You are on page 1of 9

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/234138800

Formulation Development and Optimization of Cefuroxime Axetil tablets by


Direct Compression Method and its Stability Studies

Article  in  LATIN AMERICAN JOURNAL OF PHARMACY · March 2012

CITATIONS READS

5 910

5 authors, including:

Iyad Naeem Muhammad Muhammad Harris Shoaib


University of Karachi, Faculty of Pharmacy and Pharmaceutical Scie… University of Karachi
47 PUBLICATIONS   97 CITATIONS    123 PUBLICATIONS   584 CITATIONS   

SEE PROFILE SEE PROFILE

Rabia Ismail Yousuf Muhammad Hanif


University of Karachi Bahauddin Zakariya University
81 PUBLICATIONS   458 CITATIONS    89 PUBLICATIONS   234 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Pharmaceutics View project

Medication disposal; Household practices in Karachi, Pakistan. Need for a medication take-back program. View project

All content following this page was uploaded by Iyad Naeem Muhammad on 06 March 2015.

The user has requested enhancement of the downloaded file.


Latin American Journal of Pharmacy Regular Article
(formerly Acta Farmacéutica Bonaerense) Received: February 3, 2012
Revised version: March 21, 2012
Lat. Am. J. Pharm. 31 (2): 271-8 (2012) Accepted: March 22, 2011

Formulation Development and Optimization of Cefuroxime Axetil


tablets by Direct Compression Method and its Stability Studies
Iyad N. MUHAMMAD, Muhammad H. SHOAIB, Rabia I. YOUSUF,
Muhammad HANIF, Sabahat JABEEN & Tariq ALI

Department of Pharmaceutics, Faculty of Pharmacy, University of Karachi,


Karachi - Sind - 75270 - Pakistan

SUMMARY. Cefuroxime axetil (CA) immediate release (IR) tablets were developed and optimized by di-
rect compression method. Ten formulations were designed and optimized using central composite design
with two main variables, microcrystalline cellulose PH 102 and croscarmellose. Pharmaceutical evaluation
of the formulations was conducted emphasizing on dissolution profile of the drug by USP dissolution test
using apparatus II in 0.07 N HCl and in medium of pH 1.2, 4.5 and 6.8 to determine the dissolution pat-
tern of the low soluble drug. Test formulations were compared against reference brand using f2 similarity
factor. Test formulations were assayed by a validated HPLC method, with acetonitrile and 10 mM ammo-
nium acetate solution (pH = 5.2) in a ratio of 15:85 as mobile phase. Stability studies under stress were
conducted on selected formulations according to ICH guidelines. It was conclusive that stable CA formula-
tions could be developed by direct compression method.

INTRODUCTION
Cefuroxime axetil (CA) is a prodrug which is of cefuroxime axetil has been observed to be
hydrolyzed by esterases of the gut mucosa, re- improved by the use of superdisintegrant and
leasing the active cefuroxime base. After oral amorphous form of CA 8,11,12.
administration, the absorption of CA is carried Optimization techniques have been widely
out from the gastrointestinal tract 1,2. Cefuroxime used in pharmaceutical industries on different
axetil has in vitro microbial activity same as the pharmaceutical dosage forms for the last many
parent cefuroxime 3-5. Cefuroxime is a poorly years specially using statistical design of experi-
water soluble drug with BCS class II characters, ments 13. These includes to observe the physical
low solubility but high permeability 6. Depend- properties of tablets, like disintegration time for
ing on the physical state of the drug powder a directly compressed tablets 14 or to evaluate
some also includes it in BCS class IV drug 7. It is the release properties and bio-adhesiveness of
a well known fact that CA forms a gel like mass newly developed drug delivering formulations.
which hinders the absorption of the drug in vivo Since recent past the trend to substitute the
8. The bioavailability and kinetics of the drug tablet process has overwhelmingly inclined to-
has been observed to vary depending on the wards direct compression due to its convenience
physical form from crystalline to amorphous, and productivity 15, the present study was fo-
with a better absorption of the later one 9 and cused on developing tablet formulations contain-
having more stability in dosage forms 10. Several ing CA by direct compression method that pre-
reports conforms improving the bioavailability sents the best responses after being optimized
of the drug by using different excipients with showing the effects of superdisintegrant and sol-
optimization process and improvising the per- ublisers in providing appropriate release pattern
meability issues of the drug. The release pattern of the drug in different dissolution media.

KEY WORDS: Cefuroxime axetil, Centre composite design, Formulation development, Formulation optimiza-
tion, Similarity factor f2, Stability under stress conditions.
* Author to whom correspondence should be addressed. E-mail: harrisshoaib2000@yahoo.com

ISSN 0326-2383 271


MUHAMMAD I.N., SHOAIB M.H., YOUSUF R.I., HANIF M., JABEEN S. & ALI T.

MATERIAL AND METHODS stant and adding fixed quantities of SLS and
Design of Experiments for optimization magnesium stearate summarized in Table 2. All
Formulations were developed applying a the ingredients were accurately weighed,
central composite response surface method with screened through 40 mesh sieve and geometri-
two independent variables, i.e the directly com- cally mixed in a polyethylene bag for 10 min.
pressible diluent binder microcrystalline cellu- Ac-Di-Sol was then added to the polyethylene
lose (Avicel PH-102®), and super disintegrant bag’s content and again mixed for the next 5
croscarmellose (AC-Di-sol) and observing the min, finally magnesium stearate was added at
response variables that included tablet weight this stage and the content was mixed for anoth-
variation, hardness, thickness, friability, disinte- er 5 min. The content was pressed in single
gration time, and dissolution as mentioned in punch tablet machine (Korsch, Erweka, Ger-
Tables 1 and 2. A total of 10 formulations, in- many); at an appropriate pressure that produced
cluding the centre formulation, in duplicate, caplet shaped tablets having hardness ranging
were developed and optimized according to 2nd between 6 to 10 kg.
order center composite response surface
method. Pharmaceutical evaluation: in vitro
comparative tests with reference product
Active and Excipients The developed tablet formulations were
Amorphous CA compacted powder; obtained evaluated through different pharmacopeial and
with gratitude from Medicaid Pharma, Karachi. non-pharmacopeial pharmaceutical tests against
Microcrystalline cellulose (Avicel® PH 102), and Zinnat® as reference product of cefuroxime ax-
croscarmellose Sodium (Ac-Di-Sol), were of etil to evaluate the pharmaceutical quality.
FMC Corporation, USA, Sodium Lauryl Sulphate,
SLS and magnesium stearate were of Fisher Sci- Tablet Characteristics
entific, UK. Different tablet attributes were observed as
response variables. Weight variation is a direct
Process of Tablet manufacturing by direct indicative to the dosage form uniformity 16, so
compression method the tablet weight variation was carried out in 20
Formulations were designed with varying tablets randomLy selected from each formula-
percentages of excipients keeping the API con- tion and weighing it carefully on a type 1 ana-

Levels (% content)
Factors % Content per tablet
-1.414 -1 0 +1 +1.414

Cefuroxime axetil (equivalent to 250 mg)


Avicel® PH 102 (X1) 20-50 22.93 25 30 35 38
Ac-Di-Sol® (X2) 1-5 1.59 2 3 4 4.41

Table 1. A two factor central composite response surface Design of Experiments.

Coded factors Factors amount Factors amount


Formulation Amount % mg Total wt. Total wt.
Run
Code of Tab rounded up
X1 X2 X1 X2 X1 X2

1 Test 1 -1 -1 25.00 2.00 115.00 9.00 432.00 435


2 Test 2 1 -1 35.00 2.00 160.00 9.00 477.00 480
3 Test 3 -1 1 25.00 4.00 115.00 18.00 441.00 440
4 Test 4 1 1 35.00 4.00 160.00 18.00 486.00 490
5 Test 5 -1.414 0 22.93 3.00 105.68 13.50 427.18 430
6 Test 6 1.414 0 37.07 3.00 169.32 13.50 490.82 490
7 Test 7 0 -1.414 30.00 1.59 137.50 7.14 452.64 455
8 Test 8 0 1.414 30.00 4.41 137.50 19.86 465.36 465
9 Test 9 0 0 30.00 3.00 137.50 13.50 459.00 460
10 Test 10 0 0 30.00 3.00 137.50 13.50 459.00 460
Table 2. Second-order Central Composite Design: 2 Factors 10 Runs with center in duplicate.

272
Latin American Journal of Pharmacy - 31 (2) - 2012

lytical balance (Mettler Toledo, Germany). Bus Module CBM 102, and a UV detector SPD
Tablet friability was carried out using (H Jurgens 10-AVP (Shimadzu Corp, Tokyo, Japan). The data
friabilator GmbH & Co. D-2800 Bremann, Ger- were processed using HPLC Software Class GC
many), after being carefully dedusted and accu- 10 ver. 2.0 (1993-2000), Shimadzu Corp., Kyoto,
rately weighed, and rotated at a speed of 100 Japan. The separation of drug was done by Su-
rotations in 4 min followed by re-dedusting and pelcosil® column LC-18-DB 250 x 4.6 mm, 5 µm
weighing 17. The disintegration test was ob- (Supelco, Bellefonte, PA, USA). The proposed
served to ensure the dispersibility of the tablet method was validated according to International
content in purified water at a temperature of 37 Conference on Harmonization guidelines for re-
± 2 °C within an appropriate time interval. coveries, accuracy and reproducibility 21.
Tablets were placed in each tube of the basket
rack assembly of disintegrator (Erweka disinte- Assay test method
gration tester, ZT2, Heusenstamm, Germany), Twenty tablets were accurately weighed and
and oscillated in water and the time of complete crushed in porcelain mortar; powder, weighing
disintegration noted. averagely for one tablet was taken and dis-
solved in mobile phase and diluted till 0.025 %
Dissolution Release Profile Comparison and filtered through 0.45µ Millipore filter. Sam-
Preparation of dissolution medium ples of the solution were injected and the same
The dissolution medium pH 1.2 medium was procedure was carried out for reference stan-
a 0.1 M solution of HCl, while pH 4.5 phosphate dard solution of CA. Area under the curve was
buffer was 0.68 w/v solution of potassium dihy- measured using standard reference peak area.
drogen phosphate monobasic hydrogen phos-
phate (KH2PO4) pH adjusted with HCl. For pH Accelerated Stability studies under stress
6.8 buffer solution, a 0.2 M solution each of The best formulation observed (Test 8, Test
potassium dihydrogen phosphate (2.74 % w/v) 9 and TEST 10 ) were kept in stability chamber
and sodium hydroxide (0.8 % w/v) was mixed NuAire, USA for accelerated stability studies ac-
with water in a ratio of 50:22:28 and pH adjust- cording to ICH guidelines at 40 ± 1 °C and RH
ed with sodium hydroxide 18. of 75 ± 1 % for 1, 3, and 6 months during which
Method the formulations were tested physically and
United States Pharmacopeial dissolution test chemically for quality attributes.
1 for cefuroxime axetil tablets with USP appara-
tus II was applied to observe the drug release in
900 mL of 0.07 N Hydrochloric acid as dissolu- RESULTS AND DISCUSSION
tion medium, 5 mL samples were withdrawn at Design expert® software was used for the
prescribed time interval of 5, 10, 15, 30, 45, 60, development of immediate release tablets con-
90 and 120 mins. and was replaced by fresh taining CA. The levels of independent variables
medium 19. Cefuroxime axetil was determined at are mentioned in Table 1 and the coded and ac-
278 nm using double beam UV-Vis spectropho- tual quantities for individual ingredients in Table
tometer (1800, Shimadzu, Kyoto, Japan). The 2. The tablets were compressed conveniently by
dissolution profile was also established in pH direct compression using compacted drug pow-
1.2, 4.5 and 6.8 buffer solutions as dissolution der, directly compressible ingredients and single
medium to observe the release of the drug in punch machine. The formulations were evaluat-
the developed formulations. ed for pharmaceutical attributes. All the formula-
Assay method tions were manufactured having thickness under
The test formulations were assayed and uni- the prescribed range of ± 5 %. Variation in tablet
formity of dosage form units was determined by thickness is often governed by the flowability of
a modified and validated high performance liq- the tablet content and filling of the dye 22. Avicel
uid chromatographic (HPLC) method reported PH102 has been observed to acquire better
by Darouiche & Hamill 20. The mobile phase flowability with promising tables binding capa-
was composed of 15 % v/v acetonitrile and 85 % bilitites and this indicates the binding properties
v/v 10 mM ammonium acetate solution (pH = of the binders 23. The hardness of the ten pre-
5.2) with a flow rate of 1.0 mL/min and a detec- pared tablet formulations were found to have a
tion wavelength of 254 nm and injection volume range between 6.05 and 10.34 kg.N with a mean
100 µL. The liquid chromatography system con- value 7.98 kg.N. The hardness is a direct indica-
sisted of a pump LC- 10 ADVP, Communication tive of the binding charectrstics of the filler

273
MUHAMMAD I.N., SHOAIB M.H., YOUSUF R.I., HANIF M., JABEEN S. & ALI T.

binder and the compressional force with which In the present study a modified, more sim-
the tablet content has been compressed 24. The ple, precise and less tedious method was ap-
results of the present study explains well about plied that could be used for the formulations
the compaction tendency of Avicel® PH102 and and biological fluids. The method was having
its role in tablet hardness and thickness as was linearity over a range of 50 µg/mL to 0.39
previously reported 25. The developed formula- µg/mL with r2 value of 0.9849 and retention
tions has shown a consistency in weight varia- time between 4-6 min the accuracy for LLOQ
tion and the tablet weight lied under the accept- was 90 % with CV% of 1.88 % as precision. The
able range. A direct relationship between the within day (n = 5) accuracy for the concentra-
tablets content with that of pharmaceutical at- tions was ranging from 94.35 to 98.58 with a
tributes was observed, the summary of the phar- precision of 0.8-4.27. The between day accuracy
maceutical attributes are mentioned in Table 3. and precision (n = 3) for the same set of con-

Thickness Hardness Weight variation Friability


Formulations
(mm) (kg) (mg) (%)

REF coated 5.10 ± 0.06 NA 460.99 ± 0.53 NA


Test 1 4.12 ± 0.07 6.29 ± 0.17 436.80 ± 2.27 1.09
Test 2 5.19 ± 0.07 6.08 ± 0.15 482.43 ± 3.38 0.80
Test 3 4.15 ± 0.04 7.11 ± 0.15 442.95 ± 6.28 1.06
Test 4 5.25 ± 0.07 6.73 ± 0.33 489.86 ± 4.53 1.80
Test 5 4.04 ± 0.07 6.05 ± 0.21 433.02 ± 2.50 1.03
Test 6 5.23 ± 0.10 6.99 ± 0.23 489.68 ± 5.19 1.00
Test 7 4.32 ± 0.11 9.88 ± 0.41 455.01 ± 4.10 1.27
Test 8 4.76 ± 0.14 10.22 ± 0.06 465.68 ± 4.80 0.65
Test 9 4.77 ± 0.06 10.34 ± 0.09 462.89 ± 4.23 0.91
Test 10 4.74 ± 0.06 10.10 ± 0.13 462.67 ± 2.07 0.65
Table 3. Relationship between tablets content and pharmaceutical attributes.

Conc. range for Linearity µg/mL 50 - 0.2604


Coefficient r2 0.9849
LLOQ/Sensitivity 0.39
LLOD 0.2604

Conc. µg/mL After 4 h After 7 h After 9 h

25 98.46 97.64 96.48


Within -Day Accuracy
6.25 98.58 97.24 95.74
3.125 98.04 94.72 95.62
0.781 96.78 97.85 94.35

25 4.27 0.23 0.64


6.25 0.80 2.79 3.76
Within - Day Precision
3.125 0.83 1.25 2.73
0.781 1.67 2.72 2.34
Day 1 Day 2 Day 3

25 97.43 97.47 97.83


Between - Day Accuracy
6.25 101.25 97.82 96.82
3.125 97.88 97.74 96.35
0.781 97.24 93.83 96.36

25 2.07 2.14 3.12


6.25 2.55 2.75 1.47
Between - Day Precision
3.125 2.47 3.82 4.84
0.781 2.82 2.78 2.18
Table 4. Validation parameters.

274
Latin American Journal of Pharmacy - 31 (2) - 2012

centrations within three days were ranging from The conventional release oral solid dosge
93.83 to 101.25 and 1.47-4.84. The validation form needs to be disintegrated from its intact
summary is mentioned in Table 4. The assay re- form, and get dissolved to show its release pat-
sult showed the pharmacopeial requirement for tern. In any such case where the disintegration
the developed formulations i.e. 90-110 %. There is retarding or is slow, the dissolution is affected
are some other works that has proposed some drastically by the disintegration, hence it be-
modified methods for the determination of CA comes the rate limiting step in release pattern.
by other techniques 26-29, an assay method in The formulation composition of the immedite
plasma was developed by Szlagowska et al. 30, release dosage form hence should be fabricated
in which a modified and validated method was such that it assures the disintegration and simul-
presented for the determination of CA in plas- taneous dissolution in the upper gastrointestinal
ma. tract 31. According to the Food and Drug Admin-

Figure 1. Release pattern of developed formulations in different dissolution media.

Formulations 0.07 N HCl pH 1.2 pH 4.5 pH 6.8

TEST 1 38.23 31.33 30.47 41.27


TEST 2 43.30 42.20 41.41 41.81
TEST 3 46.12 36.14 38.77 48.80
TEST 4 43.37 46.02 45.64 40.33
TEST 5 29.58 29.87 30.11 34.41
TEST 6 48.71 47.40 46.06 48.25
TEST 7 46.83 34.22 37.94 41.07
TEST 8 49.36 49.66 49.12 48.27
TEST 9 49.77 46.67 41.50 49.97
TEST 10 49.70 46.61 40.74 46.31
Table 5. Drug Release at different dissolution medium f 2 Similarity.

275
MUHAMMAD I.N., SHOAIB M.H., YOUSUF R.I., HANIF M., JABEEN S. & ALI T.

Formulation
Study Period Tests
T8 T9 T 10

Physical Hardness Kg.N 10.22 10.34 10.10


Friability % 00.65 00.91 00.65
0 month Pharmaceutical Two point Dissolution at 15 min 75.12 75.89 75.12
0.07 N HCl % Drug Release 45 min 86.64 86.95 87.41

Assay % 94.12 90.46 93.27

Physical Hardness Kg.N 10.17 09.97 09.76


Friability % 00.72 00.98 00.77

1 month Pharmaceutical Two point Dissolution 15 min 68.23 69.34 73.54


at 0.07 N HCl 45 min 82.65 82.43 85.98

Assay % 89.12 89.83 89.78

Physical Hardness Kg.N 09.65 09.72 08.54


Friability % 00.71 01.01 00.98
3 month Pharmaceutical Two point Dissolution at 0.07 N HCl 15 min 67.45 67.24 72.54
45 min 80.13 78.378 84.23

Assay % 86.33 88.76 88.43

Physical Hardness Kg.N 09.64 08.76 08.52


Friability % 00.85 01.12 01.08
6 month Pharmaceutical Two point Dissolution at 0.07 N HCl 15 min 68.34 62.77 67.43
45 min 79.34 77.33 77.43

Assay % 87.43 87.35 86.32

Table 6. Stability studies of selected cefuroxime axetil 250 mg formulations.

istration, US, the dissolution profiles of a tablet weak alkaline in nature the dissolution at pH
formulation should be obtained in at least three 6.8 showed a little hindered release, but has
dissolution media having pH values i.e 1.2, 4.5, demonstrated similarity in release with an f 2 val-
and 6.8. In the present study, the in vitro release ue of 49.9 as compared to reference formula-
pattern of the drug was observed in pharma- tion. The summary of the release pattern is
copeial dissolution medium and was compared mentioned in Table 5, while Figure 1 represents
using f 2 similarity factor with the reference drug the release pattern graphically.
formulation. The drug release in the present As for the optimization, the response surface
study was observed in pH 1.2 dissolution medi- method graph (Fig. 2) represents the best possi-
um, and pH 4.5 and 6.8 phosphate buffer solu- ble combination of avicel and crosscarmellose
tions. As amorphous form of CA was reportedly that would show the desired similarity factor.
having better solubility and permeability than The possible test designs that were predic-
the crystalline form, the same was established in tive to be optimized are mentioned in Figure 3.
the current study. It is reported in the literature Formulation TEST 8 was the closest formulation
that the dissolution and pH-solubility data of and declared as the optimized and stable formu-
both prodrugs and drugs could be relevant 32,33 lation of CA with sufficient dissolution pattern.
and the FDA and ICH recommends f 2 similarity The stability studies on the test formulations
factor to be adopted for comparing the dissolu- (Table 6) has shown TEST-8 and center point
tion data profile 34. The comparison of the de- formulations TEST 9 and TEST-10 to be the sta-
veloped formulations based on f 2 similarity fac- ble ones of which TEST 8 has shown a stability
tor showed formulation TEST 8, TEST 9 and till six months of storage.
TEST 10 among all the developed formulations Although the stability of the formulations
that had good similarity in vitro in pharma- were observed for the uncoated tablets, under
copeial dissolution medium. As the drug is a stressed conditions, amorphous form of CA has

276
Latin American Journal of Pharmacy - 31 (2) - 2012

Figure 3. Possible points of optimization.

Figure 2. RSM optimization graphical representation 11. Chen, J.F., J.Y. Zhang, Z.G. Shen, J. Zhong & J.
based on f 2 similarity for the developed formulations. Yun (2006) Ind.Eng. Chem. Res. 45: 8723-7.
12. Mosharrafand, M. & C. Nystrom. (1995) Int. J.
Pharm. 122: 35-47.
been reported to obey the first order reversible 13. Madgulkar, A., M. Bhalekar & M. Swami (2009)
degradation reaction 35 which was rapid in pres- AAPS PharmSciTech 10: 743-51.
14. Abed, K.K., A.A. Hussein, M.M. Ghareeb &
ence of high humidity and temperatures 36 and
A.A. Abdulrasool. (2010) AAPS PharmSciTech
similar results were also observed in tablet
11: 356-61.
dosage form 37. The use of uncoated tablet in 15. Shangraw, R.F. & D.A. Demarest (1993)
the present study was to observe the stability of Pharm. Technol. 17: 32-44.
unprotected directly compressed tablets which 16. USP 32 NF 27 (2009) Uniformity of dosage
was suggestive that the stability could be im- units <905>. The United States Pharmacopeial
proved by the addition of stabilizers and coat- Convention; p. 382.
ing. 17. USP 32 NF 27 (2009) Tablet friability test
<1216>. The United States Pharmacopeial Con-
vention; p.725.
REFERENCES 18. Siewert, M. (1997) Pharm. Ind. 59: 760-6.
1. Harding, S.M., P.E. Williams & J. Ayrton. 19. USP 30 NF 25 (2007) Cefuroxime axetil tablets.
(1984) Antimicrob. Agents Chemother. 25: 78- The United States Pharmacopeial Convention;
82. p. 1684.
2. Fuchs, T. (2007) “Case study: Cefuroxime ax- 20. Darouiche, R.O. & R.J. Hamill. (1994) Antimi-
etil: An oral prodrug of cefuroxime”. 1195-205. crob. Agents Chemother. 38: 1059-64.
3. Marx, M.A. & W.K. Fant. (1988) Drug. Intell. 21. ICH. Harmonised tripartite guideline (2005)
Clin. Pharm. 22: 651-8. Q2(r1)-validation of analytical procedures:
4. Kayser, F.H. & J. Bille (1990) Schweiz. Rund- Text and methodology. International confer-
sch. Med. Prax. 79:1383-6. ence on Harmonisation of technical require-
5. Emmerson, A.M. (1988) J. Antimicrob. Chemo- ments for registration of pharmaceuticals for
ther. 22:101-4. human use.
6. Kanfer, I. (2000) J. Pharm. Pharm. Sci. 5: 1-4. 22. Davies, P.N. & J.M. Newton1(1995) “Pharma-
7. Therapeutic Systems Research Laboratories, ceutical powder compaction technology”. New
TSRL Inc. Available at <http://www.tsrlinc. York: Marcel Dekker;
com/resources/services/bcs/search.cfm. Bio- 23. Guy, A. (2009) “Cellulose, microcrystalline”. 6th
pharmaceutics classification system>. ed. Grayslake and Washington DC: Pharma-
8. Jennewein, H. & J. Raneburger (2004) Compo- ceutical Press and American Pharmacists Asso-
sitions comprising cefuroxime axetil. United ciation.
States: Biochemie, Gesellschaft M.B.H. United 24. Siddiqui, A. & S. Nazzal (2007) Int. J. Pharm.
States Patent 6727243. 341:173-80.
9. Crisp, H.A. & J.C. Clayton (1985) Amorphous 25. Lahdenpaa, E., M. Niskanen & J. Yliruusi.
form of cefuroxime ester. US Pat 4562181. USA. (1997) Eur. J. Pharm. Biopharm. 43: 315-22.
10. Harding, S.M. (1990) Res. Clin. Forums 12: 23- ˛
26. Krzek, J. & M. Dabrowska-Tylka (2003) Chro-
9. matographia 58: 231-4.

277
MUHAMMAD I.N., SHOAIB M.H., YOUSUF R.I., HANIF M., JABEEN S. & ALI T.

27. Can, N.O., Altiokka G. & H.Y. Aboul-Enein 34. Food and Drug Administration (1997) Guid-
(2006) Anal. Chim. Acta 576: 246-52. ance for industry: Dissolution testing of imme-
28. Ivana, I., Z. Ljiljana & Z. Mira (2006) J. Chro- diate release solid oral dosage forms. Rockville,
matogr. A 1119: 209-15. MD: U.S.
29. Ranjane, P.N., S.V. Gandhi, S.S. Kadukar & 35. Jelinska, A., I. Dudzinska, M. Zajac & I. Os-
K.G. Bothara (2010) J. Chromatogr. Sci. 48: 26- zczpowicz. (2006) J. Pharm. Biomed. Anal. 41:
8. 1075-81.
30. Szlagowska, A., M. Kaza & P.J. Rudzki. (2010) 36. Zajac, M., A. Jelinska, L. Dobrowolski & I. Os-
Acta Pol. Pharm. 67: 677-81. zczapowicz (2003) J. Pharm. Biomed. Anal.
31. Dressman, J.B. & C. Reppas. (2000) Eur. J. 32: 1181-7.
Pharm. Sci. 11: S73-80. 37. Jelinska, A., I. Dudzinska, M. Zajac, I. Oszcza-
32. Food and Drug Administration (2001). powicz & W. Krzewski (2005) Acta Pol.
33. Food and Drug Administration (2003) Guid- Pharm. 62: 183-7.
ance for industry: Bioavailability and bioe-
quivalence studies for orally administered drug
products-general considerations.

278

View publication stats

You might also like