chromatographic system Labelling. The label states (1) the number of Units per mg; (2)
– a stainless steel column 20 cm x 4.6 mm, packed with the quantity of Tylosin Tartrate in terms of equivalent amount
octadecylsilane bonded to porous silica (5 µm) (such as of tylosin; (3) the date after which the material is not intended
Nucleosil C18), to be used; (4) the storage conditions; (5) where applicable,
– column temperature 35°, that it is suitable for use in the manufacture of parenteral
– mobile phase: a filtered and degassed mixture of 60 preparations; (6) that the preparation is intended for veterinary
volumes of 0.85 M sodium perchlorate and 40 volumes use.
of acetonitrile adjusted to pH 2.5 with 1 M hydrochloric
acid,
– flow rate. 1 ml per minute, Tylosin Tartrate and Sulphathiazole
– spectrophotometer set at 290 nm,
– a 20 µl loop injector.
Sodium Veterinary Oral Powder
Inject reference solution (b). If necessary, adjust the molarity Tylosin Tartrate and Sulphathiazole Sodium Veterinary Oral
of the sodium perchlorate or increase the temperature of the Powder is a mixture of Tylosin Tartrate and Sulphathiazole
column to a maximum of 50° so as to obtain a retention time of Sodium. It contains 3 parts of Sulphathiazole Sodium for 1
about 12 minutes for tylosin A. The test is not valid unless the part, by weight, of Tylosin.
resolution between the peaks due to tylosin A and tylosin D is Tylosin Tartrate and Sulphathiazole Sodium Veterinary Oral
at least 2.0. Powder contains not less than 90.0 per cent and not more than
Inject reference solution (a). The column efficiency, determined 110.0 per cent of the stated amounts of tylosin and
using the peak due to tylosin A, should be not less than sulphathiazole sodium sesquihydrate, C9H8NaO2S2,11/2H2O.
22,000 theoretical plates per metre. Identification
Inject alternatively the test solution and the reference solution
A. Triturate a quantity of the powder containing 0.25 g of
(a). The order of elution of the major components of the
Tylosin with two quantities, each of 25 ml, of dichloromethane
substance under examination is desmycinosyltylosin, tylosin
and filter. Reserve the dichloromethane-insoluble matter for
C, tylosin B, tylosin D, tylosin A aldol and tylosin A.
test B. Wash the combined filtrates by shaking for 1 minute
Calculate the percentage content of components from the areas with 20 ml of 0.1 M sodium hydroxide and dry the
of the peaks in the chromatogram obtained with test solution. dichloromethane layer by the addition of anhydrous sodium

1573
TYLOSIN TARTRATE AND SULPHATHIAZOLE SODIUM VETERINARY ORAL POWDER IP 2007

sulphate. Evaporate the filtrate to dryness and dry the residue Nucleosil C18),
over phosphorus pentoxide at a pressure not exceeding 0.7 – column temperature 35°,
kPa for 1 hour. The residue complies with the following test. – mobile phase: a filtered and degassed mixture of 60
Determine by infrared absorption spectrophotometry (2.4.6). volumes of 0.85 M sodium perchlorate and 40 volumes
Compare the spectrum with that obtained with tylosin RS or of acetonitrile adjusted to pH 2.5 with 1 M hydrochloric
with the reference spectrum of tylosin. acid,
– flow rate. 1 ml per minute,
B. Dry the dichloromethane-insoluble matter reserved in test – spectrophotometer set at 290 nm,
A at 105° for 1 hour. The residue complies with the following – a 20 µl loop injector.
test.
Inject reference solution (b). If necessary, adjust the molarity
Determine by infrared absorption spectrophotometry (2.4.6).
of the sodium perchlorate or increase the temperature of the
Compare the spectrum with that obtained with sulphathiazole
column to a maximum of 50° so as to obtain a retention time of
sodium RS or with the reference spectrum of sulphathiazole
about 12 minutes for tylosin A. The test is not valid unless the
sodium.
resolution between the peaks due to tylosin A and tylosin D is
Tests at least 2.0.
Inject reference solution (a). The column efficiency, determined
Sulphonamide-related substances. Determine by thin-layer
using the peak due to tylosin A, should be not less than
chromatography (2.4.17), coating the plate with silica gel H.
22,000 theoretical plates per metre.
Solvent mixture. A mixture of 9 volumes of ethanol (95 per
cent) and 1 volume of strong ammonia solution. Inject alternatively the test solution and the reference solution
(a). The order of elution of the major components of the
Mobile phase. A mixture of 90 volumes of 1-butanol and 18 substance under examination is desmycinosyltylosin, tylosin
volumes of 10 M ammonia. C, tylosin B, tylosin D, tylosin A aldol and tylosin A.
Test solution. Shake a quantity of the powder containing 0.1 g Calculate the percentage content of components from the areas
of sulphathiazole sodium sesquihydrate with 10 ml of the of the peaks in the chromatogram obtained with the test
solvent mixture. solution by normalisation. In the chromatogram obtained with
Reference solution. A 0.0050 per cent w/v solution of the test solution the content of tylosin A is not less than 80.0
sulphanilamide in the solvent mixture. per cent and the sum of the contents of tylosin A, tylosin B,
Apply to the plate 10 µl of each solution. After development, tylosin C and tylosin D is not less than 95.0 per cent.
dry the plate by heating it at 105° for 10 minutes and spray Other tests. Complies with the tests stated under Veterinary
with a 0.1 per cent w/v solution of Oral Powders.
4-dimethylaminobenzaldehyde in a mixture of 99 volumes of
Assay. For tylosin activity - Weigh accurately a quantity of
ethanol (95 per cent) and 1 volume of hydrochloric acid.
the powder containing about 0.2 g of Tylosin, transfer to a
Any secondary spot in the chromatogram obtained with the
100-ml volumetric flask with three quantities, each of 10 ml, of
test solution is not more intense than the spot in the
methanol, swirl to dissolve and add sufficient sterile
chromatogram obtained with reference solution (0.5 per cent).
phosphate buffer pH 7.0 to produce 100.0 ml. Filter and dilute
Tylosin A and other tylosins. Determine by liquid 5.0 ml of the filtrate to 100.0 ml with sterile phosphate buffer
chromatography (2.4.14). pH 7.0. Carry out the microbiological assay of antibiotics
NOTE - Use freshly prepared solutions. (2.2.10). Calculate the content of tylosin taking each 1000 Units
found to be equivalent to 1 mg of tylosin.
Test solution. Dissolve a quantity containing 20 mg of tylosin
in 100 ml of a mixture of equal volumes of acetonitrile and For sulphathiazole sodium - Weigh accurately a quantity of
water. the powder containing about 0.4 g of sulphathiazole sodium
Reference solution (a). A 0.02 per cent w/v solution of tylosin sesquihydrate, dissolve in a mixture of 75 ml of water and 10
RS in a mixture of equal volumes of acetonitrile and water. ml of hydrochloric acid, add 3 g of potassium bromide, cool
in ice and titrate slowly with 0.1 M sodium nitrite, stirring
Reference solution (b). A solution containing 0.02 per cent w/ constantly and determine the end-point potentiometrically
v each of tylosin A RS and tylosin D RS in a mixture of equal (2.4.25).
volumes of acetonitrile and water.
1 ml of 0.1 M sodium nitrite is equivalent to 0.03043 g of
Chromatographic system C9H8N3NaO2S2,11/2H2O.
– a stainless steel column 20 cm x 4.6 mm, packed with
octadecylsilane bonded to porous silica (5 µm) (such as Storage. Store protected from moisture.

1574
IP 2007 TYLOSIN TARTRATE AND SULPHATHIAZOLE SODIUM VETERINARY ORAL POWDER

Labelling. The label states the strength of Tylosin Tartrate in Sulphathiazole Sodium in terms of the equivalent amount of
terms of the equivalent amount of tylosin and that of sulphathiazole sodium sesquihydrate.

1575
IP 2007 AVIAN INFECTIOUS BRONCHITIS VACCINE, INACTIVATED

Anthrax Spore Vaccine, Live stated on the label for sheep and observe the animal for 21
days. None of the sheep shows any untoward reaction. If
Anthrax Spore Vaccine, Live consists of a suspension of live more than 2 animals die from non-specific causes, repeat the
spores of an attenuated, non-capsulated strain of Bacillus test.
anthracis.
Inject subcutaneously into each vaccinated guinea pig or
Production rabbit or sheep at least 100 MLD and to each of these control
animals 10 MLD of a strain of B.anthracis pathogenic for the
The strain used may either be not lethal to guinea pigs or the species of animal used in the test. Observe all the animals for
mouse or lethal to guinea pigs but not to the rabbit or lethal to 10 days, all vaccinated animals survive and all the controls die
some rabbits. At the end of growth the spores are suspended from anthrax during the observation period. If a vaccinated
in 50 per cent glycerin saline and counted. The vaccine may animal dies after the challenge, repeat the test. If in the second
contain an adjuvant. test, a vaccinated animal dies, the vaccine fails the test.
Identification Labelling. The label states (1) the strain used for the
preparation of the vaccine; (2) the number of viable spores
B. anthracis present in the vaccine is identified by means of
per ml.
morphological, serological test, culture and biochemical test.

Tests
Avian Infectious Bronchitis Vaccine,
Safety. Carry out the test on one of the species for which the Inactivated
vaccine is intended. If the vaccine is intended for several
species including the goat, carry out the test on sheep and Avian Infectious Bronchitis Vaccine, Inactivated consists of
goats. Administer subcutaneously or intramuscularly to each an emulsion or a suspension of one or more serotypes of
of two animals having no antibodies against B. anthracis, avian infectious bronchitis virus which have been inactivated
twice the dose (example, 2 million spores) stated on the label in such a manner that the immunogenic activity is retained.
for the species used and observe the animals for 10 days. No The vaccine may contain one or more suitable adjuvants.
abnormal systemic reaction is produced but a mild local These vaccines intended to protect against a drop in egg
reaction may occur at the site of injection. The severity of the production or quality; for vaccines also intended to protect
local reaction may vary according to the strain of the spores against respiratory symptoms, a demonstration of efficacy
and the adjuvants used in the preparation, but necrosis does (2.7.12) additional to that described under Potency is required.
not occur.
Production
Sterility (2.2.11). Complies with the test for sterility.
The virus is propagated in fertilised hen eggs (2.7.7) from SPF
Spore count. Determine the number of viable spores by plate flock or in suitable cell cultures. The master seed lot complies
count. The number of live spores is not less than 80 per cent with the tests for extreneous agents in seed lot (2.7.10).
of that stated on the label.
Inactivation
Potency. For a strain of B. anthracis which is not lethal to the
guinea pig or the mouse, the test may be carried out in the An amplification test for residual live avian infectious
guinea pigs. For a strain which is lethal to the guinea pig but bronchitis virus is carried out on each batch of antigen
not to the rabbit, the test may be carried out in rabbits. For a immediately after inactivation and on the final bulk vaccine or,
strain which is lethal to some rabbits, carry out the test in if the vaccine contains an adjuvant, on the bulk antigen or
sheep. mixture of bulk antigens immediately before the addition of
adjuvant; the test is carried out in fertilised hen eggs from
If the test is carried out in guinea pigs or rabbits. Inject into flocks free from specified pathogens (SPF) or in suitable cell
animals (each guinea pig weighing not less than 500 g and cultures and the quantity of inactivated virus used is
each rabbit weighing not less than 4.5 kg) subcutaneously or equivalent to not less than 10 doses of vaccine. No live virus
intramuscularly, 1/10th of the smallest dose of the vaccine is detected.
stated on the label for sheep. Observe the animals for 21
days. If more than two animals die from non-specific causes, Identification
repeat the test.
In susceptible birds, the vaccine stimulates the production of
If the test is carried out in sheep use 5 healthy animals. Inject specific antibodies against each of the virus serotypes in the
subcutaneously or intramuscularly into each sheep weighing vaccine, detectable by virus neutralisation and protects
not less than 20 kg, 1/10th of the smallest dose of the vaccine chicken against infectious bronchitis.

1577
AVIAN INFECTIOUS BRONCHITIS VACCINE, LIVE IP 2007

Tests (2.7.7 ). Use ten similar chickens as controls and house them
together with the vaccinated chickens. After 28 days, collect
Sterility (2.2.11). Complies with the test for sterility. serum samples from each of the vaccinated and control
Safety. Inject intramuscularly a quantity equivalent to 2 doses chickens and perform haemagglutination inhibition (HI) test
into each of twenty healthy chickens, 2 to 4 weeks old, free on each serum using 8 haemagglutinating (HA) units of antigen
from specific antibodies (2.7.7). Observe the chickens for 14 and chicken erythrocytes, testing all serum samples at the
days. No abnormal systemic or local reaction is seen. same time. The vaccine passes the test if the mean antibody
titre of the vaccinated group is not less than 1:64 and no
Inactivation specific antibody is detected in the control chickens.
Alternatively, serum neutralization test may be carried out in
A. For vaccine prepared with embryo-adapted strains of virus, SPF eggs. Serum neutralization titer should not be less than
inject 2/5 of a dose into the allantoic cavity of ten 9 to 11-day- 102 neutralization units.
old fertilised hen eggs from an SPF flock and incubate. Observe
for 5 to 6 days and pool separately the allantoic liquid from If the immunogenicity test (potency test) has been performed
eggs containing live embryos and that from eggs containing with satisfactory results on representative batch of the vaccine
dead embryos, excluding those that die within the first 24 hours from the same seed lot, it may be omitted as a routine control
after injection. Examine for abnormalities all embryos which test during production of other batches of the vaccine prepared
die after 24 hours of injection or which survive 5 to 6 days. No from the same seed lot.
death or abnormality attributable to the vaccine virus occurs. Labelling. The label states (1) the strain of virus used in
Inject into the allantoic cavity of each of ten 9 to 11-day-old preparing the vaccine; (2) the name of any added adjuvant.
fertilised hen eggs from an SPF flock, 0.2 ml of the pooled
allantoic liquid from the live embryos and into each of 10 similar
eggs 0.2 ml of the pooled liquid from the dead embryos and Avian Infectious Bronchitis Vaccine,
incubate for 5 to 6 days. Examine for abnormalities all embryos
which die after 24 hours of injection or which survive 5 to Live
6 days. No death or abnormality attributable to the vaccine Avian Infectious Bronchitis Vaccine, live is a preparation of
virus occurs. one or more suitable strains of different types of avian
If more than 20 per cent of the embryos die at either stage infectious bronchitis virus.
repeat the test from that stage. The vaccine complies with the
test if there is no death or abnormality attributable to the Production
vaccine virus. The vaccine virus is grown in embryonated hens’ eggs or in
B. For vaccine prepared with cell-culture-adapted strains of cell cultures.
virus, inoculate 10 doses of the vaccine into suitable cell
cultures. If the vaccine contains an oil adjuvant, eliminate it Substrate for virus propagation
by suitable means. Incubate at 37 ± 1° for 7 days. Make a Embryonated hens’ eggs. If the vaccine virus is grown in
passage on another set of cell cultures and incubate at 37 ± 1° embryonated hens’ eggs, they are obtained from SPF (2.7.7).
for 7 days. None of the cultures shows signs of infection.
Identification
Extraneous agents. Use the chickens from the test for safety.
21 days after injection of the double dose of vaccine, inject Carry out either the test A or B.
1 dose by the same route into each chicken. Collect serum
A. Inoculate 0.2 ml undiluted vaccine in the allantoic sac of
samples from each chicken 2 weeks later and carry out tests
SPF embryonated eggs and incubate at 37° for 7 days. Lesions
for antibodies to the following agents: avian encephalomyelitis
typical of infectious bronchitis are observed in the embryos
virus, avian leucosis viruses, haemagglutinating avian
and the allantoic fluid does not agglutinate chicken
adenovirus, infectious bursal disease virus, infectious
erythrocytes.
laryngotracheitis virus, influenza A virus, Marek’s disease
virus, Newcastle disease virus. The vaccine does not stimulate B. Specific antiserum against the strain or each of the strains
the formation of antibodies against these agents. of the avian infectious bronchitis virus used in the vaccine
are neutralised by the vaccine.
Potency. Inject one dose by the route stated on the label into
each of twenty chickens, 3 to 4 weeks old, complying with the Tests
requirements stated under Test on chickens flocks free from
pathogens for the production and quality control of vaccines Water (2.3.43). Not more than 3.0 per cent.

1578
IP 2007 BLACKQUARTER VACCINE

Mycoplasmas (2.7.9). Complies with the test for mycoplasmas. Avian Spirochaetosis Vaccine
Safety. Inject 10 times the dose by the route stated on the Avian Spirochaetosis Vaccine is a suspension prepared from
label into each of 10 healthy chickens free from specific viscera and membranes of developing chicken embryos of
antibodies (2.7.7) to 10 days old. Use five healthy chickens SPF eggs infected with antigenic strains of Borrelia anserina
from the same source and stock as controls. Observe the birds which has been inactivated in such a manner that its
for 21 days. Not more than one of the vaccinated chickens immunogenic activity is retained.
shows symptoms of or dies from infectious bronchitis. The
test is not valid unless all the control chickens are free of Production
clinical symptoms of infectious bronchitis. If during the period
of observation more than 2 of the vaccinated chickens die The organism is grown in embryonated eggs derived from
from causes not attributable to the vaccine, repeat the test. SPF flocks (2.7.7).

Sterility (2.2.11). Complies with the test for sterility. Identification


Virus titre. Titrate the vaccine in cell cultures derived from Protects chickens against infection with B. anserina.
SPF embryos or by inoculating into the allantoic sac of SPF
embryonated eggs, 9 to 11 days old. One dose of the vaccine Tests
contains not less than 103.5 TCID50/EID50. Safety. Complies with the requiremented stated under
Immunogenicity. Carry out a test for each route of Veterinary Vaccine.
administration recommended on the label and for each Sterility (2.2.11). Complies with the test for sterility.
serotype against which protection is claimed. Administer to
each of 20 healthy chickens, free from specific antibodies (2.7.7) Potency. Inject at least twelve healthy chicken, free from
3 to 4 weeks old, for each of the stated routes a volume of antibodies, 8 to 12 weeks old, with the minimum dose of the
reconstituted vaccine containing a quantity of virus equivalent vaccine by the route stated on the label. Use five chickens of
to the minimum titre stated on the label. Ten additional healthy the same stock as controls. Ten days later challenge all the
chickens of same flock for each serotype against which chickens intraperitoneally with an adequate dose of a virulent
protection is claimed are used as unvaccinated controls. Three culture of B. anserina used to prepare the vaccine or with a
to four weeks later, administer by eye drop a virulent strain of suspension of liver or kidney tissue obtained from infected
bronchitis virus with a titre of at least 103.5 EID50 per ml to all chicken. Observe the chickens for 10 days. The vaccinated
the vaccinated and control birds. Between the fourth and chickens do not show any symptoms of the disease. The test
seventh day after the challenge, take tracheal swabs from is not valid unless the control chickens show typical symptoms
each of the vaccinated and control birds. Place each swab in a of spirochaetosis.
sterile test tube containing 3 ml of tryptose phosphate broth If the potency test has been performed with satisfactory
and antibiotics. Swirl thoroughly the tubes and swabs and results on representative batch of the vaccine from the same
store at 70° pending inoculation into eggs. For each chicken seed lot, it may be omitted as a routine control test during
swab, inoculate at least 5 chicken embryos, 9 to 11 days old, production of other batches of the vaccine prepared from the
with 0.2 ml of the broth from each tube into the allantoic cavity. same seed lot.
All the embryos surviving on the third day after inoculation
Labelling. The label states (1) strain of virus used; (2)
are used in the evaluation. A tracheal swab is considered
recommended age for vaccination.
positive for recovery of the virus if any of the embryos shows
typical infectious bronchitis lesions such as stunting, curling,
kidney urates, clubbing down or death between the fourth
and seventh day after inoculation. The vaccine complies with
Blackquarter Vaccine
the test if not less than 80 per cent of the controls and not Blackquarter Vaccine is a culture of suitable strain or strains
more than 20 per cent of the vaccinated chickens are positive of Clostridium chauvoei grown in a suitable anaerobic fluid
for virus recovery. If less than 80 per cent of the vaccinated medium and rendered sterile and non-toxic by addition of
chickens are negative for virus recovery the stock seed is solution of formaldehyde in such a manner that it retains its
unsatisfactory. The stock seed virus may be tested for immunizing properties. The vaccine may contain a suitable
immunogenicity once in 5 years provided it is maintained under adjuvant.
standard conditions of storage of the bronchitis virus.
Identification
Labelling. The label states (1) the minimum virus titre per
dose; (2) the recommended age of the birds in which the Protects susceptible animals against infection with
vaccine is to be used. Cl. chauvoei.

1579
CANINE CONTAGIOUS HEPATITIS VACCINE, INACTIVATED IP 2007

Tests Identification
Safety. Use two healthy susceptible animals of one of the When injected into a susceptible dog, the animal develops
species for which the vaccine is intended. Inject into each specific neutralizing antibodies.
animal by the recommended route twice the maximum dose
stated on the label. Observe the animals for 7 days. No Tests
significant local or systemic reaction is produced.
Water (2.3.43). Not more than 3.0 per cent (for freeze dried
Sterility (2.2.11). Complies with the test for sterility. vaccine only).
Potency. Inject ten healthy adult guinea pigs weighing between Safety. Inject each of two healthy dogs, between 8 and 14
350 and 450 g with a quantity of the product not greater than weeks old, that have been previously tested and shown to be
the minimum dose and route as stated on the label. Repeat the free from canine adenovirus neutralizing antibodies, with twice
inoculation with the same dose of the vaccine after 28 days. the dose and by the route stated on the label. Observe the
None of the vaccinated guinea pigs shows any systemic animals for 14 days. No abnormal systemic or local reaction
reaction. However a minimal local reaction may be observed occurs.
in the animal.
Sterility (2.2.11). Complies with the test for sterility.
Fourteen days after the second vaccination, inoculate
intramuscularly into each of 10 vaccinated and into each of Potency. Inject each of two healthy susceptible dogs, between
5 control guinea pigs with a suitable quantity of virulent culture 8 and 14 weeks old, that have been previously tested and
or spore suspension of Cl. Chauvoei, activated if necessary, shown to be free from canine adenovirus neutralizing
with the solution of calcium chloride. antibodies, with the minimum dose and by the route stated on
the label. After14 days inject a second dose. Between 14 and
The vaccine complies with the test, if not more than
21 days after the second injection collect serum from each
10 per cent of the vaccinated guinea pigs die from Cl. chauvoei
dog separately and examine each sample as described below.
infection within 5 days and all the control animals die from Cl.
chauvoei infection within 48 hours of challenge if virulent Inactivate the serum by heating at 56° for 30 min and prepare
culture was used or within 72 hours if a spore suspension was serial dilutions in a suitable medium. Add to each dilution an
used for the challenge. Repeat the test if 20 per cent of the equal volume of serum-virus suspension containing
vaccinated animals die. approximately 102 TCID50. Incubate the mixtures for 1 hour at
37°. Add suitable cell culture with minimum of four replicates
On repetition, the vaccine complies with the test if not more
for each dilution and incubate at 37° for 4 to 8 days. Examine
than 10 per cent of the vaccinated animals die within 5 days
each culture for evidence of specific cytopathic effect.
and all of the control animals die within 48 hours of challenge
Calculate the antibody titre. The serum from each dog contains
if virulent culture was used or within 72 hours if a spore sus-
not less than 80 SN50 per ml.
pension was used for challenge.
Labelling. The label states (1) the strain used for the
Labelling. The label states (a) the method of preparation; (b) preparation; (2) the name of any added adjuvant.
the strains of bacteria used to prepare the vaccine.

Canine Contagious Hepatitis Vaccine, Canine Contagious Hepatitis Vaccine,


Inactivated Live
Canine Contagious Hepatitis Vaccine, Inactivated is a Canine Contagious Hepatitis/Canine Adenovirus Vaccine is a
preparation containing canine contagious hepatitis virus freeze dried preparation containing one or more attenuated
inactivated in such a manner that its immunogenic activity is strains of canine adenovirus.
retained. It may be a freeze-dried preparation or a liquid
preparation containing a suitable adjuvant. Production

Production The virus is propagated in a suitable cell cultures, harvested,


titrated and may be mixed with a suitable stabilizing solution.
The virus is propagated in suitable cell culture systems, the The stock seed virus should be tested for immunogenicity at
viral suspension is harvested, titrated and may contain a least once in 5 years, if maintained under suitable conditions
suitable stabilizing solution. of storage.

1580
IP 2007 CANINE CORONA VIRUS VACCINE, INACTIVATED

Identification liquid or as a freeze-dried preparation to be reconstituted with


a suitable liquid immediately before use. The liquid vaccine
The vaccine, mixed with one or more specific antisera of canine may contain a suitable adjuvant.
adenovirus(s), does not produce specific cytopathic effects
in susceptible cell cultures. Production
Tests The virus is grown in suitable cell cultures systems. The
Water (2.3.43). Not more than 3.0 per cent. vaccine may contain a suitable adjuvant. Only an inactivated
viral suspension that complies with the requirements
Virus titre. Not less than103 TCID 50 virus per dose, mentioned under final bulk vaccine of each batch is used in
determining the titre of the vaccine in a suitable cell culture. the preparation of the final vaccine.
Safety. Inject each of two susceptible dogs, between 8 and 14
weeks old, that have been previously tested and shown to be Identification
free from canine adenovirus neutralising antibodies, with twice
the dose and by the route stated on the label. Observe the When inoculated into dogs, the vaccine stimulates the
animals for 21 days. No abnormal systemic or local reaction production of specific neutralizing antibodies against canine
occurs. corona virus as determined by suitable serological tests.

Sterility (2.2.11). Complies with the test for sterility. Tests


Potency. Inject five healthy susceptible dogs, between 8 and
14 weeks old, that have been previously tested and shown to Water (2.3.43). Not more than 3.0 per cent (for freeze dried
be free from canine adenovirus neutralizing antibodies, with a vaccine only).
quantity of the vaccine equivalent to the minimum titre and by Safety. Inject each of two healthy susceptible dogs, between
the route stated on the label. Observe the animals for 21 days. 8 and 14 weeks old, free from canine corona virus antibodies
Inject intravenously each of the five vaccinated animals and with a quantity equivalent to 2 doses by the route stated on
each of two control animals of the same stock and weight the label. Observe the animals for 14 days. No abnormal
range with a quantity of a virulent strain of canine contagious systemic or local reaction occurs.
hepatitis virus sufficient to cause death or typical signs of the
disease in a susceptible dog. Observe the animals for a further Sterility (2.2.11). Complies with the test for sterility.
period of 21 days. The vaccinated animals remain in normal
Potency. Either of the test A or B may be carried out.
health and the controls die from hepatitis or show typical
signs of serious infection. If one of the controls does not A. Inject each of five healthy susceptible guinea pigs, each
show any signs of the disease, repeat the test. The vaccinated weighing between 350 and 400 g, with half the minimum dose
animals of the second group remain in normal health and the and by the route stated on the label. Repeat the injection after
control animals die from hepatitis or show typical signs of 14 days. Fourteen days after the second injection collect blood
serious infection. samples and obtain the serum from each guinea pig separately.
If the potency test has been performed with satisfactory Inactivate each serum by heating at 56° for 30 min. Examine
results on a representative batch of the vaccine from the seed the serum samples for antibodies by the following method.
lot, it may be omitted as a routine control test during production Prepare 2-fold serial dilutions of serum in a medium suitable
on other batches of the vaccine prepared from the same seed for canine cells. Add to each dilution an equal volume of a
lot. virus suspension containing approximately 102 TCID50 and
Labelling. The label states (1) the minimum virus titre; (2) the incubate the mixtures at 37° for 1 hour. Inoculate a suitable
species of the animals in which use of the vaccine is volume of canine cells into at least 4 replicates of serum virus
recommended. mixtures and incubate at 37° for 4 days. Examine for evidence
of specific cytopathic effects and calculate the antibody titre.
The vaccine complies with the test if the mean antibody titre
is not less than 32 SN50 per 0.05 ml of serum.
Canine Corona Virus Vaccine,
B. Inject each of two healthy susceptible dogs, between 8 and
Inactivated 14 weeks old, having antibody titre less than 4 SN50 per 0.05
Canine Corona Virus Vaccine, Inactivated is a preparation ml of serum, with the dose and by the route recommended on
containing canine corona virus inactivated in such a manner the label. Fourteen days later collect the serum samples from
that its immunogenic activity is retained. It may be issued as a each dog separately. Inactivate each serum by heating at 56°

1581
CANINE DISTEMPER VACCINE, LIVE IP 2007

for 30 min. Examine the serum samples individually for tenths of the dose of the vaccine is injected. Inject each mouse
neutralizing antibodies by the method described in test A. intracerebrally with 0.03 m1 of the vaccine. Observe for 21
If one dog fails to respond, i.e., the antibody titre is less than days. Not more than two mice die within the first 48 hours. If
4 SN50 per 0.05 ml of serum, repeat the test with two more dogs more than two mice die within the first 48 hours, repeat the
and calculate the mean titres of the three dogs that have test. From the third day to 21 days after the injection, the mice
responded. The vaccine complies with the test if the median do not show any abnormalities attributable to the vaccine.
antibody titre of the sera is not less than 32 SN50 per 0.05 ml. Mycoplasmas (2.7.8). Complies with the test for freedom from
Labelling. The label states (1) the recommended routes of mycoplasmas.
administration; (2) that the preparation should be shaken well Safety. Reconstitute the vaccine as recommended on the label
before use; (3) that the liquid preparation should not be allowed and carry out the following tests.
to freeze; (4) that the vaccine should be used immediately
after reconstitution. A. For chicken embryo-adapted vaccine only. Inject 0.3 m1
intracerebrally into each of a group of eight mice, between 3
and 4 weeks old, and 0.5 m1 intraperitoneally into each of
another eight mice of the same age group.
Canine Distemper Vaccine, Live
Observe both the groups for 7 days. Not more than one mouse
Canine Distemper Vaccine, Live is a freeze-dried preparation in either group shows any abnormal local or systemic reaction
of a strain of canine distemper virus that has been attenuated attributable to the vaccine.
for dogs and is grown either in SPF embryonated eggs or in
suitable cell cultures. B. Inject each of two susceptible dogs, between 8 and 14
weeks old which do not have antibodies against canine
Production distemper virus with twice the dose and by the route stated
on the label. Observe the animals for 21 days. None of the
The virus is propagated in suitable cell culture systems, the
dogs shows any abnormal local or systemic reaction.
viral suspension is harvested, titrated and may contain a
suitable stabilizing solution. The stock seed virus should be Sterility (2.2.11). Complies with the test for sterility.
tested for potency at least once in 5 years, if maintained
Potency. Inject each of five susceptible dogs, between 8 and
under suitable conditions of storage.
14 weeks old, that have been previously tested and shown to
Identification be free from distemper virus neutralizing antibodies with a
volume of the reconstituted vaccine containing a quantity of
A. The vaccine infects the chorio-allantoic membranes of SPF the virus equivalent to the minimum titre and by the route
embryonated eggs or suitable cell cultures. This effect is stated on the label. Use another two dogs of the same stock
neutralized by canine distemper antiserum. and age group as unvaccinated controls. Observe the animals
B. An injection into susceptible ferrets or dogs does not cause for 21 days. Inject intravenously each of the seven animals
distemper but immunizes them against the disease. The with a quantity of virulent strain of canine distemper virus
vaccine strain is satisfactory with respect to absence of sufficient to cause death or produce typical signs of the disease
increase in virulence. in a susceptible dog. Observe the animals for a further 21
days. The vaccinated animals survive and show no clinical
Tests signs of canine distemper. The test is not valid unless the
Water (2.3.43). Not more than 3.0 per cent. control dogs die or show symptoms typical of canine distemper.
If one of the control animals does not show any sign of
Virus titre. Not less than 103 EID50/TCID50 of the virus per distemper, repeat the test. The vaccinated animals of the second
dose, determining the titre of the vaccine in a suitable cell group remain in normal health and the control animals die
culture or SPF eggs (2.7.7). from distemper or show symptoms typical of canine distemper.
Extraneous pathogens If the potency test has been performed with satisfactory
A. Mix the vaccine under examination with a mono specific results on a represntative batch of the vaccine from the seed
distemper antiserum. It no longer provokes cytopathic effects lot, it may be omitted as a routine control test during production
in susceptible cell cultures and shows no evidence of on other batches of the vaccine prepared from the same seed
haemagglutinating or haemadsorbing agents. lot.
B. Use a sufficient number of mice, not less than ten, each Labelling. The label states (1) the strain used for the
weighing between 11 and 15 g, such that a total of three- preparation; (2) virus titre per dose.

1582
IP 2007 CANINE PARVOVIRUS VACCINE, INACTIVATED

Canine Parainfluenza Virus Vaccine, If the potency test has been performed with satisfactory
results on a representative batch of the vaccine it may be
Live omitted as a routine test during production on the other
Canine Parainfluenza Virus Vaccine, Live is a freeze-dried batches of vaccine prepared from the same seed lot.
preparation of tissue culture fluid containing the cell culture- Labelling. The label states (1) the strain used for the
adapted attenuated canine parainfluenza virus of stock seed preparation; (2) the virus titre per dose.
which has been established as pure, safe and immunogenic.

Production
The virus is propagated in suitable cell culture systems, the Canine Parvovirus Vaccine,
viral suspension is harvested, titrated and may contain a Inactivated
suitable stabilizing solution.
Canine Parvovirus Vaccine, Inactivated is a liquid or freeze
Identification dried preparation of canine parvovirus inactivated by a suitable
method so that its immunogenic activity is retained.
When inoculated into dogs, the vaccine stimulates the
production of specific neutralizing antibodies against canine Production
parainfluenza virus as determined by suitable serological tests.
The virus is grown in suitable cell culture systems. The virus
Tests may be cloned, purified and concentrated. The vaccine may
Water (2.3.43). Not more than 3.0 per cent. contain a suitable adjuvant.

Virus titre. Not less than 103 TCID50 per dose, determining Identification
the titre of the vaccine in a suitable cell culture.
When inoculated into dogs, the development of specific
Safety. Inject each of two susceptible dogs, between 8 and 14 neutralizing antibodies against canine parvovirus can be
weeks old, free from canine parainfluenza virus demonstrated by suitable serological tests.
haemaggulatinating antibodies with a dose of the vaccine
reconstituted with the sterile diluent equivalent to 10 times Tests
the dose and by the route stated on the label. Observe the
animals for 14 days. None of the dogs shows any systemic or Safety. Inject into each of two healthy susceptible dogs,
local reactions. between 8 and 14 weeks old, preferably sero negative ones
with a quantity equivalent to 2 doses by the route stated on
Sterility (2.2.11). Complies with the test for sterility. the label. Observe the animals for 14 days. No abnormal
Potency. Use two healthy susceptible dogs, between 8 and 14 systemic or local reaction occurs.
weeks old, having haemagglutination inhibition antibody titres Sterility (2.2.11). Complies with the test for sterility.
less than 1:4 per 0.1 ml of serum. Keep equal number of dogs
as unvaccinated controls. Vaccinate each test group dog with Potency. Either of the test A or B may be carried out.
the vaccine as per schedule stated on the label. After 21 days, A. Inject subcutaneously each of five healthy susceptible
collect the serum from each dog separately and examine each guinea-pigs, each weighing between 350 and 400g, with half
sample as described below. Heat the serum of each animal at of the dose stated on the label. After 14 days, inject again half
56° for 30 minutes and prepare serial dilutions with saline of the dose stated on the label. Fourteen days after the second
solution. To 0.025 ml of each dilution add 0.025 ml of a canine injection collect blood samples and obtain the serum from
parainfluenza virus suspension containing 4 haemagglutinating each guinea pig separately. Inactivate each serum by heating
units. Allow the mixture to stand at room temperature for 30 at 56 ° for 30 min. To 1 volume of each serum add 9 volumes of
minutes and add 0.05 ml of a suspension of chicken a 20 per cent suspension of light kaolin in phosphate buffered
erythrocytes containing 2 x 107 erythrocytes per ml. Allow to saline pH 7.4. Shake the mixture for 20 minutes and centrifuge
stand for 1 hour and note the last dilution of serum that at 2,000 rpm for 10 minutes. Collect the supernatant liquid and
completely inhibits haemugglutination. Calculate the median mix with 1 volume of a concentrated suspension of Rhesus
antibody litre of the sera which should not be less than 1:32 monkey/pig erythrocytes. Allow the mixture to stand at 4° for
per 0.1 ml of the serum. If one dog fails to respond, repeat the 60 minutes and centrifuge at 2,000 rpm for 10 minutes and
test using two more dogs and calculate the result as the mean collect the supernatant serum obtained in 10-fold dilution.
of titres obtained from all of three dogs that have responded Using each serum, prepare a series of 2-fold dilutions. To
which is not less than 1 :32 per 0.1 ml of the serum. 0.025 ml of each of the latter dilutions add 0.025 ml of a

1583
CANINE PARVOVIRUS VACCINE, LIVE IP 2007

suspension of canine parvovirus antigen containing Virus titre. Not less than 103 TCID50 per dose, determining
approximately 8 haemagglutinating (HA) units and allow to the titre of the vaccine in a suitable cell culture.
stand at 37° for 30 minutes. Add 0.05 ml or other suitable
Safety. Inject each of two susceptible dogs, between 8 and 14
quantity of a 1 per cent suspension of Rhesus monkey/pig
weeks old, free from canine parvovirus haemaggulatinating
erythrocytes containing 30 x 106 cells per ml to at least four
antibodies, a quantity of the vaccine reconstituted with the
replicates of each dilution. Allow to stand at 4° for 90 minutes
sterile diluent equivalent to 10 times the dose and by the route
and note the last dilution of the serum that completely inhibits
stated on the label. Observe the animals for 21 days. No
haemagglutination. The vaccine complies with the test if the
abnormal systemic or local reaction occurs.
median antibody titre of the sera is not less than 1:80.
Sterility (2.2.11). Complies with the test for sterility.
B. Inject each of two healthy susceptible dogs, between 8 and
14 weeks old, having antibody titres less than 4 ND50 (50 Potency. Inject each of seven dogs, between 8 and 14 weeks
percent neutralizing dose) per 0.1 ml of serum, with the dose old, free from canine parvovirus haemagglutinating antibodies,
and by the route recommended on the label. Fourteen days subcutaneously with the minimum dose stated on the label.
later collect the blood/serum samples from each dog separately. Use two dogs of the same stock, weight and age range as
Inactivate each serum by heating at 56° for 30 min. Examine controls. Not less than 21 days after injection of the vaccine,
the serum samples individually for neutralizing antibodies as challenge the vaccinated and control animals through the
follows: oronasal route with a virulent strain of infectious canine
parvovirus. Observe the animals for 14 days. Not less than
Prepare 2-fold serial dilutions of serum in a medium suitable
five of the vaccinated dogs survive. The test is not valid
for canine cells. Add to each dilution an equal volume of virus
unless the control dogs die or show clinical signs of canine
suspension containing approximately 104 TCID50 and incubate
parvovirus infection.
the mixtures at 37° for one hour. Inoculate a suitable volume of
canine cells into at least four replicates of serum virus mixtures If the potency test has been performed with satisfactory
and incubate at 37° for 7 days. Examine for evidence of specific results on a representative batch of the vaccine it may be
cytopathic effects and calculate the antibody titre. The vaccine omitted as a routine test during production of the other batches
complies with the test if the mean antibody titre is not less of vaccine prepared from the same seed lot.
than 32 ND50 per 0.1 ml of serum. If one dog fails to respond Labelling. The label states (1) the strain used for the
repeat the test using two more dogs and calculate the mean preparation; (2) virus titre per dose.
titres of the three dogs that have responded.
Labelling. The label states (1) the method of preparation; (2)
the types and strains of virus used to prepare the vaccine.
Clostridium Multicomponent Vaccine,
Inactivated
Canine Parvovirus Vaccine, Live
Clostridium Multicomponent Vaccine, Inactivated consists of
Canine Parvovirus Vaccine, Live is a freeze-dried preparation five highly antigenic components containing the toxoids of
of a strain of canine parvovirus that is attenuated for the C. perfringens type ‘B’, C. perfringens type ‘C’, C. perfringens
target species of dogs. type ‘D’, C. oedematiens and C. septicum which are prepared
Production in double strength and then combined in such a proportion
that would invoke adequate anti-toxin response in the
The attenuated virus is grown in suitable cell culture systems. vaccinated sheep against each antigen incorporated in the
The viral harvest is titrated and mixed with a suitable stabilizing vaccine.
solution. The stock seed virus should be tested for
immunogenicity at least once in 5 years, if maintained under Identification
suitable conditions of storage.
When injected into susceptible animals, it stimulates the
Identification production of epsilon and beta antitoxin against C. perfringens
When inoculated into dogs, the development of specific types ‘B’, ‘C’ and ‘D’ and also antitoxins against C. septicum
neutralizing antibodies against canine parvovirus can be and toxin of C. oedematiens.
demonstrated by suitable serological tests.
Tests
Tests Safety. Four sheep each are inoculated with two times the
Water (2.3.43). Not more than 3.0 per cent. dose of vaccine subcutaneously and are observed for 7 days

1584
IP 2007 CLOSTRIDIUM NOVYI (TYPE B) VACCINE FOR VETERINARY USE

during which period the animals do not show any local or Tests
systemic reaction.
Sterility (2.2.11). Complies with the test for sterility.
Sterility (2.2.11). Complies with the test for sterility.
Potency. Inject subcutaneously into each of not less than
Potency. Eight sheep each are inoculated with 2 doses of 10 healthy rabbits, 3 to 6 months old, a quantity of vaccine
vaccine subcutaneously at an interval of 21 to 28 days and not exceeding the minimum dose stated on the label as the
bled on 10th day after second inoculation for collection of first dose. After 21 to 28 days, inject into the same animals a
serum for assessing the antitoxin titre against each antigen quantity of the vaccine not exceeding the minimum dose stated
incorporated in the vaccine. The post-inoculation serum on the label as the second dose. 10 to 14 days after the second
contains not less than 2 IU of epsilon antitoxin and 10 units of injection, bleed the rabbits and pool the sera.
beta antitoxins of C. perfringens types ‘B’ and ‘C’ and 2.5 IU
of C. septicum antitoxin and 3.5 IU of C. oedematiens antitoxin. The alpha antitoxin titre of the pooled sera is not less than
3.5 IU per ml.
Labelling. The label states (1) the types of Clostridia contained
in the vaccine; (2) the preparation should be shaken before The International Unit is the specific neutralising activity for
use. C. novyi alpha toxin contained in a stated amount of the
International standard, which consists of a quantity of dried
immune horse serum. The equivalence in International Units
Clostridium novyi (Type B) Vaccine for of the International standard is stated by the World Health
Organisation.
Veterinary Use
The potency of the pooled sera obtained from the rabbits is
Clostridium novyi (Type B) Vaccine for Veterinary Use is determined by comparing the quantity necessary to protect
prepared from a liquid culture of a suitable strain of mice or other suitable animals against the toxic effects of a
Clostridium novyi type B. fixed dose of C. novyi alpha toxin with the quantity of a
reference preparation of Clostridium novyi alpha antitoxin,
Production
calibrated in International Units, necessary to give the same
The whole culture or its filtrate or a mixture of the two is protection. For this comparison, a suitable preparation of
inactivated in such a manner that toxicity is eliminated and C. novyi alpha toxin for use as a test toxin is required. The
immunogenic activity is retained. Toxoids and/or inactivated dose of the test toxin is determined in relation to the reference
cultures may be treated with a suitable adjuvant, after preparation; the potency of the serum under examination is
concentration if necessary. determined in relation to the reference preparation using the
test toxin.
Choice of vaccine composition. The vaccine is shown to be
satisfactory with respect to safety and efficacy (2.7.12). For Preparation of test toxin. Prepare the test toxin from a sterile
the latter, it shall be demonstrated that for each target species filtrate of an approximately 3 to 5 day culture in liquid medium
the vaccine, when administered according to the recommended of C. novyi type B and dry by a suitable method. Select the
schedule, stimulates an immune response (for example, test dose of the toxin in mice by determining the L+ 10 dose
induction of antibodies) consistent with the claims made for and the LD50 the observation period being 72 hours.
the product. A suitable alpha toxin contains not less than one L+/10 dose
Batch testing in 0.05 mg and not less than 10 LD50 in each L+/10 dose.
Determination of test dose of toxin. Prepare a solution of the
Safety. Administer by a recommended route, to each of 2 sheep
reference preparation in a suitable liquid so that it contains
that have not been vaccinated against C. novyi type B twice
1 IU per ml. Prepare a solution of the test toxin in a suitable
the maximum dose of the vaccine stated on the label. Observe
liquid so that 1 ml contains a precisely known amount such as
the animals for not less than 14 days. No abnormal local or
1 mg. Prepare mixtures of the solution of the reference
systemic reaction occurs.
preparation and the solution of the test toxin such that each
Residual toxicity. Inject 0.5 ml of the vaccine subcutaneously mixture contains 1.0 ml of the solution of the reference
into each of 5 mice, each weighing between 17 and 22 g. preparation (1 IU), one of a series of graded volumes of the
Observe the animals for 7 days. No abnormal local or systemic solution of the test toxin and sufficient of a suitable liquid to
reaction occurs. bring the total volume to 2.0 ml. Allow the mixtures to stand at
room temperature for 60 min. Using not less than 2 mice, each
Identification
weighing between 17 and 22 g, for each mixture, inject a dose
The vaccine stimulates the formation of C. novyi type ‘B’ of 0.2 ml subcutaneously into each mouse. Observe the mice
alpha antitoxin when injected into animals. for 72 hours. If all the mice die, the amount of toxin present in

1585
CLOSTRIDIUM PERFRINGENS VACCINE, INACTIVATED IP 2007

0.2 ml of the mixture is in excess of the test dose. If none of the valid estimates. The test is valid only if the reference
mice dies, the amount of toxin present in 0.2 ml of the mixture preparation gives a result within 20 per cent of the expected
is less than the test dose. Prepare fresh mixtures such that value.
2.0 ml of each mixture contains 1.0 ml of the solution of the
The confidence limits (P = 0.95) have been estimated to be (a)
reference preparation (1 IU) and one of a series of graded
85 per cent and 114 per cent when 2 animals per dose are used;
volumes of the solution of the test toxin separated from each
other by steps of not more than 20 per cent and covering the (b) 91.5 per cent and 109 per cent when 4 animals per dose are
expected end-point. Allow the mixtures to stand at room used; (c) 93 per cent and 108 per cent when 6 animals per
temperature for 60 min. Using not less than two mice for each dose are used.
mixture, inject a dose of 0.2 ml subcutaneously into each mouse. Labelling. The label states (1) whether the product is a toxoid,
Observe the mice for 72 hours. Repeat the determination at a vaccine prepared from a whole inactivated culture or a
least once and combine the results of the separate tests that mixture of the two; (2) that the preparation is to be shaken
have been made with mixtures of the same composition so before use; (3) for each target species, the immunising effect
that a series of totals is obtained, each total representing the produced (for example, antibody production, protection
mortality due to a mixture of a given composition. against signs of disease or infection).
The test dose of toxin is the amount present in 0.2 ml of that
mixture which causes the death of one half of the total number
of mice injected with it.
Clostridium Perfringens Vaccine,
Determination of the potency of the serum obtained from Inactivated
rabbits
The vaccines contains cultures strains of C. perfringens type
Preliminary test. Dissolve a quantity of the test toxin in a B (Lamb Dysentery Vaccine), type C (Struck Vaccine) or type
suitable liquid so that 1 ml contains 10 times the test dose D (Enterotoxaemia Vaccine; Pulpy Kidney Vaccine) or any
(solution of the test toxin). Prepare a series of mixtures of the combination of these types.
solution of the test toxin and of the serum under examination
such that each mixture contains 1.0 ml of the solution of the Production
test toxin, one of a series of graded volumes of the serum
under examination and sufficient of a suitable liquid to bring The organisms grown in an anaerobic medium, the whole
the final volume to 2.0 ml. Allow the mixtures to stand at room culture or their filtrates or a mixture of the two are inactivated
temperature for 60 min. Using not less than 2 mice for each in such a manner that toxicity is eliminated and immunogenic
mixture, inject a dose of 0.2 ml subcutaneously into each mouse. activity is retained. Toxoid and or inactivated cultures may
Observe the mice for 72 h. If none of the mice dies, 0.2 ml of contain a suitable adjuvant.
the mixture contains more than 0.1 IU. If all the mice die, 0.2 ml
Batch tesing
of the mixture contains less than 0.1 IU.
Safety. Inject subcutaneously two healthy susceptible sheep
Final test. Prepare a series of mixtures of the solution of the
weighing about 18 kg each or two healthy susceptible rabbits
test toxin and of the serum under examination such that 2.0 ml
weighing between 1.5 and 2.0 kg each with twice the dose
of each mixture contains 1.0 ml of the solution of the test toxin
stated on the label and observe for 7 days. No systemic or
and one of a series of graded volumes of the serum under
local reaction is observed.
examination, separated from each other by steps of not more
than 20 per cent and covering the expected end-point as Observe the animals for 14 days. No abnormal local or systemic
determined by the preliminary test. Prepare further mixtures of reaction occurs.
the solution of the test toxin and of the solution of the reference Residual toxicity. Inject 0.5 ml of the vaccine subcutaneously
preparation such that 2.0 ml of each mixture contains 1.0 ml of into each of 5 mice, each weighing 17 to 22 g. Observe the
the solution of the test toxin and one of a series of graded animals for 7 days. No abnormal local or systemic reaction
volumes of the solution of the reference preparation, in order occurs.
to confirm the test dose of the toxin. Allow the mixtures to
stand at room temperature for 60 min. Using not less than 2 Sterility (2.2.11). Complies with the test for sterility.
mice for each mixture, proceed as described in the preliminary
Potency
test. The test mixture which contains 0.1 IU in 0.2 ml is that
mixture which kills the same or almost the same number of C. perfringens Type B Vaccine. Inject subcutaneously into
mice as the reference mixture containing 0.1 IU in 0.2 ml. Repeat each of six healthy susceptible sheep weighing about 18 kg or
the determination at least once and calculate the average of all ten healthy susceptible rabbits weighing between 1.5 and 2.0

1586
IP 2007 CLOSTRIDIUM PERFRINGENS VACCINE, INACTIVATED

kg with the minimum dose of the vaccine stated on the label. The test dose of each toxin is established in relation to the
Repeat the dose after an interval of 21 to 28 days. Bleed the appropriate Standard preparation of antitoxin and the potency
animals 10 to 14 days after the second dose of the vaccine and of antitoxin under examination is then determined in relation
determine beta antitoxin titres in the pooled serum sample by to the appropriate Standard preparation using the appropriate
testing in mice as per the method described for C. perfringens test toxin.
Type D Vaccine. Product passes the test if the post-inoculation
International standard for the standard preparations
pooled serum contains not less than 10 IU of beta antitoxins,
and not less than 5 IU of episilon toxin per milliliter. The International units of the antitoxin is the specific
neutralizing activity of C. perfringens epsilon toxin contained
C. perfringens Type C Vaccine. Carry out the test for potency
in the stated amount in relation to International standards in
as described for C. perfringens Type B Vaccine.
the dried Horse serum.
1 ml of serum contains not less than 10 IU of beta antitoxin per
ml. The International units of the antitoxins is the specific
neutralizing activity for the C. perfringens beta toxin contained
C. perfringens Type D (Enterotoxaemia) Vaccine. Carry out in the stated amount in relation to International standards in
the test as described below. the dried Horse serum.
1 ml of serum contains not less than 5 IU of C. perfringens
epsilon toxin per ml. Test animals
Use healthy mice having body weights such that the
Identification
difference between the lightest and heaviest is not more than
A. When injected into susceptible animals, the C. perfringens 5 g.
Type B Vaccine stimulates the production of C. perfringens Suggested method for preparation of test toxin. Prepare C.
beta and epsilon antitoxins. perfringens toxins from sterile supernatants/filtrates of early
B. When injected into susceptible animals, the C. perfringens cultures of C. perfringens type B, type C or type D. The
Type C Vaccine stimulates the production of C. perfringens supernatants may be purified by precipitation with ammonium
beta antitoxin. sulphate and the resulting precipitate collected. This may then
be dried over phosphorus pentoxide at a pressure of 1.5 to
C. When injected into susceptible animals, the C. perfringens
2.5 kPa, powdered and kept dry or re-dissolved and freeze-
Type D Vaccine stimulates the production of C. perfringens
dried.
epsilon antitoxin.
Selection of test toxin. Select toxin for use as the test toxin by
Tests determining the following quantities.
Potency test. Inject subcutaneously into each of at least six L+ and L+/10 doses. These are the smallest quantities of toxin
healthy susceptible sheep weighing about 18 kg or ten healthy that when mixed respectively with 1 Unit of antitoxin and with
susceptible rabbits weighing between 2.0 and 2.5 kg with the 0.1 Unit of antitoxin and injected intravenously into mice cause
minimum dose of the vaccine stated on the label. Repeat the the death of the animals within 72 hours.
dose in each sheep/rabbit after an interval of 21 to 28 days. LD50. This is the quantity of toxin that when injected
The rabbits are inoculated with the same dose after one month intravenously into mice causes the death of one-half of the
of the first inoculation. Bleed the animals 10 to 14 days after mice injected within 72 hours.
the second inoculation. The sera of sheep or rabbits are pooled
separately and estimated for the antitoxin levels. A suitable C. perfringens beta toxin is one that has an L+ dose
in 0.2 mg or less and contains not less than 25 LD50 in an L+
Biological assay of C. perfringens antitoxins dose.

The potency of C. perfringens beta and epsilon antitoxins is A suitable Clostridium perfringens epsilon toxin is one that
determined by comparing the dose of antitoxin necessary to has an L+/10 dose in 0.005 mg or less and contains not less
protect mice or other suitable animals against the toxic effects than 20 LD50 in an L+/10 dose.
of C. perfringens beta toxin or epsilon toxin with the dose of a Determination of test dose of C. perfringens beta toxin.
standard preparation of the respective antitoxin necessary to Dissolve a quantity of dried toxin in a suitable liquid such that
give the same protection. For this comparison, the Standard 1.0 ml contains a precise amount such as 10 mg. Reconstitute
preparations of C. perfringens beta antitoxin and the Standard preparation of C. perfringens beta antitoxin with
C. perfringens epsilon antitoxin and suitable preparations of a suitable liquid to give a solution containing 5 Units of C.
C. perfringens beta and epsilon toxins are required. perfringens beta antitoxin in 1 ml.

1587
CLOSTRIDIUM PERFRINGENS VACCINE, INACTIVATED IP 2007

Prepare mixtures such that 5.0 ml of each mixture contains 2.0 all the mice die, 0.5 ml of the mixture contains less than I Unit
ml of the solution of the Standard preparation (10 Units) and of antitoxin. If none of the mice dies 0.5 ml of the mixture
one of a series of graded volumes of the solution of the toxin. contains more than 1 Unit of antitoxin.
Dilute each mixture to the same final volume with a suitable
Final test. Prepare similar fresh mixtures such that 5.0 ml of
liquid. Allow the mixtures to stand at room temperature,
each mixture contains 2.0 ml of the solution of the toxin and
protected from light, for 30 minutes and then inject a dose of
one of a series of graded volumes of the preparation under
0.5 ml of each mixture intravenously into each of not less than
examination, separated from each other by steps of not more
two mice. Observe the mice for 72 hours. If all the mice die, the
than 20 per cent and covering the expected end-point. Prepare
amount of toxin present in 0.5 ml of the mixture is in excess of
further mixtures of 5.0 ml containing 2.0 ml of the solution of
the test dose. If none of the mice dies, the amount of toxin
the toxin and graded volumes of the Standard preparation of
present in 0.5 ml of the mixture is less than the test dose.
C. perfringens beta antitoxin to confirm the test dose of the
Prepare similar fresh mixtures such that 5.0 ml of each mixture
toxin. Allow the mixtures to stand at room temperature,
contains 2.0 ml of the solution of the Standard preparation (10
protected from light, for 30 minutes. Inject a dose of 0.5 ml of
Units) and one of a series of graded volumes of the solution
each mixture intravenously into each of not less than two
of the toxin, separated from each other by steps of not more
mice and observe the mice for 72 hours. The mixture of the
than 20 per cent and covering the expected end-point.
antitoxin under examination which contains 1 Unit of
Allow the mixtures to stand at room temperature, protected C. perfringens beta antitoxin in 0.5 ml is that mixture which
from light, for 30 minutes, Inject a dose of 0.5 ml of each mixture causes the death of the same or almost the same number of
intravenously into each of not less than two mice. Observe mice as the mixture containing 1 Unit of the Standard preparation
the mice for 72 hours. Repeat the determinations at least once of C. perfringens beta antitoxin in 0.5 ml. Repeat the
and add together the results of the separate tests that have determinations at least once and calculate the average of all
been made with mixtures of the same composition such that a valid estimates. Estimates are not valid unless the Standard
series of totals is obtained, each total representing the preparation gives a result within 20 per cent of the expected
mortality due to a mixture of a given composition. value.
The test dose of toxin is the amount present in 0.5 ml of that Determination of potency of C. perfringens epsilon antitoxin.
mixture that causes the death of one-half of the total number Carry out the preliminary test and final test as described for
of mice injected with it within 72 hours. the determination of potency of C. perfringens beta antitoxin
Determination of test dose of C. perfringens epsilon toxin. with the following amendments.
Carry out the method described for the determination of test Dilute a quantity of the test toxin in a suitable liquid such that
dose of C. perfringens beta toxin with the following 2.0 ml contains 10 times the test dose. The Standard preparation
modification. Dissolve a quantity of dried toxin in a suitable used in these tests is that of C. perfringens epsilon antitoxin.
liquid such that 1.0 ml contains a precise amount such as 1
mg. The mixture of the antitoxin under examination which contains
0.1 Unit of C. perfringens epsilon antitoxin in 0.5 ml is that
Reconstitute the Standard preparation of C. perfringens mixture which causes the death of the same or almost the
epsilon antitoxin with a suitable liquid to give a solution same number of mice as the mixture containing 0.1 Unit of the
containing 0.5 Unit in 1 ml (the prepared mixtures will therefore Standard preparation of C. perfringens epsilon antitoxin in 0.5
contain 1 Unit of the Standard preparation in 5 ml). ml.
The test dose of toxin is the amount present in 0.5 ml of that
Limits of error. For the suggested method, the limits of error
mixture that causes the death of one-half of the total number
(P = 0.95) have been estimated to be 85 to 114 per cent when
of mice injected with it within 72 hours.
two mice are used per dose, 91.5 to 109 per cent when four
Determination of potency of C. perfringens beta antitoxin mice are used per dose, and 93 to 108 per cent when six mice
are used per dose.
Preliminary test. Dilute the test toxin with a suitable liquid
such that 2.0 ml contains 10 times the test dose. Prepare Labelling. The label states (a) the type or types of
mixtures such that 5.0 ml of each mixture contains 2.0 ml of the C. perfringens from which the vaccine has been prepared;
solution of the toxin and one of a series of graded volumes of (b) whether the preparation is a toxoid or a vaccine prepared
the preparation under examination. Adjust each mixture to the from a whole inactivated culture or a mixture of the two;
same final volume with a suitable liquid. Allow the mixtures to (c) that the preparation is to be shaken before use; (d) for each
stand at room temperature, protected from light, for 30 minutes. target species, the immunising effect produced (for example,
Inject a dose of 0.5 ml of each mixture intravenously into each antibody production, protection against signs of disease or
of not less than two mice and observe the mice for 72 hours. If infection).

1588
IP 2007 CLOSTRIDIUM SEPTICUM VACCINE

Clostridium Septicum Vaccine serum (supplied in ampoules containing 500 Units) or another
suitable preparation the potency of which has been determined
Clostridium Septicum Vaccine is a suspension of a culture of a in relation to the International standard.
highly toxigenic strain of C. septicum grown in an anaerobic
medium, or a filtrate from such a culture. Safety. Inject subcutaneously each of two healthy susceptible
sheep, between 8 and 12 months old, with twice the dose
Production stated on the label. Observe the animals for 7 days; none of
the animals shows any systemic or local reaction. Observe
The whole culture or its filtrate or a mixture of the two is
the animals for 14 days.
inactivated in such a manner that toxicity is eliminated and
immunogenic activity is retained. Toxoid and/or inactivated Test animals. Use healthy mice having body weights such
cultures may be treated with a suitable adjuvant. that the difference between the lightest and heaviest is not
more than 5 g.
Batch testing
Preparation of test toxin. Prepare C. septicum toxin by growing
Residual toxicity. Inject 0.5 ml of the vaccine subcutaneously C. septicum in a liquid culture medium, filtering the supernatant
into each of 5 mice, each weighing between 17 and 22 g. aseptically and precipitating with ammonium sulphate. The
Observe the animals for 7 days. No abnormal local or systemic resulting precipitate, which contains the toxin, is collected,
reaction occurs. dried over phosphorus pentoxide at a pressure of 1.5 to 2.5
Sterility (2.2.11). Complies with the test for sterility. kPa, powdered and kept dry.
Potency. Inject subcutaneously each of eight healthy Selection of test toxin. Select toxin for use as the test toxin by
susceptible sheep, between 8 and 12 months old, or ten rabbits, determining the following quantities.
between 3 and 6 months old, with the minimum dose of the
L+/5 dose. This is the smallest quantity of the toxin which
vaccine stated on the label. Repeat the dose after an interval
when mixed with 0.2 Unit of antitoxin and injected intravenously
of 21 to 28 days. Ten days after the second inoculation, bleed
into mice causes the death of the animals within 72 hours.
the animals. Pool the sera samples from individual animals
and determine the antitoxin titre by the biological assay of LD50. This is the quantity of toxin which when injected
C. septicum antitoxin described below. intravenously into mice causes the death of one-half of the
1 ml of serum contains not less than 2.5 Units of C. septicum animals within 72 hours.
antitoxin by biological assay of C. septicum antitoxin. A suitable C. septicum toxin is one that has an L+/5 dose in 1
The potency of Clostridium septicum antitoxin is determined mg or less and contains not less than 10 LD50 in an L+/5 dose.
by comparing the dose of antitoxin necessary to protect mice Determination of test dose of toxin. Weigh accurately a
or other suitable animals against the lethal effects of quantity of the dried toxin and dissolve it in a suitable liquid
Clostridium septicum toxin with the dose of a Standard so that 1.0 ml contains a precise known amount, such as 4 mg.
preparation of Clostridium septicum antitoxin necessary to Prepare a solution of the Standard preparation in a suitable
give the same protection. For this purpose, the Standard liquid such that 1.0 ml contains 1 Unit.
preparation of C. septicum antitoxin and a suitable preparation
of C. septicum toxin for use as a test toxin are required. Prepare mixtures such that 5.0 ml of each mixture contains 2.0
ml of the solution of the Standard preparation (2 Units) and
Identification one of a series of graded volumes of the solution of the toxin.
Dilute each mixture with a suitable liquid to the same final
When injected into healthy susceptible animals, it stimulates
volume (5.0 ml). Allow the mixtures to stand at room
the production of antitoxins to C. septicum.
temperature, protected from light, for 60 minutes and then
Tests inject a dose of 0.5 ml of each mixture intravenously into each
of not less than 2 mice. Observe the mice for 72 hours. If all the
The test dose of the toxin is determined in relation to the
mice die the amount of toxin present in 0.5 ml of the mixture is
Standard preparation of antitoxin and the potency of antitoxin
in excess of the test dose. If none of the mice dies, the amount
under examination is then determined in relation to the Standard
of toxin present in 0.5 ml of the mixture is less than the test
preparation using the test toxin.
dose. Prepare similar fresh mixtures such that 5.0 ml of each
Assay mixture contains 2.0 ml of the solution of the Standard
preparation (2 Units) and one of a series of graded volumes of
Standard preparation
the solution of the toxin separated from each other by steps of
The Standard preparation is the 3rd International standard, not more than 20 per cent and covering the expected end-
established in 1957, consisting of dried hyperimmune horse point.

1589
DUCK PASTEURELLA VACCINE, INACTIVATED IP 2007

Allow the mixtures to stand at room temperature, protected The vaccine passes the test if the pooled serum contains
from light, for 60 minutes. Inject a dose of 0.5 ml of each mixture 2.5 IU of C. septicum antitoxins.
intravenously into each of not less than two mice. Observe
Labelling. The label states (1) whether the preparation is a
the mice for 72 hours. Repeat the determinations at least once
toxoid or a vaccine prepared from a whole inactivated culture,
and add together the results of the separate tests that have
or a mixture of the two; (2) that the preparation is to be shaken
been made with mixtures of the same composition such that a
before use; (3) for each target species, the immunising effect
series of totals is obtained, each total representing the
produced (for example, antibody production, protection
mortality due to a mixture of a given composition.
against signs of disease or infection).
The test dose of toxin is the amount present in 0.5 ml of that
mixture that causes the death of one-half of the total number
of mice injected within 72 hours.
Duck Pasteurella Vaccine, Inactivated
Determination of potency of the antitoxin Duck Pasteurella Vaccine, Inactivated consists of an emulsion
Preliminary test. Dilute the test toxin with a suitable liquid or suspension of a virulent strain of Pasteurella multocida
such that 2.0 ml contains 10 times the test dose. Prepare which has been inactivated in such a manner that the toxicity
mixtures such that 5.0 ml of each mixture contains 2.0 ml of the is eliminated and the immunogenic activity is retained.
solution of the toxin and one of a series of graded volumes of
Identification
the preparation under examination. Adjust each mixture to the
same final volume with a suitable liquid. Protects susceptible ducks against infection with P. multocida.
Allow the mixtures to stand at room temperature, protected Tests
from light, for 60 minutes. Inject a dose of 0.5 ml of each mixture
intravenously into each of not less than two mice and observe Safety. Either test A or test B may be carried out.
the mice for 72 hours. If all the mice die, 0.5 ml of the mixture
A. Inject 5 ml subcutaneously into each of four healthy rabbits,
contains less than 0.2 Unit of antitoxin. If none of the mice
weighing between 1.0 and 1.5 kg. Observe the animals for 7
dies, 0.5 ml of the mixture contains more than 0.2 Unit of
days. No untoward reaction except slight and transient local
antitoxin.
swelling occurs.
Final test. Prepare similar fresh mixtures such that 5.0 ml of B. Inject 5 ml subcutaneously into each of two healthy rabbits,
each mixture contains 2.0 ml of the solution of the toxin and each weighing between 1.0 and 1.5 kg, and 0.5 ml
one of a series of graded volumes of the preparation under subcutaneously into each of six mice, each weighing between
examination, separated from each other by steps of not more 25 and 30 g. Observe the animals for 7 days. No untoward
than 20 per cent and covering the expected end-point. Prepare reaction except slight and transient local swelling occurs in
further mixtures of 5.0 ml containing 2.0 ml of the solution of both species of animals.
the toxin and graded volumes of the Standard preparation to
confirm the test dose of the toxin. Sterility (2.2.11). Complies with the test for sterility.

Allow the mixtures to stand at room temperature, protected Potency. Either test A or test B may be carried out.
from light, for 60 minutes. Inject a dose of 0.5 ml of each mixture A. Inject subcutaneously with the minimum dose of the vaccine
intravenously into each of not less than two mice and observe stated on the label three healthy susceptible ducks, between
the mice for 72 hours. The mixture of the antitoxin under 4 and 6 weeks old. Use another two ducks of the same stock
examination which contains 0.2 Unit in 0.5 ml is that mixture and age as unvaccinated controls. Three weeks later, challenge
which causes the death of the same or almost the same number each of the vaccinated and control ducks, subcutaneously
of mice as the mixture containing 0.2 Unit of the Standard with 102 mouse LD50 viable organisms in 0.2 ml of a suitably
preparation in 0.5 ml. Repeat the determinations at least once diluted 18-hour broth culture of the homologous virulent strain
and calculate the average of all valid estimates. Estimates are of P. multocida. Observe the ducks for 7 days. Not less than
not valid unless the Standard preparation gives a result within two of the vaccinated ducks remain in normal health and both
20 per cent of the expected value. the controls die of pasteurellosis.
Limits of error. For the suggested method, the limits of error B. Inject subcutaneously each of six mice, each weighing
(P = 0.95) have been estimated to be (a) 85 per cent and 114 per between 25 and 30 g, with 0.2 ml of the vaccine under
cent when 2 animals per dose are used; (b) 91.5 per cent and examination. Use another six mice of the same stock and weight
109 per cent when 4 animals per dose are used; (c) 93 per cent range as unvaccinated controls. Three weeks later, challenge
and 108 per cent when 6 animals per dose are used. each of the vaccinated and control mice subcutaneously with

1590
IP 2007 EGG DROP SYNDROME 76 (ADENOVIRUS) VACCINE, INACTIVATED

0.2 ml of a suitably diluted 18-hour broth culture of the clinical symptoms of plague. The test is not valid unless the
homologous virulent strain of P. multocida containing 50 control ducks die from duck plague or show typical signs of
mouse LD5o viable organisms. Observe the animals for 7 days. serious infection during the observation period.
All the vaccinated mice survive. The test is not valid unless If potancy test has been performed with satisfactoery results
all the control mice die of pasteurellosis during the observation on a representative batch of the vaccine, it may be omitted as
period. a vaccine test during production on the other batches of
Labelling. The label states (1) type of strain; (2) the vaccine prepared from the same seed lot.
recommended age for vaccination. Labelling. The label states (1) the minimum virus titre per
dose; (2) the recommended age of the birds in which the
vaccine is to be used.
Duck Plague Vaccine, Live
Duck Plague Vaccine, Live is a preparation of attenuated Egg Drop Syndrome 76 (Adenovirus)
strain of duck plague virus. This monograph applies to
vaccines intended for administration to duck for active Vaccine, Inactivated
immunisation against duck plague disease. Egg Drop Syndrome 76 (Adenovirus) Vaccine, Inactivated
consists of an emulsion or a suspension of a suitable strain of
Production egg drop syndrome ‘76 virus (haemagglutinating avian
The vaccine virus is grown in SPF eggs (2.7.7) or in cell adenovirus) which has been inactivated in such a manner that
cultures. The master seed lot complies with the tests for immunogenic activity is retained.
extreneous agents in seed lot (2.7.10).
Production
Substrate for virus propagation
The vaccine strain is propagated in fertilized SPF hen or duck
The vaccine virus is grown in embryonated hens’ eggs or in eggs from healthy flocks (2.7.7) or in suitable cell cultures.
cell cultures obtained from flocks free from specified pathogens
SPF (2.7.7). If the vaccine virus is grown in cell cultures, they Test for inactivation.
comply with the requirements for cell cultures for production The test for inactivation is carried out in fertilized duck eggs
of veterinary vaccines. The vaccine virus is filled with suitable from a flock free from egg drop syndrome ‘76 virus infection
stabilizing agent and freeze dried. or hen eggs from a flock free from specified pathogens, or in
suitable cell cultures, whichever is the most sensitive for the
Identification
vaccine strain; the quantity of virus used in the test is
Protects ducks against duck plague. equivalent to not less than ten doses of the vaccine. No live
virus is detected.
Tests
The vaccine may contain adjuvant
Water (2.3.43). Not more than 3.0 per cent.
Identification
Safety: Inject subcutaneously each of four healthy susceptible
ducks, between 8 and 12 weeks old and each weighing not When inoculated into chicken, the development of specific
less than 600 g, with 1 ml of a 1 : 10 dilution of the reconstituted neutralizing antibodies against egg drop syndrome ‘76
vaccine. Observe the ducks for 14 days. None of the ducks (adenovirus) can be demonstrated by suitable serological
shows any untoward reaction. tests.
Sterility (2.2.11). Complies with the test for sterility. Tests
Potency. Inject subcutaneously each of four healthy Safety. Inject each of ten chickens between 2 and 4 weeks old,
susceptible ducks, between 8 and 12 weeks old and each with two doses and by the route stated on the label. Observe
weighing not less than 600 g, with a volume of the reconstituted the chicken for 14 days. None of the chicken shows any
vaccine containing a quantity of the virus equivalent to the abnormal local or systemic reaction.
minimum dose stated on the label. Fourteen days later,
Sterility (2.2.11). Complies with the test for sterility.
challenge each of the vaccinated ducks and each of two control
ducks of the same stock and weight range, subcutaneously Potency. Inject each of twenty healthy chickens free of
with 10 2 ID50 of virulent duck plague virus. Observe the ducks antibodies (2.7.7) between 3 to 4 weeks old, with the dose and
for 14 days. None of the vaccinated ducks dies or shows any by the route stated on the label. After 21 days, collect serum

1591
FOOT-AND-MOUTH DISEASE VACCINE, INACTIVATED IP 2007

samples from each of the birds as well as ten-control chickens Only a final bulk vaccine that complies with the following
of the same stock and perform haemagglutination inhibition requirements may be used in the preparation of the final lot.
test on each serum using 4 haemagglutinating units of antigen
and chicken erythrocytes. The vaccine passes the potency
Tests
test if the mean antibody titre of the vaccinated group is greater Inactivation. A proportion of each batch of bulk inactivated
than 1:128. The test is valid only if no specific antibody is antigen representing at least 200 doses are tested for freedom
found in the control chicken. from infectious virus by inoculation into sensitive cell culture.
If the potency has been performed with satisfactory results A sample of inactivated antigen is concentrated to volumes
on representative batch of the vaccine from the same seed lot, adequate for inoculation into cell cultures and it must show
it may be omitted as a routine control test during production that the concentrated antigen does not interfere with the
of other batches of the vaccine prepared from the same seed sensitivity or reading of the assay. The sample is passaged 3
lot. times at an interval of 24 to 48 hours and inoculated cell
cultures are examined for the presence of foot-and-mouth
Labelling. The label states (1) the strain used for the disease virus by suitable tests. No cytopathic changes
preparation; (2) the name of any added adjuvant; (3) the route attributable to foot-and-mouth disease virus replication should
of administration. be detected. If infectious foot-and-mouth disease virus is
detected, the bulk antigen is rejected.

Foot-and-Mouth Disease Vaccine, FINAL LOT


Inactivated The final bulk vaccine is distributed aseptically into sterile
containers. The containers are closed so as to avoid
Foot-and-Mouth Disease Vaccine, Inactivated is a liquid
contamination.
preparation containing one or more types of foot-and-mouth
disease virus that have been inactivated in such a manner Only a final lot that complies with each of the requirements
that its immunogenic activity is retained. Depending on the given below under Identification, Tests and Assay may be
number of types of virus incorporated, the vaccine is described released for use
as monovalent, bivalent, trivalent or polyvalent.
Identification
Production
The serum of a foot-and-mouth disease susceptible animal
The virus is grown in suitable cell cultures. The virus is that has been immunized with the vaccine neutralizes the types
separated from cellular material by filtration or other suitable of virus used to prepare the vaccine, when tested by a suitable
procedures and the virus is inactivated using binary method.
ethyleneimine (BEI) in suitable conditions. The antigen may
be concentrated and purified. The antigen is used for the Tests
preparation of vaccine. The vaccine contains a suitable Safety. Use two non-vaccinated cattle not less than 6 months
adjuvant. old free from foot-and-mouth disease antibodies. Inoculate
Only an inactivated antigen suspension that complies with each animal with whole vaccine in case of gel adjuvant vaccine
the requirements mentioned under final bulk vaccine may be and the final bulk in case of oil adjuvant vaccine intradermally
used in the preparation of the final lot. into the tongue at not less than twenty sites with 0.1 ml per
site. Observe the animals for 4 days. No lesions or signs of
FINAL BULK VACCINE foot-and-mouth disease infection should occur. At the end
The final bulk vaccine is prepared from one or more inactivated of the observation period, inject into the same animals three
antigen suspensions. times the dose by the prescribed route as stated on the label.
Observe the animals for a further period of 6 days. No lesions
During inactivation of the virus, samples should be taken at or clinical signs due to foot-and-mouth disease infection should
regular intervals for the purpose of monitoring the rate and occur on the feet or tongue and no evidence of toxicity shall
linearity of the inactivation process. Virus titres in the samples be noticed.
are determined by inoculation into sensitive cell culture. The
infectivities of the timed samples are plotted against time, and Sterility (2.2.11). Complies with the test for sterility.
the inactivation procedure is not considered to be satisfactory Assay. Use three groups of not less than five cattle per group,
unless the extrapolation indicates that there would be less not less than 6 months old, which have never been vaccinated
than one infectious particle per 104 litres of liquid preparation and are free from antibodies neutralizing the different types of
at the end of the inactivation period. foot-and-mouth disease virus in the vaccine. Vaccinate the 3

1592
IP 2007 FOWL POX VACCINE, LIVE

groups by the route stated on the label. Use different doses Tests
of the vaccine for each group without diluting the vaccine.
For example, if 3 ml is one dose, a 1/3 dose of vaccine would Safety. Administer double dose of vaccine subcutaneously
be obtained by injecting 1 ml, and a 1/10 dose would be into each of ten healthy chickens, free of antibodies (2.7.7) of
obtained by injecting 0.3 ml. Three weeks later, challenge all 4 to 6 weeks age. Observe the chickens for 7 days; none of the
the vaccinated animals and a control group of two, susceptible chicken shows untoward reaction other than slight transient
to foot-and-mouth disease with a suspension of virus that is local swelling.
fully virulent and of the same type as that used for preparation Sterility (2.2.11). Complies with the test for sterility.
of the vaccine by inoculating 10,000 ID50 (50 per cent bovine
Potency. Inject subcutaneously each of ten healthy chickens
infectious dose) intradermally into two sites into the tongue
free from antibodies (2.7.7) between 4 to 6 weeks old, with the
(0.1ml per site). The challenge of oil adjuvanted vaccines is
minimum dose stated on the label. Use five healthy chickens
effected 28 days post vaccination. Observe the animals for 8
of the same age group and from the same stock as controls.
days and then sacrifice them. Unprotected animals show
Three weeks later challenge the vaccinated and control
lesions at sites other than the tongue. Protected animals may
chickens by injecting subcutaneously with 0.2 ml of an
display lingual lesions. The test is not valid unless control
18- hour broth culture of the homologous virulent strain of
animals show lesions on at least three feet. From the number
P. multocida diluted suitably so as to contain 102 mouse LD50
of animals protected in each group, calculate the PD50 content
or 200 to 300 CFU in the injected dose in each chicken. Observe
of the vaccine. The potency of the vaccine is expressed as the
the chickens for 14 days; not less than eight out of ten of the
number of 50 per cent cattle protective doses (PD50) contained
vaccinated chickens survive. The test is not valid unless 100
in the dose stated on the label. The vaccine must contain at
per cent of the control chickens die of pasteurellosis within
least 3 PD50 per dose for cattle.
the observation period.
Indirect tests, including post vaccination measurement of virus
Labelling. The label states (1) the serovar(s) used to prepare
neutralizing antibodies in cell culture, or ELISA, may be used
the vaccine, the serovar(s) against which protection is
to assess the potency of a vaccine provided that a statistical
claimed; (2) the method of preparation.
evaluation has established a satisfactory correlation between
the results obtained by the test on the relevant vaccine
serotype and the potency test in cattle.
Fowl Pox Vaccine, Live
The description applies to the testing of a monovalent vaccine.
Polyvalent vaccines may be potency tested by challenging Fowl Pox Vaccine, Live is a preparation of a suitable strain of
each valency as described above. avian pox virus. This monograph applies to vaccines intended
for administration to chickens for active immunization.
Labelling. The label states (1) the method of preparation; (2)
the types and strains of virus used to prepare the vaccine. Production
The vaccine virus is grown in embryonated hens’ eggs or in
Fowl Cholera Vaccine, Inactivated cell cultures.

Fowl Cholera Vaccine, Inactivated is a preparation of 1 or Substrate for virus propagation


more suitable strains of 1 or more serovars of Pasteurella The vaccine virus is grown either in embryonated hens’ eggs
multocida. This monograph applies to vaccines intended for or in avian cell cultures obtained from flocks free from specified
the active immunisation of chickens, turkeys, ducks and geese pathogens SPF (2.7.7). The master seed lot complies with the
against acute fowl cholera. test for extreneous agents in seed lot (2.7.10).
Production Identification
The seed material is inoculated in a suitable medium. If the
The vaccine protects susceptible chicken against fowl pox.
vaccine contains more than 1 strain of bacterium, the different
strains are grown and harvested separately. The bacterial Carry out an immunostaining test in cell cultures to
harvests are inactivated with suitable agent. The vaccine may demonstrate the presence of the vaccine virus. For egg adapted
contain suitable adjuvant. strains, inoculate the vaccine into eggs and notice the
characteristic lesions.
Identification
Tests
Protects susceptible chicken against infection with P.
multocida. Water (2.3.43). Not more than 3.0 per cent.

1593
GOAT POX VACCINE, LIVE IP 2007

Mycoplasmas (2.7.4). The vaccine complies with the test for Production
mycoplasmas.
The virus is propagated in suitable cell culture. The viral
Extraneous agents (2.7.11). The vaccine complies with the suspension is harvested, titrated and may be mixed with a
tests for extraneous agents in batches of finished product. suitable stabilizing agents. The vaccine is then freeze-dried.
Safety. Administer 10 doses of the vaccine to each of ten
chicken 6 to 8 weeks old complying with the requirements of
Identification
test B of the test for freedom from specified pathogens and The vaccine protects susceptible animals against goat pox.
antibodies (2.7.7), SPF chicks and by the route stated on the
label. Observe the birds for 21 days. No chicken dies from Tests
causes attributable to the vaccine or shows signs of toxicity Water (2.3.43). Not more than 3.0 per cent.
other than mild, transient, local reactions. If during the
observation period more than two chickens die from causes Safety
not attributable to the vaccine, repeat the test. A. Inoculate 10 doses of the reconstituted vaccine in each
Virus titre. Not less than 102 EID50/TCID50 of the virus per animal of the three species mentioned below by the route
dose, determining the titre by inoculation into the chorio- stated against each. Administer 0.2 ml intraperitoneally to each
allantoic membrane of SPF embryonated eggs, between 9 and of six mice and 0.5 ml and 1.0 ml subcutaneously to each of
11 days old, or in a cell culture derived from SPF embryos three guinea pigs and three rabbits respectively. Observe the
(2.7.7). animals for 10 days. None of the animals shows an abnormal
reaction.
Sterility (2.2.11). Complies with the test for sterility.
B. Inject 100 doses of the vaccine contained in 1 ml of the
Potency. Carry out a separate potency test for each of the
reconstituted vaccine subcutaneously into each of two
routes of administration stated on the label. Use not less than
susceptible goats, 6 to 8 months old. Observe the goats for 10
ten susceptible chickens, 6 to 8 weeks old, complying with the
days. None of the animals shows abnormalities other than
requirements of test B of the test for freedom from specific
local erythema of not more than 3 cm in diameter around the
pathogens and antibodies (2.7.7), and of the minimum age for
site of injection.
vaccination stated on the label. Use ten birds from the same
flock and weight range as controls. Administer to each chicken Virus titre. Not less than 103 TCID50 of the virus per dose,
a volume of the reconstituted vaccine containing a quantity determining the titre in a suitable cell culture or by inoculation
of the virus equivalent to the minimum titre stated on the into the chorio-allantoic membrane of SPF embryonated eggs
label. After 21 days, challenge each chicken by intrafollicular (2.7.7), 9 to 11 days old.
administration or by scarification with a virulent strain of fowl Sterility (2.2.11). Complies with the test for sterility.
poxvirus. Observe the birds for 14 days. The vaccinated
chickens survive and show no signs of disease except transient Potency. Use nine susceptible goats, 8 to 10 months old. Divide
local reactions of fowl pox within 6 days following the them into three groups of three goats each. Inject
challenge. All control chickens show lesions of fowl pox. subcutaneously 1/20th of the dose of the vaccine stated on
the label into each goat of one group. Administer a quantity
If the potency test has been performed with satisfactory equivalent to the dose of the vaccine stated on the label into
results on a representative batch of the vaccine it may be each goat of the second group. Use the third group as
omitted as a routine test during production of the other batches unvaccinated controls which should be kept along with the
of the vaccine prepared from the same seed lot. inoculated goats. Observe the animals for 14 days and record
Labelling. The label states (1) the minimum virus titre; (2) the the rectal temperature daily of each goat during the observation
age of vaccination; (3) the types and strains of virus used to period. None of the vaccinated goats shows any thermal
prepare the vaccine. reaction or local or generalised lesion. After 21 days, challenge
the vaccinated and control animals with sufficient quantity of
a virulent goat pox virus by intradermal injection. Observe the
animals for 14 days and record the rectal temperature daily of
each goat during the observation period. None of the
Goat Pox Vaccine, Live vaccinated goats shows any thermal reaction or local or
Goat Pox Vaccine, Live is a freeze-dried preparation of an generalised lesion. The test is valid only if the control animals
attenuated strain of goat poxvirus propagated in a suitable develop high fever or show local or generalised lesions. If the
cell culture. It is reconstituted immediately before use by a test for potency has been carried out with satisfactory results
suitable diluent. on a representative batch of vaccine, this test may be omitted

1594
IP 2007 INCLUSION BODY HEPATITIS (IBH) VACCINE, INACTIVATED

as a routine control on other batches of vaccine prepared weight and of the same stock as controls. After 21 days,
from the same seed lot, subject-to agreement by the competent challenge each of the vaccinated rabbits as well as the control
authority. rabbits with an 18 hour old culture of P. multocida containing
not less than 10 LD50 of virulent organisms. Observe the
Labelling. The label states (1) the strain used for the
animals for 7 days; none of the vaccinated animals dies of
preparation; (2) virus titer; (3) dose and age of vaccination.
pasteurellosis. The test is not valid unless both the control
rabbits die of pasteurellosis.
(c) Test on calves. Inject each of not less than 3 healthy
Haemorrhagic Septicaemia Vaccine susceptible calves, weighing not less than 140 kg each with 2
ml of vaccine. Three weeks later, these animals along with two
Haemorrhagic Septicaemia Vaccine (Inactivated) is a healthy animals of the same type are challenged
preparation of Pasteurella multocida. The whole culture is subcutaneously with 18-hours old broth culture of
inactivated which may be heated with a suitable adjuvant. P. multocida equivalent to at least 50 million mouse minimum
infective. Observe the animals for 7 days. Both the controls
Identification should die of pasteurellosis and at least two out of three
The vaccine protects susceptible animals against infection vaccinated animals should survive.
with P. multocida. Potency is conducted on each seed lot or for every fifth batch
produced from the seed lot.
Tests
Labelling. The label states (1) the type and strains of bacteria
Safety. Inject intraperitoneally or intramuscularly into each of used to prepare the vaccine; (2) adjuvant used.
six mice, weighing not less than 18 g, with 0.2 m1 of the vaccine
under examination. Observe the animals for 5 days; no abnormal
systemic reaction occurs.
Inclusion Body Hepatitis (IBH) Vaccine
Inject two seronegative cattle with twice the maximum dose
stated on the label and observe for 10 days for adverse effects.
Inactivated
Sterility (2.2.11). Complies with the test for sterility. Inclusion Body Hepatitis (IBH)/Hydropericardium Syndrome
(HPS) Vaccine (Inactivated) consists of an emulsion or a
Potency. Carry out any of the following three tests. suspension of avian adenovirus type 4 virus which have been
(a) Test on mice. Inject intramuscularly each of fifty mice, inactivated in such a manner that the immunogenic activity is
weighing not less than 18 g, with 0.2 ml of the preparation retained. The vaccine may contain one or more suitable
under examination. Repeat the dose 14 days later. After 7 days, adjuvants.
divide the mice into ten groups of five each. Use another fifty
Production
mice of the same weight and from the same stock as controls.
Divide the controls also into ten groups of five each. Challenge
Substrate for virus propagation
the vaccinated and the control mice with an 8 to 12-hour old
broth culture of a virulent strain of P. multocida in the range Vaccine virus is grown in SPF chicks (2.7.7).
of 10-1 to 10-10. Observe the mice for 5 days and record the
number of vaccinated and control mice found dead in each Inactivation
group. Calculate the median lethal dose (LD50) for the An amplification test for residual live IBH/HPS disease virus
vaccinated and control mice by standard methods. is carried out on each batch of antigen immediately after
The protection provided by the vaccine is calculated as inactivation and on the final bulk vaccine or, if the vaccine
number of protection units using following formula: contains an adjuvant, on the bulk antigen or the mixture of
bulk antigens immediately before the addition of any adjuvant,
Number of protection units=LD50 in control animals - LD50 in
to confirm inactivation; the test is carried out on chickens
vaccinated animals.
from a flock free from specified pathogens. The quantity of
The vaccine passes the test if it provides minimum protection inactivated virus used in the test is equivalent to not less than
of 104 units ten doses of the vaccine. No live virus is detected.
(b) Test on rabbits. Inject intramuscularly each of not less Identification
than six rabbits, each weighing not less than 2.0 kg, with 2 m1
of the vaccine under examination. Use two rabbits of the same Protects chickens against infection of IBH/HPS.

1595
INFECTIOUS AVIAN ENCEPHALOMYELITIS VACCINE, LIVE IP 2007

Tests Tests
Safety. Inject subcutaneously a quantity equivalent to 2 doses Water (2.3.43). Not more than 3.0 per cent.
into each of 20 healthy chickens free from specific antibodies,
of the recommended age at which vaccine is to be used. Safety. Administer twenty five healthy chickens of 6 to 10
Observe the chickens for 14 days, no abnormal systemic or weeks old free from specific antibodies (2.7.7) ten doses of the
local reaction is seen. vaccine by the recommended route. Observe the chickens for
21 days. No chicken develops signs of the disease or dies
Sterility (2.2.11). Complies with the test for sterility. from causes attributable to the vaccine. Repeat the test if
Potency. Inject one dose by the route stated on the label into more than two chickens die from causes not attributable to
each of 20 healthy chickens free from specific antibodies at the vaccine during the observation period.
the minimum recommended age at which vaccine is to be used. Virus titre. Not less than 102.5 TCID50/EID50 of the virus per
Use 10 similar chickens from same source as controls. dose, determining the titre of the virus in cell cultures derived
Challenge chickens after 21 days by intraperitoneal route from SPF embryos or by inoculation into the yolk sac of SPF
with 104 Chick Infective Dose (CID50) of virulent strain of IBH/ embryonated hen eggs (2.7.7), between 5 and 6 days old.
HPS. Observe the chickens of both groups for 14 days. Vaccine
complies with the test if not more than 2 of vaccinated chickens Sterility (2.2.11). Complies with the test for sterility.
die or show signs of disease. At the end of observation period Potency. Carry out a separate potency test for each of the
sacrifice all the chickens. The test is valid only if at least 80 per routes of administration stated on the label. For each of the
cent of control chicks either die or show symptoms or show stated routes, use not less than ten susceptible SPF chickens,
post-mortem lesions on sacrifice at the end of observation 3 weeks old. Administer to each chicken a volume of the
period post challenge. reconstituted vaccine containing a quantity of the virus
If the potency test has been performed with satisfactory equivalent to the minimum virus titre stated on the label. Use
results on representative batch of the vaccine from the same ten chickens of the same flock and age as controls. After 21
seed lot, it may be omitted as a routine control test during days, challenge each chicken in the vaccinated and control
production of other batches of the vaccine prepared from the groups with intracerebral injection of a suitable quantity of a
same seed lot. virulent avian encephalomyelitis virus. Observe the chickens
for another 21 days. Not less than 80 per cent of the vaccinated
Labelling. The label states (1) strain used for vaccine
chickens survive or show no signs of disease and not less
production; (2) recommended age for vaccination.
than 70 per cent of the controls die or develop signs or lesions
of avian encephalomyelitis.
If the immunogenicity test (potency test) has been performed
Infectious Avian Encephalomyelitis with satisfactory results on representative batch of the vaccine
Vaccine, Live from the same seed lot, it may be omitted as a routine control
test during production of other batches of the vaccine prepared
Infectious Avian Encephalomyelitis Vaccine, Live is a freeze- from the same seed lot.
dried preparation of an attenuated strain of infectious avian
encephalomyelitis virus.

Production
Infectious Bursal Vaccine, Inactivated
The virus is grown in SPF embryonated eggs (2.7.7) or in
InInfectious Avian Bursal Disease Vaccine, Inactivated
suitable cell cultures. The master seed lot complies with the
consists of an emulsion or a suspension of a suitable strain of
test for extraneous agents in seed lot (2.7.10).
infectious avian bursal disease virus type 1 which has been
Identification inactivated in such a manner that immunogenic activity is
retained. The vaccine may contain one or more suitable
Inoculate 0.1 ml of the undiluted reconstituted vaccine into adjuvant.
the yolk sac of SPF embryonated eggs, between 5 and 6 days
old. Keep them in an incubator and transfer to the brooder for Production
hatching. Observe the hatched chickens for 7 days. Not less
than 50 per cent of the chickens show the typical symptoms The virus is propagated in fertilized eggs from SPF flocks, in
characteristic of infectious avian encephalomyelitis-like suitable cell cultures or in chickens from a flock free from
weakness or paralysis of legs and tremors. specified pathogens (2.7.7).

1596
IP 2007 INFECTIOUS CORYZA VACCINE

Inactivation the bursa of each chicken from the first group and the second
group. No evidence of infectious bursal disease is seen and
An amplification test for residual live infectious avian bursal
no abnormal local reaction develops.
disease virus is carried out on each batch of antigen
immediately after inactivation and on the final bulk vaccine or, Safety. Inject each of twenty healthy chickens free from specific
if the vaccine contains an adjuvant, on the bulk antigen or the antibodies (2.7.7) between 14 and 28 days old with twice the
mixture of bulk antigens immediately before the addition of minimum vaccinating dose and by one of the routes stated on
any adjuvant, to confirm inactivation; the test is carried out in the label. Observe the chickens for 14 days. No abnormal
fertilized hen eggs or in suitable cell culture or, where chickens local or systemic reaction is seen.
have been used for production of the vaccine, in chickens Sterility (2.2.11). Complies with the test for sterility.
from a flock free from specified pathogens the quantity of
inactivated virus used in the test is equivalent to not less than Potency. Inject each of twenty healthy chickens free from
ten doses of the vaccine. No live virus is detected. specific antibodies (2.7.7) between 3 and 4 weeks old, with
the minimum dose and by the route stated on the label. Use
Identification ten chickens of the same flock and age as controls. After 21
days, collect serum samples from each bird including the ten-
Protects susceptible chickens against infectious bursal disease control chickens and perform quantitative agar gel precipitation
by producing specific antibodies on inoculation. test on each serum sample. The mean antibody titre of sera in
vaccinated group shall be 8.0 and there are no specific
Tests
antibodies in the sera of control chicken.
Inactivation If the potency test has carried out been with satisfactory
For vaccine prepared with embryo-adapted strains of the results on representative batch of the vaccine from the same
virus. Inject two-fifths of a dose into the allantoic cavity or seed lot, it may be omitted as a routine control test during
onto the chorio-allantoic membrane of the SPF embryonated production of other batches of the vaccine prepared from the
hen eggs, between 9 and 10 days old, and incubate at 37°. same seed lot.
Observe for 6 days and pool separately the allantoic fluid Labelling. The label states (1) the type of strain; (2) the
from eggs containing live embryos, and that from eggs recommended age for vaccination.
containing dead embryos, excluding those dying from non-
specific causes within the first 24 hours after inoculation.
Inject into the allantoic cavity of each of the SPF embryonated
hen eggs, between 9 and 10 days old, 0.2 ml of the pooled
Infectious Coryza Vaccine
allantoic fluid from the live embryos or membrane from the Infectious Coryza Vaccine for chickens is a suspension of
dead embryos and incubate at 37° for 6 days. Examine each inactivated culture of one or more virulent strain of
embryo for lesions of infectious bursal disease. The vaccine Haemophilus paragallinarium. A suitable adjuvant may be
complies with the test if there is no evidence of lesions of added.
infectious bursal disease.
Identification
The test is valid only if not more than 20 per cent of the
embryos die at either stage of the test. If more than 20 per cent Protects susceptible chicken against infection with
of the embryos die at either one of the stages of the test, H. paragallinarium organism.
repeat that stage. In any repeat test, not more than 20 per cent
of the embryos die from non-specific causes. Antibiotics may Tests
be used to control extraneous bacterial infection.
Safety. Inject 1.0 ml subcutaneously into each of 10 healthy
For vaccine prepared with strains of virus not adapted to chickens free from antibodies (2.7.7) at the minimum age group
embryos. Inject two doses intramuscularly into each of twenty at which vaccine is intended. Observe these birds for 7 days;
chickens, between 14 and 28 days old, complying with the no bird shows untoward reactions other than slight transient
requirements stated under Test on chicken flocks free from local swelling.
pathogens for the production and quality control vaccines
Sterility (2.2.11). Complies with the test for sterility.
(2.7.7). Four day later, kill ten of the chickens and remove
bursa of fabricius from each chicken, pool the bursa and Potency. Inject subcutaneously each of 10 healthy chickens
homogenise in an equal volume of a suitable liquid. Inject 1 ml free from antibodies (2.7.7) of the minimum age group at which
of the homogenate into each of a further ten chickens of the vaccine is used, with minimum dose stated on the label. Repeat
same flock and age. After 21 days, examine microscopically the vaccination after 2 weeks. Use 10 healthy chickens of

1597
LARYNGOTRACHEITIS VACCINE, LIVE IP 2007

same age group and of same stock as controls. Three weeks more than 20 per cent of the chickens show abnormal clinical
later, challenge vaccinated and control chickens by instillation signs or die from causes not attributable to the vaccine. The
with 0.2 ml of 18-hour broth culture of homologous virulent vaccine complies with the test if no chicken shows notable
strain of H. paragallinarium diluted suitably so as to contain clinical signs of disease or dies from causes attributable to the
lxl06 Chick ID50 by infra-orbital sinus inoculation. Observe the vaccine.
chickens for 7 days for unilateral eye swelling, nasal discharge, Sterility (2.2.11). Complies with the test for sterility.
isolation of the virulent organisms from infra-orbital sinus in a
suitable medium. Not less than 7 vaccinated chickens show Potency. A test is carried out for each route and method of
prevention from lesions and isolation of homologous virulent administration to be recommended using in each case chickens
organisms. The test is not valid unless 70 per cent of control not older than the youngest age to be recommended for
chickens exhibit typical symptoms of eye swelling and nasal vaccination. The quantity of the vaccine virus administered
discharge typical of infectious coryza. to each of 20 chickens is not greater than the minimum virus
titre to be stated on the label and the virus is at the most
Labelling. The label states (1) strains used for preparation; attenuated passage level that will be present in a batch of the
(2) route of administration. vaccine. Vaccinate by a recommended route not less than 20
chickens. Maintain not less than 10 chickens as controls.
Challenge each chicken after 21 days by the intratracheal route
Laryngotracheitis Vaccine, Live with a sufficient quantity of virulent infectious
laryngotracheitis virus. Observe the chickens at least daily
Laryngotracheitis Vaccine, Live is a preparation of a suitable for 7 days after challenge. Record the deaths and the number
strain of Avian infectious laryngotracheitis virus (gallid herpes of surviving chickens that show clinical signs of disease. At
virus 1). This monograph applies to vaccines intended for the end of the observation period kill all the surviving chickens
administration to chickens for active immunization against and carry out examination for macroscopic lesions: mucoid,
laryngotracheitis disease in chickens. hemorrhagic and pseudomembraneous inflammation of the
trachea and orbital sinuses. The test is not valid if during the
Production observation period after challenge less than 90 per cent of the
The vaccine virus is grown in embryonated hens’ eggs or in control chickens die or show severe clinical signs of avian
cell cultures. infectious laryngotracheitis or notable macroscopic lesions
of the trachea and orbital sinuses, or if during the period
If the vaccine virus is grown in embryonated hens’ eggs, they between the vaccination and challenge more than 10 per cent
are obtained from flocks free from specified pathogens (SPF) of the vaccinated or control chickens show notable clinical
(2.7.7). If the vaccine virus is grown in cell cultures, they signs of disease or die from causes not attributable to the
comply with the requirements for cell cultures for production vaccine. The vaccine virus complies with the test if during the
of veterinary vaccines . The vaccine virus is filled with suitable observation period after challenge not less than 90 per cent of
stabilizing agent and freeze dried. The master seed lot complies the vaccinated chickens survive and show no notable clinical
with the tests for extraneous agents in seed lot (2.7.10). signs of disease and/or macroscopically lesions of the trachea
Identification and orbital sinuses.
If the potency test has been performed with satisfactory
When mixed with mono specific laryngotracheitis disease results on a representative batch of the vaccine from the seed
virus antiserum the vaccine no longer infects susceptible cell lot, it may be omitted as a routine control test during production
cultures or embryonated hen eggs, 9 to 11 days old. on other batches of the vaccine prepared from the same seed
lot.
Tests
Labelling. The label states (1) strain of virus used; (2)
Water (2.3.43). Not more than 3.0 per cent. recommended age for vaccination.
Virus titre. Titrate the vaccine virus by inoculation into
embryonated hens’ eggs from an SPF flock or into suitable
cell cultures. The vaccine complies with the test if 1 dose
contains not less than the minimum titre stated on the label.
Leptospira Veterinary Vaccine,
Safety. Use not less than 10 chickens from a healthy flock and
Inactivated
of the youngest age recommended for vaccination. Administer Canine Leptospirosis Vaccine (Inactivated) is a suspension
by eye-drop to each chicken 10 doses of the vaccine. Observe of inactivated whole organisms and/or antigenic extract(s) of
the chickens at least daily for 21 days. The test is not valid if one or more suitable strains of one or more of Leptospira

1598
IP 2007 PESTE DES PETITIS RUMINANTS VACCINE, LIVE

interrogans serovar canicola, serovar icterohaemorrhagiae Not less than four of the control animals die showing typical
or any other epidemiologically appropriate serovar, inactivated leptospira infection. Not less than four of the vaccinated
and prepared in such a way that adequate immunogenicity is animals remain in good health for not less than 14 days after
maintained. the death of the four control animals.

Production Labelling. The label states (1) the strain used for the
preparation; (2) the name of any added adjuvant.
The seed material is cultured in a suitable medium; each strain
is cultivated separately. During production, various parameters
such as growth rate are monitored by suitable methods; the
values are within the limits approved for the particular product. Peste Des Petitis Ruminants Vaccine,
Purity and identity are verified on the harvest using suitable Live
methods. After cultivation, the bacterial harvests are collected
separately and inactivated by a suitable method. The antigen Peste Des Petitis Ruminants Vaccine, Live is a preparation of
may be concentrated. The vaccine may contain an adjuvant. a suitable strain of PPR virus that is attenuated for sheep and
goats.
Inactivation
Production
Carry out a test for inactivation by inoculation on to a specific
medium. Inoculate 1 ml of the vaccine into100 ml of the medium. The vaccine strain is grown in suitable cell cultures. The viral
Incubate at 30° for 14 days, subculture into a further quantity suspension is harvested, mixed with a suitable stabilizing liquid
of the medium and incubate both media at 30° for 14 days: no and freeze-dried.
growth occurs in either medium. At the same time, carry out a
control test by inoculating a further quantity of the medium Batch testing
with the vaccine together with a quantity of a culture containing If the test for potency has been carried out with satisfactory
approximately 100 leptospirae and incubating at 30° Growth results on the representative batch of vaccine, this test may
of leptospirae occurs within 14 days. be omitted as a routine control on other batches of vaccine
prepared from the same seed lot, subject to agreement by a
Identification National Regulatory Authority.
When administered to experimental animals causes the
Identification
appearance of agglutinating antibodies against the serotype
or serotypes used to prepare the vaccine. When injected into the target animals, the vaccine stimulates
the production specific neutralizing antibodies.
Tests
Safety. Use 2 dogs of the minimum age recommended for Tests
vaccination and which do not have antibodies to the leptospira Safety. Administer 0.5 ml of vaccine equivalent to 5 doses
serovar(s) present in the vaccine. Administer 2 doses of the intramuscularly into each of two healthy guinea pigs weighing
vaccine to each dog by a recommended route. Observe the between 200 and 250 g and 0.5 ml each into two healthy
animals for 14 days. The animals remain in good health and no guinea pigs weighing between 200 and 250 g intraperitoneally
abnormal local or systemic reaction occurs. and 0.1 ml of vaccine equivalent to one dose intraperitoneally
Sterility (2.2.11). Complies with the test for sterility. in each of six healthy mice weighing between 17 and 22 g.

Potency. Carry out a separate potency test for each serotype Keep two guinea pigs and two mice as uninoculated controls.
if the vaccine is prepared with different serotypes. Inject each Observe the animals for 3 weeks. At the end of 3 weeks of
of five hamsters not more than 3 months old, the animals observation, all animals are killed for post-mortem examination.
being drawn from the same stock, subcutaneously with 1/40 The vaccine is considered safe if during the first or second
of the dose of the vaccine stated on the label for dogs. Use an test at least 80 per cent of animals remain in good health during
equal number of animals of the species used for the test as the period of observation, and no significant post-mortem
controls. After 15 to 20 days inject intraperitoneally into each lesion is found.
of the vaccinated and control animals an adequate dose of a Inject two susceptible goats of one year old free from
virulent culture of leptospirae of the serotype used to prepare antibodies to rinderpest or PPR by subcutaneous route with a
the vaccine or a suspension of liver or kidney tissue obtained 100 times the dose of vaccine stated on the label. Observe the
from animals infected with the serotype used to prepare the animals for 21 days. No sign of illness attributable to PPR is
vaccine. Observe the animals for 14 days after the injection. noticed

1599
RABIES VETERINARY VACCINE, INACTIVATED (CELL CULTURE) IP 2007

Water (2.3.43). Not more than 3 per cent. virus suspension is harvested on one or more occasions within
5 28 days of inoculation. Multiple harvests from a single
Virus titer. Virus titer not less than 10 TCID50 per dose.
production cell culture may be pooled and considered as a
Extraneous viruses. The reconstituted vaccine when mixed single harvest. The rabies virus is inactivated by a suitable
with specific anti-PPR serum should not produce cytopathic method. The vaccine may contain one or more adjuvants.
effects in susceptible cell cultures and the cells should show
no evidence of the presence of haemadsorbing agents. Inactivation
Sterility (2.2.11). Complies with the test for sterility. A. The test for residual live rabies virus is carried out by
inoculation of the inactivated virus into the same type of cell
Potency. Use not less than six healthy goats and six healthy
culture as that used in the production of the vaccine or a cell
sheep of 1 year old free from antibodies to rinderpest or PPR.
culture shown to be at least as sensitive. The quantity of
Collect sera from the animals before the time of vaccination
inactivated virus used in the test is equivalent to not less than
and 3 weeks after vaccination and just before challenge.
25 doses of the vaccine. After incubation for 4 days, a
Vaccinate two goats and two sheep subcutaneously with 100
subculture is made using trypsinised cells; after incubation
doses per ml; vaccinate two goats and two sheep
for a further 4 days, the cultures are examined for residual live
subcutaneously with 1 dose per ml. Keep the remaining
rabies virus by an immunofluorescence test. No live virus is
animals as the in-contact controls. Monitor each animal for
detected.
clinical signs, in particular respiratory symptoms and record
temperature measurements daily for three weeks. Three weeks B. Inject each of twenty suckling mice, each weighing between
after vaccination challenge the vaccinated animals and in- 12 and 16 g, intracerebrally with not less than 0.03 ml of the
contact controls group with a suspension of virus containing vaccine or antigen under examination. Observe the animals
103 LD50 pathogenic PPRV by subcutaneous route. The animals for 21 days. None of the mice dies or shows any abnormalities
are observed for clinical signs and the body temperatures are attributable to the vaccine. If more than two mice die within 48
recorded daily for two weeks. The vaccine passes the test if hours, repeat the test.
all vaccinated animals resist challenge infection and all the in-
contact controls develop signs of PPR. The serum
Identification
neutralization test must be positive for PPR antibody in When injected into animals, the vaccine stimulates production
vaccinated animal only, in samples taken three weeks after of specific neutralising antibodies.
vaccination.
Tests
If the potency test has been performed with satisfactory
results on a representative batch of the vaccine from the seed Water (2.3.43). Not more than 3.0 per cent (for freeze dried
lot, it may be omitted as a routine control test during production vaccine only).
on other batches of the vaccine prepared from the same seed Safety. Inject each of twenty mice, each weighing between
lot. 12 and 16 g, intracerebrally with not less than 0.03 ml of the
Labelling. The label states (1) cell line used for vaccine vaccine under examination. Observe the animals for 21 days.
manufacture; (2) virus titer per dose; (3) recommended age for None of the mice dies or shows any abnormalities attributable
vaccination. to the vaccine. If more than two mice die within 48 hours
repeat the test. If the vaccine is intended for more than one
species including one belonging to the order of Carnivore,
Rabies Veterinary Vaccine, Inactivated carry out the test in dogs. Otherwise use one of the species
for which the vaccine is intended. Administer, by a
(Cell Culture) recommended route, a double dose of vaccine to each of 2
Rabies Vaccine for Veterinary Use is a preparation of rabies animals having no antibodies against rabies virus. Observe
fixed virus adapted to and propagated in cell culture and the animals for 14 days. No abnormal local or systemic reaction
inactivated by a suitable method. It may be issued as a liquid occurs.
containing a suitable adjuvant or as a freeze-dried preparation Sterility (2.2.11). Complies with the test for sterility.
to be reconstituted with a suitable liquid immediately before
Potency. The potency of rabies vaccine is determined by
use.
comparing the dose necessary to protect mice against the
Production clinical effects of the dose of rabies virus defined below,
administered intracerebrally, with the quantity of a reference
The vaccine is prepared from virus grown either in suitable preparation, calibrated in International Units, necessary to
cell lines or in primary cell cultures from healthy animals. The provide the same protection.

1600
IP 2007 RANIKHET DISEASE VACCINE, INACTIVATED

Preparation of the challenge suspension. Inoculate a group The vaccine complies with the test if the estimated potency is
of mice intracerebrally with the CVS strain of rabies virus and not less than 1 IU in the smallest prescribed dose.
when the mice show signs of rabies, but before they die, kill
Labelling. The label states (1) the strain used for the
the mice and remove the brains and prepare a homogenate of
preparation; (2) the name of any added adjuvant.
the brain tissue in a suitable diluent. Separate gross particulate
matter by centrifugation and use the supernatant liquid as
challenge suspension. Distribute the suspension in small
volumes in ampoules, seal and store at a temperature below
-60°. Thaw one ampoule of the suspension and make serial
Ranikhet Disease Vaccine, Inactivated
dilutions in a suitable diluent. Allocate each dilution to a group Newcastle Disease Vaccine, Inactivated
of 10 mice and inject intracerebrally into each mouse 0.03 ml of
Ranikhet Disease Vaccine, Inactivated consists of an emulsion
the dilution allocated to its group. Observe the animals for 14
or a suspension of a suitable strain of Newcastle disease
days and record the number in each group that, between the
virus (avian paramyxovirus 1) that has been inactivated in
fifth and the fourteenth day, develop signs of rabies. Calculate
such a manner that immunogenic activity is retained.
the ID50 of the undiluted suspension.
Production
Determination of potency of the vaccine
Use in the test healthy mice about 4 weeks old and from the Preparation of the vaccine
same stock. Distribute the mice into at least 10 groups of not The vaccine virus is grown either in embryonated hens’ eggs
less than 10 mice. Prepare at least three serial dilutions of the or in avian cell cultures obtained from flocks free from specified
vaccine under examination and three similar dilutions of the pathogens (SPF) (2.7.7).
reference preparation. Prepare the dilutions such that those
containing the largest quantity of vaccine may be expected to Inactivation. Inject 2/5 of a dose into the allantoic cavity of
protect more than 50 per cent of the animals into which they each of 10 embryonated hen eggs that are 9 to 11 days old
are injected and those containing the smallest quantities of SPF eggs, and incubate. Observe for 6 days and pool
vaccine may be expected to protect less than 50 per cent of separately the allantoic fluid from eggs containing live embryos
the animals into which they are injected. Allocate each dilution and that from eggs containing dead embryos, excluding those
to a different group of mice and inject intraperitoneally into dying within 24 hours of the injection. Examine embryos that
each mouse 0.5 ml of the dilution allocated to its group. Fourteen die within 24 hours of injection for the presence of Newcastle
days after the injection prepare a suspension of the challenge disease virus: the vaccine does not comply with the test if
virus such that, on the basis of the preliminary titration, it Newcastle disease virus is found.
contains about 50 ID50 in each 0.03 ml. Inject intracerebrally Inject into the allantoic cavity of each of 10 SPF eggs,
into each vaccinated mouse 0.03 ml of this suspension. Prepare 9 to 11 days old, 0.2 ml of the pooled allantoic fluid from the
3 suitable serial dilutions of the challenge suspension. Allocate live embryos and, into each of 10 similar eggs, 0.2 ml of the
the challenge suspension and the 3 dilutions one to each of 4 pooled fluid from the dead embryos and incubate for 5 to
groups of 10 unvaccinated mice and inject intracerebrally into 6 days. Test the allantoic fluid from each egg for the presence
each mouse 0.03 ml of the suspension or one of the dilutions of haemagglutinins using chicken erythrocytes.
allocated to its group. Observe the animals in each group for
The vaccine complies with the test if there is no evidence of
14 days. The test is not valid if more than 2 mice of any group
haemagglutinating activity and if not more than 20 per cent of
die within the first 4 days after challenge. Record the numbers
the embryos die at either stage. If more than 20 per cent of the
in each group that show signs of rabies in the period 5 days to
embryos die at one of the stages, repeat that stage; the vaccine
14 days after challenge.
complies with the test if there is no evidence of
The test is not valid unless (a) for both the vaccine under haemagglutinating activity and not more than 20 per cent of
examination and the reference preparation the 50 per cent the embryos die at that stage.
protective dose lies between the smallest and the largest dose
Antibiotics may be used in the test to control extraneous
given to the mice; (b) the titration of the challenge suspension
bacterial infection.
shows that 0.03 ml of the suspension contained at least 10
ID50 and not more than 50 ID50; (c) the confidence limits (P = Identification
0.95) are not less than 25 per cent and not more than 400 per
cent of the estimated potency; (d) the statistical analysis shows When injected into susceptible healthy chicken, free of
a significant slope and no significant deviations from linearity antibodies (2.7.7) the vaccine stimulates the production of
or parallelism of the dose-response lines. specific antibodies against Newcastle disease virus.

1601
RANIKHET DISEASE VACCINE, LIVE (LENTOGENIC STRAIN) IP 2007

Tests less than 10 chickens as controls. Challenge each chicken


after 21 days by the intramuscular route with 6 log10 chick
Safety. Inject ten healthy chickens, free of antibodies (2.7.7) LD50 of the virulent strain of avian paramyxovirus 1. Observe
between 2 and 4 weeks old, with twice the dose and by the the chickens at least daily for 7 days after challenge. At the
route stated on the label. Observe the birds for 21 days. No end of the observation period, calculate the PD50 by standard
abnormal local or systemic reaction’ is observed. statistical methods from the number of chickens that survive
Sterility (2.2.11). Complies with the test for sterility. in each vaccinated group without showing any signs of
Newcastle disease during the 7 days. The vaccine complies
Potency. Either test A or test B may be carried out. with the test if the smallest dose stated on the label
A. Inject intramuscularly each of ten healthy chickens, free corresponds to not less than 50 PD50 and the lower confidence
from antibodies (2.7.7) between 3 and 4 weeks old, with a limit is not less than 35 PD50 per dose. If the lower confidence
volume of the vaccine equivalent to one-fiftieth of a dose. limit is less than 35 PD50 per dose, repeat the test; the vaccine
Use ten chickens of the same stock and age group as controls. must be shown to contain not less than 50 PD50 in the repeat
After 21 days, collect serum samples from each of the test. The test is not valid unless all the control birds die within
vaccinated and unvaccinated chicken. Perform 6 days of challenge.
haemagglutination inhibition test using the method described Labelling. The label states (1) strain of virus used;
below. Use the positive control serum calibrated against a (2) recommended age for vaccination of vaccines for veterinary
Standard preparation of anti-Newcastle disease serum. use.
The vaccine passes the test if a mean HI titre of the vaccinated
group is equal to or greater than 1:16 and that of the
unvaccinated controls is equal to or less than 1:4.
Ranikhet Disease Vaccine, Live
Standard preparation (Lentogenic Strain)
The Standard preparation is the 1st International reference Newcastle Disease Vaccine, Live (Lentogenic strain)
preparation, established in 1966, consisting of freeze-dried
chicken serum (supplied in ampoules containing 320 Units), Ranikhet Disease Vaccine Live (Lentogenic Strain) is a
or another suitable preparation, the potency of which has preparation of a suitable strain of Newcastle disease/Ranikhet
been determined in relation to the International reference disease virus (avian paramyxovirus 1). This monograph
preparation. applies to vaccines intended for administration to chickens
and/or other avian species for active immunization.
Suggested method of haemagglutination inhibition test.
Inactivate the serum samples by heating at 56° for 30 minutes. Production
Add 0.05 ml of saline solution to all the wells in a microtitre
plate and 0.05 ml of the test sera to the first row of wells. Preparation of the vaccine
Prepare two-fold dilutions of the serum samples across the
plate. Add 0.05 ml ofa suspension of Newcastle disease virus The vaccine virus is grown in embryonated hens’ eggs or in
containing 4 haemagglutinating units of inactivated Newcastle cell cultures derived from SPF flocks (2.7.7). The master seed
disease virus. Incubate the plate at 4° for one hour. Add lot complies with the tests for extraneous agents in seed lot
0.05 ml of a 1 per cent suspension of erythrocytes collected (2.7.10).
from chicken, between 3 and 4 weeks old, susceptible to
Newcastle disease.
Identification

Incubate the plate at 4° for one hour. It must be ensured that The vaccine, diluted if necessary and mixed with a
negative and positive control sera are included in the test. monospecific Newcastle disease virus antiserum, no longer
The positive control serum must show a titre of 300 to 400 provokes haemagglutination of chicken red blood cells or
Units determined by calibration against the Standard reference infects embryonated hens’ eggs from an SPF flock or
Preparation. susceptible cell cultures into which it is inoculated.
B. Inject intramuscularly each of three groups of twenty Tests
healthy chicken, free from antibodies (2.7.7) between 3 and 4
weeks old, with five fold dilution of vaccine. Use minimum Water (2.3.43). Not more than 3.0 per cent.
three dilutions. Allocate a different volume to each vaccination For vaccines recommended for use in healthy chickens, free
group. Vaccinate each chicken by the intramuscular route with of antibodies (2.7.7) use not less than 10 chickens from an SPF
the volume of vaccine allocated to its group. Maintain not flock and of the youngest age recommended for vaccination.

1602
IP 2007 RANIKHET DISEASE VACCINE, LIVE (MESOGENIC STRAIN)

For vaccines recommended for use only in avian species other applies to vaccines intended for administration to chickens
than the chicken, use not fewer than 10 birds of the species for active immunization.
likely to be most sensitive to Newcastle disease, that do not
have antibodies against Newcastle disease virus and of the Identification
minimum age recommended for vaccination. Administer to each
bird by eye-drop, or parenterally if only parenteral The vaccine, diluted if necessary and mixed with a
administration is recommended, 10 doses of the vaccine in a monospecific Newcastle disease virus antiserum, no longer
volume suitable for the test. Observe the birds at least daily provokes haemagglutination of chicken red blood cells or
for 21 days. The test is not valid if more than 20 per cent of the infects embryonated hens’ eggs from an SPF flock (2.7.7) or
birds show abnormal clinical signs or die from causes not susceptible cell cultures into which it is inoculated. The master
attributable to the vaccine. The vaccine complies with the test seed lot complies with the tests for extraneous agents in seed
if no bird shows notable clinical signs of disease or dies from lot (2.7.10).
causes attributable to the vaccine.
Tests
Virus titre. Not less than 106 TCID50/EID50 of the virus per
dose, determining the titre in suitable cell culture or by Water (2.3.43). Not more than 3.0 per cent.
inoculation into the allantoic cavity of SPF embryonated eggs,
Safety. Administer fifteen healthy chickens free from
9 to 11 days old.
antibodies (2.7.7), 8 to 9 weeks old, with the minimum ten dose
Sterility (2.2.11). Complies with the test for sterility. and by the route stated on the label. Observe the chickens for
Potency. Carry out a potency test for each of the routes of 21 days. None of them shows abnormal clinical signs or dies
administration stated on the label. For each of the stated routes, due to causes attributable to the vaccine. If more than two
use at least twenty susceptible chickens and of the minimum chickens die during the period of observation due to causes
age recommended for vaccination. other than those attributable to the vaccine, repeat the test.
Administer each chicken with a volume of the reconstituted Virus titre. Not less than 105 TCID50/EID50 of the virus per
vaccine containing a quantity of the virus equivalent to the dose, determining the titre in suitable cell culture or by
minimum titre stated on the label. Use eight chicken of the inoculation into the allantoic cavity of SPF embryonated eggs,
same flock and age as controls. After 14 to 21 days, challenge (2.7.7) between 9 and 11 days old.
each chicken by intramuscular injection with 105 LD50, of a
Sterility (2.2.11). Complies with the test for sterility.
virulent strain of Newcastle disease virus. Observe the animals
for 14 days. The vaccine complies with the test if not more Potency. Carry out a potency test for each of the routes of
than two of the vaccinated chickens die or show signs of administration stated on the label. For each of the stated routes,
disease. The test is valid only if all the control animals die use not less than twenty susceptible healthy chickens free of
within 6 days of inoculation of the virulent challenge strain. antibodies (2.7.7) and of the minimum age recommended for
If the potency test has been performed with satisfactory vaccination. Administer each chicken with a volume of the
results on a representative batch of the vaccine from the seed reconstituted vaccine containing a quantity of the virus
lot, it may be omitted as a routine control test during production equivalent to the minimum titre stated on the label. Use eight
on other batches of the vaccine prepared from the same seed chickens of the same flock and age as controls. After 14 to 21
lot. days, challenge each chicken by intramuscular injection with
105 LD50 of a virulent strain of Newcastle disease virus.
Labelling. The label states (1) strain of virus used; (2) Observe the animals for 14 days. The vaccine complies with
recommended age for vaccination of vaccines for veterinary the test if not more than two of the vaccinated chickens die or
use. show signs of disease. The test is valid only if all the control
chickens die within 6 days of inoculation of the virulent
challenge strain.
Ranikhet Disease Vaccine, Live If the potency test has been performed with satisfactory
(Mesogenic Strain) results on a representative batch of the vaccine from the seed
lot, it may be omitted as a routine control test during production
Newcastle Disease Vaccine, Live (Mesogenic strain) on other batches of the vaccine prepared from the same seed
lot.
Ranikhet Disease Vaccine, Live (Mesogenic Strain) is a
preparation of a suitable strain of Newcastle disease virus Labelling. The label states (1) strain of virus used; (2)
(naturally modified avian paramyxovirus 1). This monograph recommended age for vaccination.

1603
RINDERPEST VACCINE, LIVE IP 2007

Rinderpest Vaccine, Live containing not less than 100 times the minimum dose stated
on the label, using pooled reconstituted contents of not less
Rinderpest Vaccine, Live is a freeze-dried preparation of a live than ten containers taken at random. Observe the animals for
attenuated strain of rinderpest virus that has been modified 21 days. No sign of disease attributable to the vaccine other
by adaptation to and propagation in suitable cell cultures in than mild transient pyrexia is seen.
such a manner that it remains avirulent but retains its
immunogenicity in cattle. It is reconstituted immediately before Virus titer. Not less than 103 TCID50 per dose of cell culture
use with a suitable diluent. vaccine determining the virus content of the reconstituted
vaccine in a suitable cell culture system.
Production Sterility (2.2.11). Complies with the test for sterility.
SEED LOT Potency. Inject subcutaneously each of two susceptible cattle,
The seed lots should be validated for the following tests: free from rinderpest specific antibodies, with a field dose and
a) Purity. It should be free from contaminations with viruses, 1/l0th of the minimum dose respectively stated on the label,
bacteria, fungi and mycoplasmas; considering 103 TCID50 of cell culture vaccine. Use two animals
of the same stock and age as controls. Observe the animals
b) Should not induce any abnormal clinical reaction on
for 21 days. Challenge intramuscularly each animal with a dose
inoculation into rinderpest susceptible cattle;
of not less than 104 ID50 of virulent rinderpest virus. Observe
c) Efficacy. It should induce an immunity to rinderpest in the the animals for 14 days. None of the vaccinated animals shows
susceptible cattle. any clinical signs suggestive of rinderpest. The test is not
CELLS. The primary cells/subcultured cells/continuous cell valid unless both the control animals develop signs of
lines when used should be free from BVD and other rinderpest.
contaminating viruses. If the potency test has been performed with satisfactory
results on a representative batch of the vaccine from the seed
Identification
lot, it may be omitted as a routine control test during production
A. The vaccine protects cattle against virulent rinderpest virus. on other batches of the vaccine prepared from the same seed
B. The seed and the vaccine must be titrated in a suitable cell lot, provided the National Regulatory Authority permits.
culture system capable of supporting the multiplication of the Labelling. The label states (1) the strain of the virus used;
rinderpest virus. (2) the number of doses in the container; (3) that the vaccine
C. When neutralised with a specific rinderpest antiserum, the should be used immediately after reconstitution.
vaccine is no longer capable of protecting cattle against
rinderpest infection.
Tests Salmonella Abortus Equi Vaccine
Water (2.3.43). Not more than 3.0 per cent. Salmonella Abortus Equi Vaccine is a suspension of killed
Mycoplasmas (2.7.4). Complies with the test for absence of mixture of equal parts of pure formalized cultures of smooth
mycoplasmas. laboratory strains of Salmonella abortus equi.
Extraneous pathogens. Complies with the requirements stated Production
under Veterinary Vaccines.
The whole culture or its filtrate or a mixture is inactivated in
Safety such a manner that pathogenecity is eliminated and
A. Use four healthy guinea-pigs, each weighing not less than immunogenic activity is retained. The inactivated cultures may
400 g. Inject two of them intramuscularly and two be treated with a suitable adjuvant.
intraperitoneally with 0.5 ml of the vaccine under examination.
Identification
In addition, inject intraperitoneally each of six mice, each
weighing between 18 and 25 g, with 0.1 ml of the vaccine. It protects susceptible animals against infection with
Observe the animals for 21 days. All the animals remain healthy Salmonella abortus equi.
during the observation period. At the end of the observation
period sacrifice the animals and perform autopsy on each. Tests
None of the animals shows any unusual changes. Safety. Inject 0.5 ml of the vaccine intraperitoneally to each of
B. Inject subcutaneously each of two susceptible cattle, free six mice, each weighing not less than 18 g. Observe the mice
from specific antibodies, with a quantity of the vaccine for 96 hours, none of the mice dies of salmonellosis.

1604
IP 2007 SWINE FEVER VACCINE, LIVE

Sterility (2.2.11). Complies with the test for sterility. the dose of the vaccine and by the route stated on the label.
Use two sheep of the same stock and age as un-vaccinated
Potency. Inject each of twelve mice, each weighing not less
controls. Shave the animals closely on the flank from the
than 18 g, subcutaneously with 0.5 ml of the preparation under
shoulder to the proctodal area. Challenge each animal after 21
examination. Use another twelve mice of the same weight range
days post-vaccination by inoculating intradermally with 0.1
and from the same stock as controls. Three weeks later,
ml of a suspension six ten fold dilution of the sheep pox
challenge the mice from both groups by injecting
challenge virus. Make five separate inoculations in a vertical
intraperitoneally each animal with 0.5 ml of a suspension of an
line for each serial dilution from the anterior to the posterior of
18-hour old culture containing 10 LD50 virulent organisms of
the animals. The titer of the challenge virus is calculated using
S. abortus equi. Observe the mice for 7 days. The vaccine
a standard statistical method for the vaccinated and control
passes the test if not less than nine mice of the vaccinated
sheep by the number of pox lesions observed in each dilution.
group survive. The test is not valid unless not less than nine
The titer of the challenge virus is calculated for the vaccinated
of the control mice succumb to the challenge.
and control animals. The vaccine passes the test if there is a
Labelling. The label states (1) the method of preparation; difference of log titer of more than log10 2.5.
(2) the strains of bacteria used to prepare the vaccine.
Labelling. The label states (1) the strain of virus used in
preparing the vaccine; (2) the virus titre; (3) the minimum dose
and the routes of administration; (4) the volume of the liquid
Sheep Pox Vaccine, Live Attenuated to be used for reconstitution of the vaccine.
Sheep Pox Vaccine, Live Attenuated is a freeze dried
preparation obtained by producing attenuated sheep poxvirus
in a suitable cell culture and mixed with a suitable stabilizer
Swine Fever Vaccine, Live
and freeze dried. The freeze dried vial is reconstituted with a Swine Fever Vaccine, Live is a preparation of a modified strain
suitable diluent and used immediately. of classical swine fever virus, which is devoid of pathogenicity
for the pig by adaptation either to cell cultures or to the rabbit.
Production It is prepared immediately before use by reconstitution from
The vaccine reconstituted with a suitable liquid and diluted if the dried vaccine with a suitable diulent.
necessary to provide a concentration appropriate to the Production
particular test, complies with the requirements stated under
Veterinary Vaccines with the following modifications. The virus is propagated in suitable cell culture. The viral
suspension is harvested, titrated and may be mixed with a
The seed lots used for vaccine preparation must be free from
suitable stabilizing agents. The vaccine is then freeze-dried
extraneous pathogens.
Identification
Identification
LAPINISED VACCINE. Administer 0.5 ml intravenously into
The vaccine specifically protects sheep against sheep pox.
one or more non-immunised rabbits , immunized either with an
Tests identical dose of a vaccine of the same type injected by the
same route between 10 and 60 days before hand or with a
Water (2.3.43). Not more than 3.0 per cent. sufficient dose of antiserum administered a few hours before
Safety. Inoculate not less than 2 sheep of 8 to 12 months old, the injection of the vaccine. Twenty-four hours after the
free from neutralizing antibodies against sheep pox virus, with injection, start recording the temperature of the rabbits in the
ten times the field dose of the vaccine contained in 1 ml by mornings and the evenings until the fifth day after the injection.
subcutaneous route. Observe the animals for 14 days. The The immunised rabbits do not exhibit a rise in temperature of
vaccine complies the test if none of the vaccinated animals more than 1.5°. The test is not valid unless the nonimmunised
show deep necrotic lesion and generalization. rabbits exhibit a rise in temperature of not less than 1.5°.

Virus titre. Not less than 102.5TCID50 of the virus per dose as CELL CULTURE VACCINE. For non-lapinised vaccines
determined by the titre of the vaccine in a suitable cell culture prepared in cell cultures, on administration to pigs immunised
system. with the vaccine specific neutralizing antibodies develop.

Sterility (2.2.11). Complies with the test for sterility. Tests


Potency. Administer each of three sheep, between 8 and 12 Test for extraneous pathogens. The vaccine mixed with a mono
months old, free from sheep pox neutralizing antibodies, with specific antiserum does not cause cytopathic effects in

1605
TETANUS VETERINARY VACCINE IP 2007

susceptible cell cultures. The cells also show no evidence of Tetanus Veterinary Vaccine
the presence of haemadsorbing agents and the cell-culture
fluids are free of haemagglutinating agents when tested with Tetanus Vaccine for Veterinary Use is a preparation of the
chicken erythrocytes. neurotoxin of Clostridium tetani treated in a manner that
eliminates toxicity while maintaining adequate immunogenic
Water (2.3.43) Not more than 3.0 per cent. properties.
Safety. Inject intramuscularly 10 times the minimum dose stated
Production
on the label into each of three healthy piglets, between 6 and
7 weeks old, free from swine fever virus antibodies. Observe The C. tetani strain used for production is cultured in a suitable
the animals for 21 days. Temperature curve should be normal medium. The toxin is purified and then detoxified or it may be
and animals remain in apparent good health and display normal detoxified before purification. The antigenic purity is
growth. determined in Lf units of tetanus toxoid per milligram of protein
and shown to be not less than the value approved for the
Inject intracerebrally 0.03 ml of the vaccine, reconstituted in a
particular product.
manner that 1.0 ml contains 1 ml dose, into each of ten mice,
weighing between 11g and 15g. Observe the mice for 21 days. Choice of vaccine composition
If more than two mice die within the first 48 hours repeat the
The C. tetani strain used in the preparation of the vaccine is
test. The mice show no abnormalities attributable to the
shown to be satisfactory with respect to the production of the
vaccine within the third and twenty-first day after the injection.
neurotoxin. The vaccine is shown to be satisfactory with
Virus titre. Not less than minimum virus titre per dose stated respect to safety and immunogenicity for each species of
on the label, determining the titre in a suitable cell culture. animal for which it is intended. As part of the studies to
demonstrate these characteristics, the tests described below
Sterility (2.2.11). Complies with the test for sterility.
may be used.
Potency. All the animals are healthy and must have had no Production of antigens. The production of the neurotoxin of
contact with swine fever virus and serologically must be free C. tetani is verified by a suitable immunochemical method
from CSF and BVDV antibodies. Use four healthy piglets, carried out on the neurotoxin obtained from the vaccine strain
between 6 and 7 weeks old, for each of the 1:50, 1:200 and under the conditions used for the production of the vaccine.
1:400 dilutions of the vaccine prepared in a suitable diluent or
buffer. Inject intramuscularly 1 ml of these dilutions into each Safety. Carry out the test for each recommended route of
of the piglets in respective groups. Use two healthy susceptible administration and species of animal for which the vaccine is
intended; use animals of the minimum age recommended for
piglets of the same stock and age as control animal group.
vaccination and of the most sensitive category for the species.
After 21 days, inoculate intramuscularly with a sufficient
quantity of the challenge virus in each vaccinated piglet and Use not less than 15 animals, free from antitoxic antibodies for
in each of the two unvaccinated control animals so that at each test. Administer a double dose of vaccine to each animal.
least one of the two unvaccinated control animals die within 7 Administer a single dose of vaccine to each animal after the
to 14 days. Observe the vaccinated animals for 14 days. interval stated on the label. Observe the animals until 14 days
Calculate the number of PD50 contained in the vaccine by after the last administration. The vaccine complies with the
standard statistical methods from the number of animals, which test if no animal shows abnormal local or systemic signs of
survive without showing any signs of swine fever. The disease or dies from causes attributable to the vaccine.
vaccine contains not less than 100 PD50 per dose. The test is
DETOXIFIED HARVEST
not valid unless the control animals die within 7 to 14 days
after inoculation. PD50 correlation studies with virus titres can Absence of toxin and irreversibility of toxoid. Carry out a test
replace the potency test on routine basis. for reversion to toxicity on the detoxified harvest using 2
groups of 5 guinea-pigs, each weighing between 350 to 450 g;
If the test for potency has been carried out with satisfactory
if the vaccine is adsorbed, carry out the test with the shortest
results on a representative batch of vaccine, this test may be
practical time interval before adsorption. Prepare a dilution of
omitted as a routine control on other batches of vaccine
the detoxified harvest so that the guinea-pigs each receive
prepared from the same seed lot, subject-to agreement by the
10 times the amount of toxoid (measured in Lf units) that will
competent authority. be present in a dose of vaccine. Divide the dilution into 2 equal
Labelling. The label states (1) the minimum dose; (2) the parts. Keep one part at 5 ± 3° and the other at 37° for 6 weeks.
recommended routes of administration; (3) the name of any Attribute each dilution to a separate group of guinea-pigs
added adjuvant. and inject into each guinea-pig the dilution attributed to its

1606
IP 2007 TETANUS VETERINARY VACCINE

group. Observe the animals for 21 days. The toxoid complies normally uses rabbits, the potency test described above may
with the test if no guinea-pig shows clinical signs of disease be carried out using ten healthy rabbits, between 3 and 6
or dies from causes attributable to the neurotoxin of C. tetani. months old.

FINAL LOT 1 m1 of serum contains not less than 2.5 Units.

The final bulk vaccine is distributed aseptically into sterile Biological assay of Cl. tetani antitoxin
containers. The containers are closed so as to avoid The potency of Cl. tetani antitoxin is determined by comparing
contamination. the dose necessary to protect mice or other suitable animals
against the toxic effects of a fixed dose of Cl. tetani toxin with
Identification the quantity of a Standard preparation of Cl. tetani antitoxin
Carry out test A if permitted by the nature of the adjuvant. necessary to give the same protection. For this purpose, the
Otherwise carry out test B. Standard preparation of Cl. tetani antitoxin and a suitable
preparation of Cl. tetani toxin are required.
A. Separate the toxoid from the adjuvant. For vaccines
adsorbed on aluminium hydroxide, the following treatment is The test dose of the toxin is determined in relation to the
suitable. Dissolve sufficient sodium citrate in the vaccine Standard preparation of antitoxin and the potency of the
under examination to give a 10 per cent w/v concentration. preparation under examination is then determined in relation
Maintain at 37° for about 16 hours and centrifuge. The clear to the Standard preparation using the test toxin.
supernatant liquid reacts with a suitable tetanus antitoxin and
Standard preparation
yields a precipitate.
The Standard preparation is the 2nd International standard,
B. When inoculated into healthy susceptible animals, the
established in 1969, consisting of freeze-dried hyperimmune
vaccine stimulates the formation of antitoxin to the neurotoxin
horse serum (supplied in ampoules containing 1400 Units) or
of Clostridium tetani or protects the animals against the
another suitable preparation, the potency of which has been
paralytic effects of the toxin.
determined in relation to the International standard.
Tests
Suggested method
Safety. Inject 5 ml of the vaccine subcutaneously as two equally
NOTE - The severity of tetanic paralysis to be regarded as
divided doses at separate sites into each of five guinea pigs,
the end-point is such that the paralysis is readily recognised
each weighing between 350 and 450g. Observe the guinea
but not sufficiently extensive to cause significant suffering.
pigs for 21 days. None of the animals shows any symptoms of
For humane reasons the animals should be examined at least
tetanus or dies from tetanus. If more than one animal dies of
twice a day and should be killed as soon as the end-point is
non-specific causes, repeat the test. No animal dies in the
reached.
second test.
In practice, when using high levels of toxin to determine the
Sterility (2.2.11). Complies with the test for sterility.
test dose, or when using low levels of antitoxin in the
Potency. Test A may be omitted if test B is carried out. Test B preliminary and final tests, the development of paralysis is so
may be ommited if test A is carried out. rapid that the defined end-point is usually synchronous with
A. Inject subcutaneously each of ten guinea pigs, each death. Where death occurs, the combined totals of animals
weighing between 350 and 450 g, with a quantity of the vaccine dying or reaching the paralytic end-point are used in the
not more than the minimum dose stated on the label as the calculations.
primary dose, and 28 days later with a quantity of the vaccine Preparation of test toxin. Prepare Cl. tetani toxin by growing
not more than the minimum dose stated on the label as the Cl. tetani in liquid culture for 8 to 10 days and then adding 1
secondary dose. Fourteen days after the second dose, collect volume of a sterile filtrate of the culture to 1 or 2 volumes of
the blood from each guinea pig, pool the sera and determine glycerine. Store at 0° or at temperatures slightly below it. The
the antitoxin titre by the biological assay of Cl. tetani antitoxin toxin may be dried by a suitable method.
described below.
Selection of test toxin. Select toxin for use as the test toxin by
1 ml of serum contains not less than 7.5 IU per ml or, for determining the following quantities:
vaccine intended for use in equine, not less than 30 IU per ml.
LP/10 dose (Limes paralyticum). This is the smallest quantity
When Cl. tetani vaccine is presented as a component of a of toxin that when mixed with 0.1 Unit of antitoxin and injected
mixed vaccine intended for use in animals other than equine subcutaneously into mice (or guinea-pigs) causes tetanic
and the potency test of the other component or components paralysis in the animals on or by the fourth day after injection.

1607
THEILERIOSIS VACCINE, LIVE IP 2007

Paralytic dose 50. This is the quantity of toxin that when degree of tetanus developing in each group of animals. From
injected subcutaneously into mice (or guinea-pigs) causes the results select suitable mixtures for the final test.
tetanic paralysis in one-half of the animals injected on or by
Final test. Prepare similar fresh mixtures of the test toxin and
the fourth day after injection. A suitable toxin is one that
the preparation under examination such that for each mixture
contains not less than 1000 paralytic dose 50 in an LP/10
the volume selected for injection contains the test dose of
dose.
toxin and one of a series of graded volumes of the preparation
Determination of test dose of toxin. Measure or weigh a under examination, separated from each other by steps of not
quantity of the test toxin and dilute with or dissolve in a suitable more than 20 per cent and covering the expected end-point as
liquid. Reconstitute or dilute the Standard preparation with a determined in the preliminary test. Prepare further mixtures
suitable liquid to give a solution containing 0.5 Unit in 1 ml. with the same amount of test toxin and graded volumes of the
Standard preparation, centered on 0.1 Unit in the volume
Prepare mixtures of the solution of the Standard preparation
selected for injection to confirm the test dose of the toxin.
and the solution of the test toxin such that each mixture
Adjust each mixture to the same final volume with a suitable
contains 0.1 Unit of antitoxin in the volume selected for injection
liquid. Allow the mixture to stand at room temperature, protected
and one of a series of graded volumes of the solution of the
from light, for 6 minutes. Inject a dose of the selected volume
toxin, separated from each other by steps of not more than
of each mixture subcutaneously into each of not less than two
20 per cent and covering the expected end-point. Adjust each
animals of the group to which each mixture has been allocated.
mixture to the same final volume (0.4 to 0.6 ml if mice are used
Observe the animals for 4 days and record daily the degree of
or 4.0 ml if guinea-pigs are used) with a suitable liquid. Allow tetanus developing in each group of animals. The mixture of
the mixtures to stand at room temperature, protected from antitoxin under examination that contains 0.1 Unit in the volume
light, for 60 minutes and then inject a dose of the selected injected is that mixture which causes tetanic paralysis in the
volume of each mixture subcutaneously into each of not less same, or almost the same number of animals as the mixture
than 2 animals of the group to which each mixture has been containing 0.1 Unit of the Standard preparation in the volume
allocated. Observe the animals for 4 days and record daily the injected. Repeat the determination at least once and calculate
degree of tetanus developing in each group of animals. Repeat the average of all valid estimates. Estimates are not valid unless
the determination at least once, add together the results of the the Standard preparation gives a result within 20 per cent of
separate tests that have been made with mixtures of the same the expected value.
composition such that a series of totals is obtained and
determine the mean values. The test dose of the toxin is the Limits of error. For the suggested method, the limits of error
amount present in that mixture that causes tetanic paralysis in (P = 0.95) have been estimated to be 85 to 114 per cent when
one-half of the total number of animals injected with it. When two animals are used per dose, 91.5 to 109 per cent when three
the test dose of the test toxin has been determined, a animals are used per dose, and 93 to 108 per cent when six
concentrated solution of the test toxin may be prepared in a animals are used per dose.
mixture consisting of 1 volume of saline solution and 1 or 2 B. Carry out the biologycal assay of adsorbed tetanus vaccine
volumes of glycerine. This concentrated solution may be as stated under Tetanus Vaccine (Adsorbed).
stored frozen and diluted as required. The specific activity of
This method may only be used for those preparations for
such a solution must be determined at frequent intervals.
which it has been shown to be suitable and in particular
may not be suitable for vaccine with an oil adjuvant. Where
Determination of potency of the antitoxin.
this alternative method is used the estimated potency is not
Preliminary test. Measure or weigh a quantity of the test less than 150 units in the smallest dose stated on the label.
toxin and dilute with or dissolve in a suitable liquid such that Labelling. The label states (1) the name of the adjuvant used;
the solution contains 5 test doses per ml. Prepare mixtures of (2) the preparation should be shaken before use.
the solution of the test toxin and the preparation under
examination such that for each mixture the volume selected
for injection contains the test dose of toxin and one of a series
of graded volumes of the preparation under examination.
Adjust each mixture to the same final volume with a suitable
Theileriosis Vaccine, Live
liquid. Allow the mixtures to stand at room temperature, Theileriosis Vaccine, Live is a lymphoblast cell culture
protected from light, for 60 minutes. Inject a dose of the selected containing Theileria annulata macroschizonts attenuated by
volumes of each mixture subcutaneously into each of not less passage in such a manner that it remains avirulent while it
than two animals of the group to which each mixture has been retains its immunogenicity. The concentrate of the vaccine
allocated. Observe the animals for 4 days and record daily the may be diluted with a suitable diluent after thawing.

1608
IP 2007 THEILERIOSIS VACCINE, LIVE

Production Sterility (2.2.11). Complies with the test for sterility.

Production is based on approved seed lot system. The working Potency. Inject each of three susceptible cattle not less than
seeds are prepared from the master seeds. The production 9 months old with the minimum dose by the route stated on
seed may be prepared by propagating a large number of cells the label. Use two cattle of the same stock and age as controls.
either in suspension/monolayer cultures. After 30 days, challenge each of the vaccinated as well as the
control animals with a preparation of gut homogenate of ticks
Identification containing suitable quantity of sporozoites to infect adult
cattle. Observe the animals for 30 days; none of the vaccinated
Protects susceptible cattle against theileriosis. animals shows any abnormal signs. The test is not valid unless
both the control animals show typical signs of theileriosis. If
Tests these tests have been performed with satisfactory results on
Safety. Inject each of two healthy susceptible cattle not less a representative batch of the vaccine from the seed lot, they
than 9 months old with twice the dose as recommended on the may be omitted by the manufacturer as a routine control on
label. Observe the animals for 30 days. None of the animals other batches of the vaccine prepared from the same seed lot.
shows systemic reactions other than mild pyrexia and mild Labelling. The label states (1) the number of doses in the
swelling of superficial lymph nodes. No schizonts/piroplasms container; (2) the recommended dose; (3) the method of
should be seen in the blood smears/lymph node smear. thawing and reconstitution; (4) that the reconstituted vaccine
Cell count. Contains not less than 2 million live lymphoblast should be used within 3 hours after thawing and
cells in each dose. reconstitution.

1609

You might also like