You are on page 1of 113

PREPARATION AND EVALUATION OF LIPOSPHERE BASED TOPICAL DRUG DELIVERY SYSTEM CONTAINING A NSAID DRUG

THESIS

Submitted in partial fulfillment for the degree of

MASTER OF PHARMACY
In Industrial Pharmacy
In The Faculty of Medicine Sant Gadge Baba Amravati University, Amravati.
RESEARCH SCHOLAR Mr. LEELADHAR PRAJAPATI B. PHARM RESEARCH GUIDE Dr. P. S. KAWTIKWAR (M. PHARM, Ph.D.)

2012-2013 DEPARTMENT OF PHARMACEUTICS SUDHAKARRAO NAIK INSTITUTE OF PHARMACY, PUSAD-445204 SANT GADGE BABA AMRAVATI UNIVERSITY, AMRAWATI MAHARASHTRA. (INDIA)

Affectionately Dedicated To,


God,
Who is always with me.

My family,
Whose affection and love are infinite with me in adversity and prosperity.

The Guide,
To whom, I shall remain indebted for giving new shape and path to my life.

(Research Guide) Dr. P.S. Kawtikwar


M.Pharm. Ph.D. Professor & H.O.D. Department of Pharmaceutics, S. N. Institute of Pharmacy, Pusad- 445 204.MS (India)

CERTIFICATE
This is to certify that the investigations described in this thesis entitled Preparation and evaluation of liposphere based topical drug delivery system containing a NSAID drug was carried out by Mr. Leeladhar Prajapati in the laboratories of Department of pharmaceutics, S.N. Institute of pharmacy, Pusad under my supervision and guidance. The thesis work was carried out and submitted in partial fulfillment of the requirements for the Degree of Master of Pharmacy in Industrial Pharmacy, in the faculty of Medicine of Sant Gadge Baba Amravati University, Amravati. This thesis is now ready for examination and evaluation.

Date: Place:

Dr. P. S. Kawtikwar
[Research Guide]

Dr. D. M. Sakarkar
M.pharm. Ph.D. Principal, S.N. Institute of Pharmacy, Pusad -445204, MS (India)

CERTIFICATE
This is to certify that the investigations described in this thesis entitled Preparation and evaluation of liposphere based topical drug delivery system containing a NSAID drug was carried out by Mr. Leeladhar Prajapati in the laboratories of Department of pharmaceutics, S.N. Institute of pharmacy, Pusad under my supervision and guidance. The thesis work was carried out and submitted in partial fulfillment of the requirements for the Degree of Master of Pharmacy in Industrial Pharmacy, in the faculty of Medicine of Sant Gadge Baba Amravati University, Amravati. The thesis is now ready for examination and evaluation.

Date: Place:

Dr. D. M. Sakarkar Principal

Mr. Leeladhar Prajapati


B. Pharm. Department of Pharmaceutics S. N. Institute of Pharmacy Pusad-445204, MS (India)

DECLARATION
It gives me great pleasure and satisfaction to declare that the thesis entitled Preparation and evaluation of liposphere based topical drug delivery system containing a NSAID drug was reviewed and submitted in partial fulfillment of the requirements for the Degree of Master of Pharmacy in Industrial Pharmacy, in the Faculty of Medicine of Sant Gadge baba Amravati University, Amravati under guidance and supervision of Dr. P.S. Kawtikwar and I hereby declare that This thesis is now ready for examination and evaluation.

Date: Place:

Mr. Leeladhar Prajapati

SUDHAKARRAO NAIK INSTITUTE OF PHARMACY


Nagpur road, Pusad Dist: Yavatmal (M.S) 445204 Phone (07233)-247308; Fax (07233)-247308 Web site: www.sniop.ac.in

INSTITUTIONAL ANIMAL ETHICS COMMITTEE


(Reg. No. CPCSEA/729/02/a/ CPCSEA)

Prof. S.V. Tembhurne Member Secretary (IAEC)

Ref. No. SNIOP/IAEC/2012-13/CPCSEA/IAEC/IP-PL/12-2012

Mr. Leeladhar Prajapati (M.pharm Student) Department of Pharmaceutics Sudhakarrao Naik Institute of Pharmacy, pusad Dist-Yavatmal-445204

This is to certify that the proposal of Mr.Leeladhar Prajapati for the Study entitled Preparation and evaluation of liposphere based

topical drug delivery system containing a NSAID drug was approved by


Institutional Animal Ethics Committee (IAEC) of S.N. Institute of Pharmacy Pusad in the IAEC meeting held on dated 07/01/2013 and the proposal number CPCSEA/IAEC/IP-PL/12-2012

Hence the certificate is issued.

Place: Pusad

[Prof. S.V. Tembhurne]

LIST OF TABLES
Table No. 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. 20. 21. Title Composition and active ingredients for formulations of lipospheres Factors influencing morphology of Lipospheres Factors influencing entrapment efficiency Factors influencing drug release List of instrument used List of chemical and reagent used Formulations codes of lipospheres different batchs (F1-F12) Topical gel formulation of optimized batch Physical characters of Aceclofenac drug Interpretation of Aceclofenac IR UV calibration curve reading of Aceclofenac in phosphate buffer saline pH 7.4 Evaluations of lipospehers Mean particle size (mps) of different formulation of liposphers Cumulative percentage drug release from various formulations of lipospheres (F1-F4) Cumulative percentage drug release from various formulations of lipospheres (F5-F8) Cumulative percentage drug release from various formulations of lipospheres (F9-F12) Drug release kinetics for the various formulations of lipospheres Result of pH, Viscosity, Drug content and Spreadability Cumulative percent (%) drug release of optimize batch (F8, LF8) Percentage inhibition for anti-inflammatory activity Stability studies for the formulation LF7 and LF8 Page No. 19 25 26 27 45 46 51 55 60 62 63 64 66 72 73 74 75 77 79 81 82

LIST OF FIGURES
Fig. No. 1 2 3 4 5 6 7 8 9 10 11. 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 Schematic liposphere A cross sectional view of human skin Epidermis with Stratum corneum including a corneocytes cluster Skin appendages Plasma drug concentration profile for controlled release Schematic representation of the methods of production of LS: melt dispersion and solvent evaporation UV spectrum of aceclofenac FTIR Spectra of aceclofenac drug Calibration Curve of Aceclofenac Entrapment efficiency (F1-F12) Photograph of optimized liposphers batch F7 Photograph of optimized liposphers batch F8 FT-IR spectra of Aceclofenac FT-IR Spectra of physical mixture DSC Thermogram of Aceclofenac DSC Thermogram of optimized formulation F8 and F6 SEM images of drug loaded lipospheres In vitro drug release of batch F1,F2,F3,F4 In vitro drug release of batch F4,F5,F6,F8 In vitro drug release of batch F9,F10,F11,F12 First order drug release plot of optimized batch F7 First order drug release plot of optimized batch F8 Higuchis drug release plot of optimized batch F7 Higuchis drug release plot of optimized batch F8 Peppas korsmeyers drug release plot of optimized batch F7 Peppas korsmeyers drug release plot of optimized batch F8 Title Interactions of phospholipids in aqueous media. Page No. 6 7 8 9 13 17 21 61 61 63 64 65 65 67 68 69 70 70,71 72 73 74 76 76 76 76 76 76

28 29 30

Lipospheres based gel of optimized batch In vitro release profile of optimized formulation F8 and LF8 Skin-irritation testing (Draize patch test)

78 80 80

LIST OF ABBREVATIONS

Base unit Hour Second Minute Milliliter Micrometer Nanometer Millimeter Milligram Gram Cubic meter Revolutions per minute Centimeter Potassium Bromide Centipoise BP LS

Symbol h s min ml m nm mm mg g m3 rpm cm KBr cps British Pharmacopoeia Liposphere

ACKNOWLEDGEMENT
First and foremost I would like to express my deepest pray to ALL MIGHTY, love and thanks to my father Mr. Teerath Prasad Prajapati, my mother Mrs. Shivkali Prajapati, for their extreme patience and support. I know this took a long time but your sacrifice and understanding has allowed me to persevere. Let me start by expressing my sincere and special thanks to my esteem guide Respected Dr. P. S. Kawtikwar Sir, H.O.D. Dept. of Pharmaceutics, Sudhakarrao Naik Institute of pharmacy, Pusad. I am thankful to him for giving me freedom to work, timely advice and valuable suggestions. Under his constant guidance, encouragement, and positive attitude towards work has instilled more confidence in me. Thank you Sir for all you has done. I owe a great debt of gratitude to Dr. D. M. Sakarkar Sir, Principal, Sudhakarrao Naik Institute of pharmacy, Pusad for providing the necessary facilities. This thesis would not have become a reality without his constant quest for knowledge and critical evaluation. Im grateful to Prof. R. B. Wakade Sir, Prof. A.H.harsulkar sir, Prof. A.M.Mahale sir, & Prof. J.K. Jadhav, Prof. A.R. Tapas sir and prof. S. V. Tembhurne sir for their proper orientation to my work to make it possible. I express my deepest thanks to VAV Life Science Ms. Stuti Singh mam and Mr. Swanand Malode sir for valuable suggestions and motivation. I express my deepest and special thanks to my elder Brothers Anand and Yogesh & my sister for their keen interest, love and motivation in my life.

I am thankful to all teaching and non-teaching staff of our college for valuable suggestions and motivation, which they bestowed on me right from the inception to the successful completion of the work. Im really very much grateful to Mr. M.D.Wandhare, for his tips, his guidance and his cooperation during my dissertation work. I express my deepest and special thanks to my colleagues,Prashant, Girish, Ketan, Mayur, Lukesh, Anant, Pankaj, Ghanashyam, Dhanraj, Amol, Rahul, Sushil, Nikita, Radhe, Anish, Akanksha, Nilakshi, for their keen interest, love and support in my dissertation work. There are many others whose names flashed across my mind when I enlist those who have given grateful support to me. It would rather impracticable to mention each of them separately but I am conscious my obligation and thanks them collectively.

Leeladhar Prajapati

Table of Contents
SR. S.NO. TITLE PAGE NO.

1.

INTRODUCTION

1-28 29-35

2.

LITERATURE REVIEW

3.

AIM AND OBJECTIVES

36 37 38-44

4.

PLAN OF WORK

5.

DRUG PROFILE

6.

LIST OF MATERIAL AND EQUIPMENT

45-47

7.

EXPERIMENTAL WORK

48-59 60-82 83-85

8.

RESULTS AND DISCUSSION

9.

SUMMARY AND CONCLUSION

10.

FUTURE SCOPE

86 87-97 98

11.

REFERENCES

12.

ERRATA

Abstract

Abstract
The purpose of this study was to prepare lipospheres containing aceclofenac intended for topical delivery with the aim of exploiting the favorable properties of this carrier system and developing a sustained release formula to overcome the side effects resulting from aceclofenac oral administration. Lipospheres were prepared using different lipid cores (carnauba wax, bees wax, steryl alcohol) and phospholipids coats (egg phosphatidylchoine and soya phosphatidylcholine) by melt dispersion technique. Characterization of the prepared lipospheres formulation carried out through photomicroscopy, scanning electron microscopy (SEM), particle size analysis, diffential scanning caorimetry (DSC), and In vitro drug release and stability study. It was uniformly dispersed after suitably gelled by Carbopol 940 preparation. The characterization of the prepared lipospheres was based on topical gel rheological study, pH, spreadability, drug content, skin irritation test. No oedema and erythema were observed after administration of lipospheres based aceclofenac gel on rabbit skin. The anti-inflammatory effect of liposphere systems was assessed by the rat paw edema technique and was compared to the marketed product. Results revealed that liposphere systems were able to entrap aceclofenac at very high levels (101.2%). The particle size of liposphere systems was well suited for topical drug delivery. DSC revealed the molecular dispersion of aceclofenac when incorporated in lipospheres. Lipospheres were substantially stable after 3 months storage at 28 C. Liposphere topical gel was found to possess superior anti-inflammatory activity compared to the marketed product.

Key words: Aceclofenac, entrapment efficiency, formulation of topical gel, animal experiment, stability, sustained release.
SNIOP, PUSAD 2012-2013

Chapter No.1

Introduction

1. Introduction
One approach for increasing the beneficial action of drugs and decreasing systemic adverse effects is to deliver the necessary amount of drugs to the diseased sites, where they are most needed, for the appropriate period of time1-3. Although the drug delivery system concept is not new, great progress has recently been made in the treatment of a variety of diseases. Particulate carriers (e.g., polymeric nano- and Microparticles, fat emulsion, and liposomes) possess specific advantages and disadvantages. For instance, in the case of polymeric Microparticles, the degradation of the polymer might possibly cause systemic toxic effects through the impairment of the reticuloendothelial system4 or by accumulation at the injection Site5, cytotoxic effects have indeed been observed in vitro after phagocytosis of particles by human macrophages and granulocytes6. In addition, organic solvent residues deriving from the preparation procedures, such as the solvent evaporation technique often used for liposome7 and polyester microparticles8 can be present in the delivery system and could result in severe acceptability and toxicity problems9. To solve these adverse effects, lipid microspheres, often called lipospheres (LS), have been proposed as a new type of fat-based encapsulation system for drug delivery of bioactive compounds (especially lipophilic compounds). LS consist of solid microparticles with a mean diameter usually between 0.2 and 500 m, composed of a solid hydrophobic fat matrix in which the bioactive compounds are dissolved or dispersed10-12. LS have some advantages over other delivery systems, such as good physical stability, low cost of ingredients, ease of preparation and scale-up, and high entrapment yields for hydrophobic drugs. Because of their large range in particle size, LS can be administered by different routes- such as orally, subcutaneously, intramuscularly, or topically-or they can be used in cell encapsulation, thus allowing them to be proposed for treatment of a number of diseases13 15. For instance, the in vivo distribution of LS demonstrated a high affinity to vascular wells (including capillaries),
SNIOP, PUSAD 2012-2013 Page 1

Chapter No.1

Introduction

inflamed tissues, and granulocytes16-17. LS have been used for the controlled delivery of various types of drugs, including vasodilator and antiplatelet drugs, anti-inflammatory compounds, local anesthetics, antibiotics, and anticancer agents; they have also been used successfully as carriers of vaccines and adjuvants18. For a biocompatible formulation suitable for human administration, triglycerides and monoglycerides have been chosen as the biomaterials for LS because of their high biocompatibility, high physicochemical stability, and drug delivery release. LS were prepared by two alternative approaches, namely, the melt dispersion and the solvent evaporation techniques. Liposphere formulation is an aqueous micro dispersion of solid water insoluble spherical micro the lipospheres are made of solid hydrophobic triglycerides with a monolayer of phospholipids embedded on the surface of the particle. Liposphere formulation is appropriate for oral, parenteral and topical drug delivery system. The solid core containing a drug dissolved or dispersed in a solid fat matrix and used as carrier for hydrophobic drugs. Several techniques, such as solvent emulsification evaporation, hot and cold homogenization and high pressure homogenization have been used for the production of liposphere. Benefits of liposphere drug delivery system are; a) Improving drug stability. b) Possibility for controlled drug release. c) Controlled particle size. d) High drug loading. In addition, use of lipospheres for oral administration, it can protect the drug from hydrolysis, as well as improve drug bioavailability. Therefore, the present review articles focused on achievements of lipospheres formulation to deliver the drugs in the targeted sites. Polymers as carriers19 for difficult to deliver drugs, to be targeted drugs and to achieve a desired release pattern is a popularly known and widely exploited concept in formulation Technology. With the emergence of polymer
SNIOP, PUSAD 2012-2013 Page 2

Chapter No.1

Introduction

era, delivery systems like microspheres20 nanoparticles21 found their way into pharmaceutical market. Natural, semi synthetic and synthetic polymers are now being widely explored in the formulation of multiparticulate systems apart from several other applications. Multi particulate systems comprise of several small discrete units containing drug substance entrapped or encapsulated in polymer and offer several advantages over single unit dosage forms. They show predictable gastric emptying resulting in less inter and intra-subject variability, gastric emptying independent of the state of nutrition, high degree of dispersion in the digestive tract, lower risk of dose dumping and reduced local irritation, increased bioavailability, reduced risk of systemic toxicity. Despite these advantages, use of polymers in drug delivery poses several safety concerns. Polymers used in the preparation of micro/nanospheres can produce toxic degradation products causing systemic toxic effects through the impairment of reticuloendothelial system (RES). Polymers precipitate toxic effects due to accumulation of products at injection site. In vitro studies have shown cytotoxic effects after phagocytosis of polymer particles by human macrophages and granulocytes. Eg: Pre degraded poly (Llactic acid) (P-PLLA) and non treated PLLA (N-PLLA) particles, both having diameters not exceeding 38 m, were injected intraperitoneally in mice. Nondegradable polytetrafluoroethylene (PTFE) particles and the carrier solution were used as control. Cells of the abdominal cavity were harvested after 1, 2, 3, 4, 5, and 7 days to study the effect on the morphology of cells and viability due to degradation products. TEM (Transmission Electron Microscopy) studies indicated that, upon injection of particles in the peritoneal cavity, macrophages demonstrated signs of cell damage, cell death, and cell lysis due to phagocytosis of a large amount of PPLLA particles. Methods like solvent evaporation used in the preparation of liposomes and polyester Microparticles
22

leave organic solvent residues in the product which

can result in severe acceptability and toxicity problems.


SNIOP, PUSAD 2012-2013 Page 3

Chapter No.1

Introduction

Due to several limitations with polymeric delivery systems, extensive attempts are being made to develop alternate carriers. Lipids23 especially, are now being studied widely due to their attractive properties namely physicochemical diversity, biocompatibility, biodegradability, ability to increase the oral bioavailability of poorly water soluble drug moieties, thus making them ideal candidates as carriers for problematic drugs.

1.1.1 Advantages of lipid based delivery systems:


Lipid based delivery systems disperse, solubilize and maintain solubility of drug in GI fluids. Bioavailability of most of the lipophilic drugs is altered in the presence of lipid content in food. Lipid carriers mimic such lipid food and thus reduce the food effect on bioavailability of drugs and render flexibility to dosage regimen. Transfer drug into bile-salt mixed micelle and promote lymphatic uptake of carrierdrug particles. Influence gut wall permeability. Normalize and/or modify pharmacokinetic parameters. However few concerns related to using lipids as carriers can be overcome by well characterizing physicochemical and testing methodologies for lipid drug delivery systems and are as follows: Limiting solubility of drug in lipid core which determines entrapment efficiency. Quantity of excipient required. Stability of drug. Chemical stability issues like drug and carrier compatibility. Physical stability of lipid dosage forms like polymorphic phase transitions of drug and lipid during preparation and storage and stability of semisolids.

SNIOP, PUSAD

2012-2013

Page 4

Chapter No.1

Introduction

Also research should be focused towards developing reliable in vitro and in vivo testing methodologies for lipid based delivery systems and understanding the in vivo fate of drug carried by such delivery systems and influence of co administered drugs/lipids on the bioavailability of drugs. Lipid based drug delivery systems like solid lipid nanoparticles (a technology owned by Skye Pharma) and lipospheres are now being studied widely. Solid lipid nanoparticles24 are nanosized lipid carriers in which lipidic core contain the drug in dissolved or dispersed state. These systems were designed to substitute polymeric carriers due to the inherent toxicity. Lipospheres are lipid based dispersion systems in which drug is dissolved or dispersed in lipidic core, the surface of which is embedded with emulsifier layer.

1.1.2 Advantages of lipospheres carriers:


1. Easily available, low cost, GRAS (Generally Recognized As Safe) listed raw materials. 2. Feasible simple production techniques that do not employ high energy process like homogenization which will otherwise compromise the stability of labile active pharmaceutical ingredient. 3. High entrapment for lipophilic drugs. 4. Extended release of entrapped drug after a single injection from few hours to several days 5. Good physical stability 6. Administration by several routes.

1.2. Colloidal drug delivery systems


Many of the drug substances are characterized by poor aqueous solubility, which causes many formulation problems. Besides the use of cosolvents, drug24 complexation and solubilization in surfactant micelles, incorporation in colloidal carrier systems represents an alternative way to
SNIOP, PUSAD 2012-2013 Page 5

Chapter No.1

Introduction

render poorly water soluble drugs applicable for effective therapy. Furthermore incorporation of drugs in particulate carriers provides a possibility to manipulate the drug release. In last few years the colloidal carriers have been used for site specific targeting especially in cancer chemotherapy. Based on the carrier material the conventional vehicles used as drug carriers can be divided into 2 groups 1. 2. a. b. c. d. Polymeric carriers. Lipidic carriers. Liposomes. Lipoproteins. Lipid O/W emulsions. Lipospheres. The lipidic carriers are more preferred than polymeric carriers to avoid potential toxicological problems. The vehicles of all the above lipidic carriers are composed of physiological lipids such as phospholipids, cholesterol, cholesterol esters and triglycerides.

Phospholipid molecule

SNIOP, PUSAD

2012-2013

Page 6

Chapter No.1

Introduction

Micelle

Lipid Bilayer

Lipospheres

Figure 1: Interactions of Phospholipids in Aqueous Media.

Lipospheres

Figure 2: Structure of Liposphere

Figure 3: Schematic Liposphere

1.3. The structure of skin


Skin is anatomically divided into three principal and distinct layers, from the outside of skin inward, including stratum corneum (1020 m thick), viable

SNIOP, PUSAD

2012-2013

Page 7

Chapter No.1

Introduction

epidermis (50100 m thick), and dermis (1000-2000 m thick). A fatty subcutaneous layer resides beneath the dermis. It should be pointed out that all the thickness specified here are representative only, since the actual thickness of each layer varies several fold from place to place on the body. Adnexal appendages, including hair follicles, associated sebaceous glands and pili muscles, apocrine and eccrine sweat glands, can be found dispersing throughout of the skin, varying in number and size depending on body site. The cross section of skin structure is shown is (Figure 4)

Fig 4: A cross sectional view of human skin, Source: From Ref. (Lu and Flynn, 2009)

1.3.1 Stratum corneum (SC)


Stratum corneum is the outmost superficial layer of the skin and also the principal barrier element of the skin. SC consists of several layers of completely keratinized flattened dead cells, corneocytes, each of which is about 30 m in diameter with a hexagonal shape and 0.5-0.8 m in thickness. These acutely flattened corneocytes are highly organized and stacked vertically 15 to 25 cell layers, which are embedded into a specialized and well structured intercellular lipid matrix.

SNIOP, PUSAD

2012-2013

Page 8

Chapter No.1

Introduction

The most simplistic organizational description of SC is advocated by which is the classic brick-and-mortar assembly (Figure 5). The intracellular space of corneocytes is literally packed with structural protein, semi crystal line keratin intermixed with more amorphous keratin. The intracellular space is dense, offering little freedom of movement to drug molecules. Thus, the corneocytes work as brick being thermodynamically impenetrable. While the intercellular space of corneocytes is filled with a lipid mortar formed of cholesterol, free fatty acids, and ceramides, which seals horny structure.

Fig.5: Brick-and-mortar model of Stratum corneum and penetration routes through it, Source from Ref. (Elias, 1981)

However, the brick-and-mortar skin model is not enough to describe the panorama of the SC. In fact, the cells from basal layer of epidermis, which we describe further in the text, to the SC are built up in clusters, which represent the basic skin permeation resistance unit.It is these clusters that are separated by surface corrugations (wrinkle line), which often reach several micrometers into the basal layer of the epidermis (Figure 6).

Fig.6: Epidermis with Stratum corneum including a corneocytes cluster, Source from Ref. (Cevc and Vierl, 2010)

SNIOP, PUSAD

2012-2013

Page 9

Chapter No.1

Introduction

In addition, the basolateral side of stratum corneum is in direct contact with the living epidermal mass, where the corneocytes contain water at high thermodynamic activity of the physiological milieu. On the other hand, its external surface interfaces the environment, where air tends to have a far lower water activity. Consequently a water gradient is established and water diffuses out through the stratum corneum. Under such a normal hydration situation, the stratum corneum takes up moisture to the extent of 15% to 20% of its dry weight. It should be pointed out here that the hydration condition of SC plays an important role on the drug molecules skin penetration, which would be discussed further in the text.

1.3.2 Viable epidermis


The viable epidermis is underlying the stratum corneum. It is multilayered when viewed under microscope, including, from bottom to top, basal layer (stratum germinativum), spinous layer (stratum spinosum), granular layer (stratum granulosum) and lucid layer (stratum lucidum). Each layer is defined by position, shape, and morphology and also reflects the progressive differentiation of keratinocytes which eventuates into their death and placement as chemically and physically resistant brick in stratum corneum. However, when physicochemically considered, the viable epidermis is just a group of tightly massed live cells, which results in a singular diffusion area or resistance in percutaneous absorption process. Water found in this live epidermis has an activity equivalent to that of 0.9 % NaCl. While the interface between stratum corneum and epidermis is flat, the one between epidermis and dermis is papillose, which increases their contact surface area and then allows for the diffusion of nutrients or other biological or medicated molecules between dermis and epidermis.

SNIOP, PUSAD

2012-2013

Page 10

Chapter No.1

Introduction

The epidermis itself is avascular. Besides keratinocytes, Langerhans cells also can be found in viable epidermis. They are antigen presenting cells in the skins immunological responses. Moreover, another kind of cells, melanocytes, are strategically placed in the epidermis just above the epidermis and dermis junction. When influenced by melanocyte-stimulating hormone or ultraviolet radiation, melanocytes synthesize and deposit the pigment granules into skin, which gives rise to the skin coloration.

1.3.3 Dermis
Dermis is directly adjacent to the epidermis and extends from the epidermal-dermal junction to the subcutaneous tissue (Figure 4). Dermis consists of a net-work of irregular connective tissue, which provides the mechanical support for the skin. The matrix of this connective tissue consists of structure fibers, such as collagen, reticulum, and elastin. These fibers are embedded in an amorphous mucopolysaccharidic gel called the ground substance. The dermis can be arbitrarily divided in into a superficial papillary layer and a deep reticular layer. The upper papillary layer is thin, one fifth of thickness of the dermis, and protrudes in to the epidermis giving rise to the dermal papilla, and also provides the support of the delicate capillary plexus which nurtures the epidermis. The deepest layer of the skin is a far coarser fibrous matrix, the reticular dermis, which is the main structural element of the skin. Equally importantly, the microcirculation which subserves the skin is entirely housed in the dermis. Blood flow through skin can vary by a factor of 100 fold depending on environmental conditions. The dermis is also penetrated by sensory nerve endings and an extensive lymphatic network. Moreover, skin appendages such as sweat gland, sebaceous glands, hair follicles, and arrector pili muscles are anchored within the dermis.
SNIOP, PUSAD 2012-2013 Page 11

Chapter No.1

Introduction

The main cell inhabitants of the dermis are fibroblasts, mast cells and macrophages. Fibroblasts synthesize the structural fibers, while mast cells are thought to synthesize the ground substance. Macrophages work as immune response. In addition, plasma cells, chromatophores, fat cells, nerve cells and endings can also be found along with blood vessels, nerves and lymphatics.

1.3.4 Skin appendages


Skin appendages include hair follicles and their associated sebaceous glands, eccrine glands, apocrine glands, and arrector pili muscles. The hair follicle unit is composed of the hair, hair follicle, associated sebaceous gland, and pili muscles. Hair is a compact of keratinized structures, which consist of three layers, including an outermost cuticle, a cortex of densely packed keratinized cells, and a medulla of loose flattened cells. Hairs can be found mostly everywhere on the body except for the soles of the feet, the palms of the hand, and mucocutaneous junctions. There are100 follicles per square centimeter, representing one thousandth of the skins surface. A hair emerges from a follicle, which is set within the dermis at a slight angle. The hair follicle consists of three major components, including internal root sheath, external root sheath, and dermal papilla (Figure 7). This arrangement results in a solid implantation of the hair root in the hair follicle. In addition, each follicle is anchored to the surrounding connective tissue by an individual strand of arrector pili muscle. Furthermore, each follicle is associated with one or more flask like sebaceous glands, which secret an oily secretion, sebum. Then the sebum is forced upward around the hair shaft and onto the skin surface. Sebum mainly consists of squalene, cholesterol esters, wax esters, and triglycerides. It has several biological functions including the regulation of steroidogenesis and androgen synthesis, and providing antibacterial and water resistance to skin.

SNIOP, PUSAD

2012-2013

Page 12

Chapter No.1

Introduction

Fig.7 Skin appendages: (a) Structure of the skin (b) Structure of the hair follicle (c) Cross-section of the hair, source from Ref. (Wosicka and Cal, 2010)

Eccrine glands (sweat glands) are distributed over the entire body except the genitalia and lips. These simple tubular glands open directly on the skin surfaces and extend to the footings of the dermis. There are between 150 and 600 glands per square centimeter of body surface depending on body site .However, the estimated number of actual sweating glands is much less than that value, since many of these glands remains dormant. Thus, these glandular openings occupy approximately one ten thousandth of the skin surface. Eccrine sweat is slightly acidic (pH=5) due to traces of lactic acid, which is moderately bacteriostatic.

1.3.5 Skin penetration routes


When a skin drug delivery system is applied topically, drug-containing carriers or free drug in the system could interact with either the stratum corneum or the sebum filled ducts of the pilosebaceous glands. Thus, two principle absorption routes are involved, including the

transepidermal route, where the drug delivery system interacts with or diffuses

SNIOP, PUSAD

2012-2013

Page 13

Chapter No.1

Introduction

through stratum corneum, and transfollicular route, where they interact with or diffuse through the follicles. In the case of the transepidermal route, since the impermeable character of the corneocytes, the intercellular space of corneocytes provides the only continuous phase, which is also the predominant penetration pathway (intercellular route or intercorneocyte pathway) from the skin surface to the viable epidermis. However, the tortuous zigzag bestowed by staggered corneocytes arrangement (typically 1821), corneocyte layers as well as the highly organized crystalline lamellae structures of the mortar lead to an outstanding barrier property of the labyrinthine intercellular route. The transportation of molecules across this layer is primarily passive diffusion, in accordance with Ficks law, and no active transport processes have been identified to date.Thus the permeability of stratum corneum as a penetration resistor is proportional to the diffusive mobility of drug molecules within it (diffusion coefficient, Dsc, also proportional to the capacity of the SC to solubilize the drug molecules relative to vehicle (partition coefficient, Ksc) but inversely proportional to the thickness of stratum corneum (hsc). Consequently, at the steady state and sink condition, drug permeation can be described as following:

where Jsc (g cm-2 h-1) is the steady state flux through stratum corneum. C is the concentration of drug in the topical drug delivery system. When considering transfollicular route, initially it was not considered to be a significant skin penetration route, as evidence suggested that they accounted for only approximately 0.1% of the skin surface area .Recently, it has been demonstrated that hair follicles may act as a significant penetration pathway and/or potential
SNIOP, PUSAD 2012-2013 Page 14

Chapter No.1

Introduction

reservoirs for topically applied compound. As mentioned before, owing to the presence of sebum in follicles, the permeation through follicular route can be described as following:

Where J sebum (g cm-2 h-1) is the steady state flux through sebum/hair follicle. C is the concentration of drug in the topical drug delivery system. Ksebum and Dsebum are diffusion coefficient through sebum and drug partition coefficient in sebum/water, respectively. In short, either or both routes can be important depending on the physicochemical properties of a drug as well as the condition of the skin, since the percutaneous absorption is a spontaneous passive diffusion process which takes the path of least resistance.

1.4. Sustained drug delivery system


Modified release delivery systems may be divided conveniently into the following categories 25 Delayed release Sustained release Site specific targeting Receptor targeting Sustained release, sustained action, prolonged action, controlled release, extended action, timed release, depot and repository dosage forms are terms used to identify drug delivery systems that are designed to achieve a prolonged therapeutic effect by continuously releasing medication over an extended period of time after an administration of single dose.

SNIOP, PUSAD

2012-2013

Page 15

Chapter No.1

Introduction

The term sustained release is used to describe a dosage form formulated to retard the release of a therapeutic agent such that its appearance into systemic circulation is delayed and/or prolonged and its plasma profile is sustained in duration. The onset of pharmacological action is often delayed and duration of its therapeutic effect is often sustained. Controlled release dosage forms are designed to release drug in vivo according to predictable rates that can be verified by in vitro measurements. Controlled release technology implies a quantitative understanding of the physicochemical mechanism of drug availability to the extent that the dosage form release rate can be specified.26 Various designations such as smart, targeted, intelligent, novel and therapeutic have been given to sustained release systems. The sustained release dosage forms continue to lure both the market and the researchers by virtue of improved patient compliance and reduced incidences of adverse drug reactions. The field of sustained release technology is vastly growing and as a consequence has witnessed a remarkable sophistication. Many new technologies and devices are continuously being tried for providing a more reliable control and precision over the release of the actives.27

1.4.1 Rationale of sustained drug delivery systems:


In general, the goal of sustained release dosage form is to maintain therapeutic blood or tissue level of the drug for extended period of time. This is generally accomplished by attempting to obtain zero order release from the dosage form. Zero order release constitutes drug release from the dosage form which is independent of the amount of drug in the delivery system. Sustained release system generally do not attain this type of release and usually try to mimic zero order release by providing drug in slow first order fashion (i.e.

SNIOP, PUSAD

2012-2013

Page 16

Chapter No.1

Introduction

concentration dependent). Thus sustained release dosage form consists of two parts: An immediately available dose to establish the blood level quickly in an amount sufficient to produce the desired pharmacological response (i.e. Loading dose). The remaining amount of total dose (maintenance dose) is then gradually released to maintain constant blood level of the drug. 28

Figure 8: Plasma drug concentration profiles for conventional tablet or capsule


T

formulation, a sustained release formulation and a I zero order controlled release formulation.
M E

1.4.2. Factors influencing the design and performance of sustained release products:
To establish criteria for the design of controlled release products a number of variables must be considered such as:

SNIOP, PUSAD

2012-2013

Page 17

Chapter No.1

Introduction

1.4.2.1. Drug properties:


Physicochemical properties of drug including stability, solubility, partitioning characteristics, charge and protein drug binding play a dominant role in the design and performance of controlled release systems.

1.4.2.2. Route of drug delivery:


Physiological constraints imposed by particular route, such as, first pass metabolism, GI motility, blood supply and sequestration of small foreign particles by the liver and spleen. 1. Target sites. 2. Acute or chronic therapy. 3. The disease. 4. The patient29. 1.4.3.3. Advantages of sustained drug delivery systems over conventional dosage forms Improved patient compliance and convenience due to less frequent drug administration. Reduction in fluctuations in steady state levels and therefore better control of disease condition and reduced intensity of local or systemic side effects. Increased safety margin of high potency drugs due to better control of plasma levels. Maximum utilization of drug enabling reduction in total amount of dose administered.

SNIOP, PUSAD

2012-2013

Page 18

Chapter No.1

Introduction

Reduction in health care costs through improved therapy, shorter treatment period, less frequency of dosing, reduction in personal time to dispense, administer and monitor patients. Improved bioavailability of some drugs. 30

1.5. Formulation:
Lipospheres are generally composed of: 1.5.1. Lipid core which is a combination of different lipids (fats, oils):
Table 1: Composition and Active ingredients for formulations of lipospheres

Witepsol W35, Witepsol H35; Compritol 888 ATO Triglycerides (Glyceryl behenate); Dynasan 112; Precirol ( Glyceryl palmito stearate); tricaprin, trilaurin, tripalmitin,

tristearin, trimyristin. Monounsaturated fatty Cis forms of monounsaturated fatty acids have lower acid melting point than triglycerides hence used as a mixture with higher saturated fatty esters Partially hydrogenated Soybean oil, coconut oil, cotton seed oil. vegetable oils Oils Olive oil, wheat germ oil, evenin primrose oil, arachis oil, safflower oil, corn oil, rice bran oil. Waxes Bees wax, spermaceti, cetyl palmitate, arachidyl oleate, carnauba wax, cetyl alcohol, cholesteryl butyrate

1.5.2. Active pharmaceutical ingredient 1.5.3. Emulsifiers:


Phospholipids pure-egg phosphatidyglycerol, phosphatidylethanolamine, dimyristoyl phosphatidylglycerol, soybean phosphatidylcholine Surfactants: Tween-80, butyl alcohol

SNIOP, PUSAD

2012-2013

Page 19

Chapter No.1

Introduction

1.5.4. Stabilizers:
Gelatin, pectin, carrageenan, polyvinyl alcohol, polyoxyethylene sorbitan trioleate, Pluronic PE 8100, lauryl sarcosine.

1.6. Methods of preparation:


1.6.1 Melt dispersion technique 31 In this method, drug is dissolved or dispersed in the melted lipidic phase (figure 9). Aqueous phase is composed of water or suitable buffer which is heated to the same temperature as lipid phase. The aqueous phase is kept under stirring during which emulsifier is added. To the aqueous phase containing emulsifier, lipid phase containing drug is added drop by drop while maintaining the temperature and stirring speed. After this hot emulsification phase, the temperature of the mixture is rapidly brought down to room temperature or below room temperature by adding ice cold water or ice under continuous stirring. This cold resolidification results in the formation of discrete lipospheres which can be filtered. Several drugs like bupivacaine, glypizide , aceclofenac , retinyl acetate, progesterone,sodium cromglycate, diclofenac , carbamazepine , C14-diazepam, proteins like somatostatin , thymocartin , casein , bovine serum albumin , R32NS1 malaria antigen , tripalmitin based lipospheres for labon-chip

applications have been prepared by melt dispersion methods. Lipids carrying antigens exert their adjuvant effect to immunogenicity of antigens and the effect was found to decrease in the following order for the lipids studied: ethyl stearate>olive oil>tristearin>tricaprin>corn oil>stearic acid. Also inclusion of negatively charged lipids like dimyristoyl phosphotidyglycerol in the lipid core was found to improve the antibody response to encapsulated malaria antigen.

SNIOP, PUSAD

2012-2013

Page 20

Chapter No.1

Introduction

1.6.2 Solvent evaporation method 31 In this method, lipid is dissolved in an organic solvent. Commonly used organic solvents include ethyl acetate, ethanol, acetone or dichloromethane. This lipid phase is emulsified into aqueous phase containing emulsifier. Organic solvent is evaporated by stirring the oil in water emulsion for 6-8 h under ambient conditions. Discrete lipospheres can be collected by filtration through paper filter after the water rises to the surface. Examples of the drugs formulated as lipospheres by this method include paclitaxel, thymocartin, bovine serum albumin, triptorelin leuprolide .

Fig 9: Schematic representation of the methods of production of LS: melt dispersion and solvent evaporation

SNIOP, PUSAD

2012-2013

Page 21

Chapter No.1

Introduction

1.6.3 Co-solvent solvent evaporation method 31 In this co-solvent - solvent evaporation method employing chloroform and Nmethyl pyrollidone to create a clear solution, although low yield and large particle size is obtained, which is altered by variation in the solvent used. Lipospheres made up of polar and non-polar lipids using synthetic stabilizers instead of phospholipids which are the deviation from the definition of liposphere reported by Domb in his patent. Although their work is not related to protein delivery but they tried it with hydrophilic drug and reported around 50% entrapment by double emulsification method 1.6.4 Sonication method 32 In this technique, the drug is mixed with lipid in a scintillation vial which is precoated with phospholipids. The vial is heated until the lipid melts, and then vortexed for 2min to ensure proper mixing of the ingredients. A 10 ml of hot buffer solution is added into the above mixture and sonicated for 10min with intermittent cooling until it reaches to the room temperature. 1.6.5 Rotoevaporation method 32 In this technique, lipid solution with drug is prepared in a round bottom flask containing 100 grams of glass beads (3 mm in diameter) mixed thoroughly till a clear solution is obtained. Then, the solvent is evaporated by using rotoevaporizer under reduced pressure at room temperature and a thin film is formed around the round bottom flask and the glass beads. Raise the temperature upto 40 C until complete evaporation of the organic solvent. Known amount of 0.9 % saline is added to the round bottom flask and the contents are mixed for 30 min at room temperature and then the temperature is lowered to 10 C by

SNIOP, PUSAD

2012-2013

Page 22

Chapter No.1

Introduction

placing in ice bath and mixing is continued for another 30min until lipospheres are formed.
32

1.6.6. Microfluidizer method

Lipospheres can also be prepared by using a microfluidizer which is equipped with two separate entry ports. From one entry port, a homogenous melted solution or suspension of drug and carrier is pumped and from second entry port, an aqueous buffer is pumped. The liquids are mixed in the instrument at elevated temperatures where the carrier is melted and rapidly cooled to form the lipospheres. The temperature of the microfluidizer can also be changed at any stage of the lipospheres processing to manipulate the particle size and distribution.
32

1.6.7 Polymeric lipospheres

Polymeric biodegradable lipospheres can also be prepared by solvent or melt processes. The difference between polymeric lipospheres and the standard liposphere formulations is the composition of the internal core of the particles. Standard lipospheres, as those previously described, consist of a solid hydrophobic fat core that is composed of neutral fats like tristearin, while in the polymeric lipospheres, biodegradable polymers such as polylactide (PLD) or PCL substitute the triglycerides. Both types of lipospheres are thought to be stabilized by one layer of phospholipid molecules embedded in their surface. 1.6.8 Microemulsion 32 In this method, drug is added to the melted lipid. Aqueous phase is prepared by adding surfactant like Tween 80 into water maintained at same temperature as lipid phase. This is followed by the addition of co-surfactant like butyl alcohol to the aqueous phase. The aqueous phase containing surfactant and co-surfactant is

SNIOP, PUSAD

2012-2013

Page 23

Chapter No.1

Introduction

added to lipid phase kept under stirring. Rapid cooling of the above mixture results on formation of discrete lipid particles. Flurbiprofen lipospheres prepared by this method. Presence of Tween80 at 2%, butyl alcohol at 2ml and water at 50ml found to give discrete lipospheres of superior quality. 1.6.9 Multiple Emulsions 32 In this method, drug solution (aqueous phase) is added to melted lipid. The primary emulsion formed as a result is added into aqueous solution containing emulsifier kept at the same temperature as primary emulsion. The multiple emulsions formed as a result is subjected to rapid cooling to form lipospheres. Morel et al reported a 90 % entrapment efficiency of D-Trp-6- LHRH peptide from stearic acid-egg lecithin based lipospheres prepared by this technique. Drugs like thymopentin, cyclosporine and peptides like papain were investigated for liposphere formulations by this method.

SNIOP, PUSAD

2012-2013

Page 24

Chapter No.1

Introduction

1.7. Factors influencing quality attributes of lipospheres 1.7.1. Factors influencing morphology of lipospheres
Table 2: Factors influencing morphology of lipospheres

S.No.

Factors

Influence Proportion of larger particles formed was high on

1.

Drug loading

increasing the drug amount. At maximum drug: lipid (1:1)33 insufficient coating of drug by lipid leads to the formation of aggregates during cooling phase resulting in irregular, fluffy and fragile particles. Combination of apolar (tristearin, tripalmitin or tribehenin)

2.

Type of lipid

with

polar

lipids

(glycery

monostearate,

glyceryl

monooleate) gave lipospheres satisfactory in terms of size, shape and recovery. Lipospheres were produced using different impeller types
33

3.

Type of impeller

and particle characteristics of formed lipospheres were

studied. Impellers used were of rotor (2-blade, 3-blade) type, helicoidal rotor (4-blade) type, double truncated cone rotor. Lipospheres could not be produced using 2-blade rotor and resulted in the formation of elliptical particles.

SNIOP, PUSAD

2012-2013

Page 25

Chapter No.1

Introduction

1.7.2. Factors influencing entrapment efficiency


Table 3: Factors influencing entrapment efficiency

S.No.

Factors

Influence Hydrophobicity of lipids promotes entrapment of

1.

Type of lipid

drugs.

Long

chain

triglycerides

(tristearin

and

triarachidin) are generally more hydrophobic than short chain triglycerides like tricaprin and trilaurin.

Accordingly the free drug contents of formulations containing the long chain triglycerides were found to be lower than short chain triglycerides34. Also long chain triglycerides were found to increase the bioavailability of drug as they increase in

gastrointestinal residence time of drug compared to medium chain and short chain fatty acids35 Lipid excipients reduce the activity of P-glycoprotein and MDR (multi drug resistant) associated protein 2 by down regulating the protein expression and increase in cell membrane permeability in addition to lymphatic uptake. As the phospholipid (coat) amount increases, formation 2. Amount of Phospholipid of alternative systems like liposomes was observed which will compromise drug entrapment. Experiments with triglyceride, phospholipid at a 1:0.5 to 1: 0.25 w/w
36

revealed that 70-90% of phospholipid polar

heads were accessible on liposphere surface thus enhancing the loadability of drug.

SNIOP, PUSAD

2012-2013

Page 26

Chapter No.1

Introduction

Melt dispersion method was found to be superior over 3. Effect of method of preparation solvent evaporation method in terms of entrapment efficiency as melt method promotes drug incorporation core where as solvent evaporation promotes drug incorporation in coat.

1.7.3. Factors influencing drug release


Table 4: Factors influencing drug release

S.No. Factor

Influences The release mechanism of drugs namely tetracaine, etomidate an prednisolone37 entrapped in lipid particles. Dynasan 112 (glycerol trilaurate), Compritol 888 ATO (glycerol behenate) were used as lipid carriers and Pluronic F 68 (Poloxamer 188), Lipoid S 75 (soy lecithin), Lipoid KG were used as emulsifiers. Tetracaine and etomidate lipospheres have shown burst release and prednisolone lipospheres gave prolonged release.

1.

Release pattern

Effect of Smaller particles have larger surface area exposed to 2. particle size dissolution medium and higher diffusion coefficient. If the drug resides in the outer shell diffusion distance becomes shorter resulting in fast (burst) release. Highest T8h value was obtained with stearyl alcohol 3. Type of lipid lipospheres compared to fatty acids like stearic acid. Stearyl alcohol possesses hydroxyl groups promoting matrix hydration by providing a hydrophilic pathway for water molecules to solubilize the drug and increase in dissolution rate. Lowest T 8h value was obtained from stearic acid lipospheres because of interaction of stearic acid with metal

SNIOP, PUSAD

2012-2013

Page 27

Chapter No.1

Introduction

ions in medium forming sodium soaps which are crystals that contain fatty acid and metal carboxylate ion pairs retarding the release. Lipospheres formulated with gelatin as stabilizer released 4 Effect of 80% of total drug in 8hrs resulting in sigmoid mode of stabilizer release whereas formulations with Poloxamer 40738 resulted in a biphasic pattern (burst release followed by slow release)

1.8. Applications of liposphers


1.8.1. Parenteral route Lipospheres have been exploited for the delivery of anesthetics like lidocaine bupivacaine for the parenteral delivery of antibiotics like ofloxacin, norfloxacin, chloramphenicol palmitate and oxytetracycline , and antifungal agents, such as nystatin and amphotericin B for the adjuvants. 1.8.2. Transdermal route 39 Properties of lipospheres like film forming ability, occlusive parenteral delivery of vaccines and

properties;controlled release from solid lipid matrix resulting in prolonged release of drug and retarded systemic absorption of drugs; increasing the stability of drugs which are susceptible to extensive hepatic metabolism, make them attractive candidates for topical delivery. 1.8.3. Oral delivery 40, 41 Several categories of drugs like antibiotics, anti-inflammatory compounds, vasodilators, anticancer agents, proteins and peptides are being formulated as oral lipospheres.

SNIOP, PUSAD

2012-2013

Page 28

Chapter No.2

Literature Review

2. Literature Review
Sandipan dasgupta et al (2012)42 Nanostructured Lipid carriers (NLC)

based

gel

for

Topical

Delivery

of

aceclofenac

preparation

,characterization ,and in vivo evaluation.stearic acid as the solid lipid and oleic acid as the liquid lipid ,pluronic F68 as the sur factant and phospholipon 90G as the co-surfactant were used NLC prepared by melt emulsification and high speed homogenization methods.the antiinflammatory effect of NLC gel was assesed by rat paw edema

technique and compared to marketed aceclofenac gel. Esimone et al (2012) 43 Formulation and evaluation of goat fat and shea butter based lipospheres of benzyl penicillin lipospheres of benzyl penicillin were formulated using the conventional thin film hydration technique five different combinations of shea butter, surfactant (span 80) and goat fat were the key variables employed in the formulations the

presence of goat fat however seems to impact negatively on the in vivo stability of liposphere. Abraham J. Domb et al (2012)44 Preparation and characterization of an oral pro-dispersion liposphere formulation for cyclosporin, a water insoluble drug with limited bioavailability. Pro-dispersion formulation consisted of a solid fat, dispersing agents and amphiphilic solvents as the major components besides cyclosporin A (CsA) were prepared in the present work. For preparation of this formulation, phospholipid was dissolved in pharmaceutically acceptable water soluble organic solvent, thereafter CsA along with other components was added and formulation optimization was carried out. After formulation preparation, particle size determination and in vitro release study was carried out. Additionally,
SNIOP, PUSAD 2012-2013 Page 29

Chapter No.2

Literature Review

ultracentrifugation, TEM, Cryo-TEM and DSC techniques were used for in vitro characterization of formulation. The prepared system was also compared with marketed Neoral microemulsion formulation. Vignesh Muruganandham et al (2012)45 Formulation, Development & Characterization of Ofloxacin Microspheres Ofloxacin is anti bacterial agent that has a wide range of activity against gram (-ve) and gram (+ve) microorganisms. Multiple doses of Ofloxacin are required to attain steady state concentration. The main objective of this study was to formulate, develop and characterize Ofloxacin microspheres to prolong the release rate so as to decrease the necessity of multiple dosings especially in patients with renal impairment. The Ofloxacin

microspheres were prepared using natural polymers by non-ionic crosslinking technique. Five different formulations were prepared with respective quantities of the polymer (Chitosan) with copolymer (Gelatin and sodium alginate) with drug in different drug-polymer ratio of 1:0.5, 1:0.75 and 1:1. The prepared microspheres were evaluated for percentage drug loading, entrapment efficiency, surface morphology, and in-vitro release characteristics to identify the effect of addition of these polymers.Cumulative release data were fitted into kinetic models. The Scanning Electron Microscope analysis revealed a smooth and spherical surface morphology with mean particle size of the microspheres ranging from 7 to 14 m. Drug loading was found to increase with the increase in the concentration of encapsulating polymer, chitosan, sodium alginate and gelatin concentration. Drug release obeyed the first order kinetics45. Sanming Li et al (2012)46 Nanostructured lipid carriers (NLC)-based gel was developed as potential topical system for flurbiprofen (FP) topical
SNIOP, PUSAD 2012-2013 Page 30

Chapter No.2

Literature Review

delivery. The characterizations of the prepared semisolid formulation for topical application on skin were assessed by means of particle size distribution, zeta potential analysis, X-ray analysis, in vitro percutaneous penetration, rheological study, skin irritation test, in vivo

pharmacodynamic evaluation and in vivo pharmacokinetic study. The NLC remained within the colloidal range and it was uniformly dispersed after suitably gelled by carbopol preparation. It was indicated in vitro permeation studies through rat skin that FP-NLC-gel had a more pronounced permeation profile compared with that of FP loaded mcommon gel. Pseudoplastic flows with thixotropy were obtained for all NLC-gels after storage at three different temperatures. No oedema and erythema were observed after administration of FPNLC- gel on the rabbit skin, and the ovalbumin induced rat paw edema could be inhibited by the gel.

Satheesh Babu et al (2011)47 Manufacturing techniques of lipospheres Lipid microspheres, often called lipospheres (LS), have been proposed as new type of lipid-based encapsulation system for drug delivery of bioactive compounds especially lipophilic compounds. LS consist of solid microparticles with a mean diameter usually the size range between 0.2 to 500m, composed of a solid hydrophobic fat matrix, where the bioactive compound(s) is dissolved or dispersed. The lipospheres have several advantages over other colloidal delivery systems (including nano & micro emulsions, nanaoparticles, hydrogels and liposomes). Loganathan Veerappan et al (2010)48 formulation development and evaluation of flurbiprofen liposphere microencapsulation is a rapidly expanding technology in the production of controlled release dosage
SNIOP, PUSAD 2012-2013 Page 31

Chapter No.2

Literature Review

forms. Most of the non-steroidal antiinflammatory drugs (Nsaid) have been widely used for the treatment of acute and chronic arthritic conditions. flurbiprofen appears to be more active as an antiinflammatory agent than other nsaid products and is usually well tolerated. flurbiprofen is frequently prescribed for the treatment of rheumatoid arthritis, osteoarthritis and ankylosing spondylitis. By formulating sustained release dosage form of this drug leads to minimization of damage to the gastro intestinal mucosa. Development of formulation was made with different formulation variables and suitable formulation was selected for further evaluations. Kamal Dua et al (2010)49 Aceclofenac is a new generation non-steroidal anti-inflammatory drug showing effective anti-inflammatory and analgesic properties. It is available in the form of tablets of 100 mg. Importance of aceclofenac as a NSAID has inspired development of topical dosage forms. This mode of administration may help avoid typical side effects associated with oral administration of NSAIDs, which have led to its withdrawal. Furthermore, aceclofenac topical dosage forms can be used as a supplement to oral therapy for better treatment of conditions such as arthritis. Ointments, creams, and gels containing 1 % (m/m) aceclofenac have been prepared. They were tested for physical appearance, pH, spreadability, extrudability, drug content uniformity, in vitro diffusion and in vitro permeation. Gels prepared using Carbopol 940 (AF2, AF3) and macrogol bases (AF7) were selected after the analysis of the results. They were evaluated for acute skin irritancy, anti-inflammatory and analgesic effects using the carrageenan-induced thermal hyperalgesia and paw edema method.

SNIOP, PUSAD

2012-2013

Page 32

Chapter No.2

Literature Review

Maha Nasr et al (2008)50 liposphere as a carrier for topical Delivery of Aceclofenac preparation characterization and in vivo. The aim of exploiting the favorable properties of this carrier system and developing a sustained release formula to overcome the side effects resulting from aceclofenac oral administration. Lipospheres were prepared using different lipid cores and phospholipid coats adopting melt and solvent techniques. liposphere systems were found to possess superior antiinflammatory activity compared to the marketed product in both lotion and paste consistencies. Liposphere systems proved to be a promising topical system for the delivery of aceclofenac. Jia-You Fang et al (2007)51 acoustically active lipospheres (AALs) were prepared using perfluorocarbons and coconut oil as the cores of inner phase. These AALs were stabilized using coconut oil and phospholipid coatings. A lipophilic antioxidant, resveratrol, was the model drug loaded into the AALs. AALs with various percentages of perfluorocarbons and oil were prepared to examine their

physicochemical and drug release properties. Co-emulsifiers such as Brij 98 and Pluronic F68 (PF68) were also incorporated into AALs for evaluation. AALs with high resveratrol encapsulation rates (_90%) were prepared, with a mean droplet size of 250350 nm. The AALs produced with perfluorohexane as the core material had larger particle sizes than those with perfluoropentane. Resveratrol in these systems exhibited retarded drug release in both the presence and absence of plasma in vitro; the formulations with high oil and perfluorocarbon percentages showed the lowest drug release rates. Hagalavadi Nanjappa Shivakumar et al (2007)52 Design and statistical optimization of glipizide loaded A 32 factorial design was employed to
SNIOP, PUSAD 2012-2013 Page 33

Chapter No.2

Literature Review

produce glipizide lipospheres by the emulsification phase separation technique using paraffin wax and stearic acid as retardants. The effect of critical formulation variables, namely levels of paraffin wax (X1) and proportion of stearic acid in the wax (X2) on geometric mean diameter (dg), percent encapsulation efficiency (% EE), release at the end of 12 h (rel12) and time taken for 50% of drug release (t50), were evaluated using the F-test. Mathematical models containing only the significant terms were generated for each response parameter using the multiple linear regression analysis (MLRA) and analysis of variance (ANOVA). Both formulation variables studied exerted a significant influence (p < 0.05) on the response parameters. Numerical optimization using the desirability approach was employed to develop an optimized formulation by setting constraints on the dependent and independent variables. The drug release from lipospheres followed first-order kinetics and was characterized by the Higuchi diffusion model. The optimized liposphere formulation developed was found to produce sustained anti-diabetic activity following oral administration in rats. Vandana B. Patravale et al (2007)53 to develop solid lipid nanoparticles (SLN) of tretinoin (TRE) with the help of facile and simple emulsification-solvent diffusion (ESD) technique and to evaluate the viability of an SLN based gel in improving topical delivery of TRE. The feasibility of fabricating SLN of TRE by the ESD method was successfully demonstrated in this investigation. The developed SLN were characterized for particle size, polydispersity index, entrapment efficiency of TRE and morphology. Studies were carried out to evaluate the ability of SLN in improving the photostability of TRE as compared to TRE in methanol. Encapsulation of TRE in SLN resulted in a significant improvement in its photostability in comparison to
SNIOP, PUSAD 2012-2013 Page 34

Chapter No.2

Literature Review

methanolic TRE solution and also prevented its isomerization. Furthermore, the skin irritation studies carried out on rabbits showed that SLN based TRE gel is significantly less irritating to skin as compared to marketed TRE cream and clearly indicated its potential in improving the skin tolerability of TRE. In vitro permeation studies through rat skin indicated that an SLN based TRE gel has permeation profile comparable to that of the marketed TRE cream.

SNIOP, PUSAD

2012-2013

Page 35

ChapterNo.4

Plan of Work

4. Plan of Work
Present proposed research work was planned as followsA) Literature survey. B) Selection of drug, lipid core material and coat material. C) Procurement of drug, core material and coat material. D) Preliminary study of drug, core material and coat material. E) Formulation of Liposphere. F) Evaluation of Liposphere. 1. Photo microscopic analysis. 2. Scanning Electron microscopy. 3. Particle size analysis. 4. Differential scanning Calorimetry. 5. Fourier transforms infrared spectroscopy (FTIR). 6. In vitro dissolution test. H) Formulation of lipospheres based gel. I) Evaluation of Lipospheres based gel. 1. pH. 2. Drug content. 3. Viscosity. 4. Spreadability. 5. In vitro permeation of liposphere based gel. 6. Skin-irritation testing (Draize patch test). 7. In vivo Anti-inflammatory Study of Liposphere. 8. Stability study.

SNIOP, PUSAD

2012-2013

Page 37

Chapter No. 3

Aim and Objective

3. Aim and Objectives


Aim Preparation and evaluation of liposphere based topical drug delivery system containing a NSAID drug. Objectives 1. To study formulation and evaluation of topical delivery of NSAID drug. 2. To optimize the formulation using suitable experimental technique, regarding particle size, stability, release property, surface morphology, hydrophobicity, drug entrapment efficiency etc. 3. To study in vitro release of NSAID from liposphere 4. In vivo Anti-inflammatory study of liposphere. liposphere as a carrier for

SNIOP, PUSAD

2012-2013

Page 36

Chapter No.6

List of Material and Equipment

6. List of material and equipment


Table 5: List of Materials Used

Sr. No. 1. 2.

Instrument Used (Model No.) FTIR Spectrophotometer UV1700 Spectrophotometer, double beam

Make Aligent Cary 630 ATR Shimadzu, 1700, Japan

3.

Electronic balance

Citizen scale (CY 104), Germany

4. 5.

Brookfield viscometer (Dial type) Middleboro, MA-02346, USA Differential Scanning Calorimetry (DSC 60) Mechanical Stirrer Magnetic Stirrer Intel play Qx3 microscope (200X magnification) Dissolution test apparatus Scanning Electron Microscope (JSM 6380A) Mettler Toledo, Zaventem (U.S.) Remi lab stirrer, Mumbai Remi lab stirrer, Mumbai Edmund Scientific (U.S.)

6. 7. 8.

9. 10.

Electro lab , Mumbai JOEL, Japan

SNIOP, PUSAD

2012-2013

Page 45

Chapter No.6

List of Material and Equipment

11

Centrifuge machine

Remi, Mumbai

12.

Digital pH Meter

Chemi line (CL-110)

13.

Plethysmometer

(UGO-Basile, 7140,Comerio, Italy Skylab,Mumbai

14.

Stability chamber

List of chemicals and reagents used


Aceclofenac was procured as a gift sample from Concept pharmaceutical, india. Soy phosphatidylcholine-35% was kindly gifted by perfect Biotech industries Pvt Ltd, Nagpur. LIPOVA-E120 (Egg

phosphatidylcholine) gifted by VAV Life sciences Pvt Ltd. Mumbai. All other chemicals were procured from Research lab fine Chem industries, Mumbai. They areTable 6: List of chemical used

Chemicals Absolute Ethanol Methanol Potassium chloride Sodium chloride AR grade LR grade LR grade AR grade

Grade

SNIOP, PUSAD

2012-2013

Page 46

Chapter No.6

List of Material and Equipment

Potassium dihydrogen phosphate Acetone disodium hydrogen phosphate Acetic anhydride Disodium hydrogen phosphate Carbopol 940P Triethanolamine (TEA)

AR grade LR grade AR grade AR grade AR grade AR grade AR grade

AR grade-Analytical reagent, LR grade-Laboratory reagent

SNIOP, PUSAD

2012-2013

Page 47

Chapter No.5

Drug Profile

5. Drug Profile
5.1. Drug: Aceclofenac 54,55,56,57

5.1.1. Structure:

Chemical Name

[(2, 6-dichlorophenyl)amino] phenylacetoxyacetic acid.

Molecular formula Molecular weight Melting point Description

C16H13Cl2NO4 354.18 149-153 0C A white or almost white, crystalline powder. Insoluble in water, soluble in ethanol and acetone

Mode of action Dose Plasma half-life Plasma protein binding Category

Highly selective 2

agonist

100 mg 2 to 3 h 40-50% Anti inflammatory Analgesic

SNIOP, PUSAD

2012-2013

Page 38

Chapter No.5

Drug Profile

Adverse reactions Gastrointestinal System Duodenal ulcer, gastrointestinal perforation Urinary System Interstitial nephritis Central and Peripheral Nervous System Optic neuritis Psychiatric Hallucination, Drowsiness, Confusion Skin and Appendages Epidermal necrolysis, Erythema multiforme, dermatitis Respiratory Aggravated asthma Haematological Aplastic anaemia Contraindications 1. 2. 3. 4. 5. 6. Active peptic ulceration Recurrent indigestion (relative contraindication) Care should be taken in patients on anticoagulants Care should be taken in patients with hypertension or heart failure Pregnancy and lactation History of sensitivity to aspirin or other NSAISD drugs. Drug Interactions
1. 2. 3. 4. 5. 6. 7.
SNIOP, PUSAD

Anticoagulants Alcohol and smoking Lithium Diuretics Antihypertensive drugs Diflunisal Anti-platelet agents and selective serotonin reuptake inhibitors
2012-2013 Page 39

Chapter No.5

Drug Profile

5.2. Lipid profile


5.2.1. Lecithin58 5.2.1.1. Structure:

CH2

O-CO-R1 R1 and R2 are fatty acids

CH

O-CO-R2 _ O O P O

C H2

+ OCH2CH2N(CH3)3

Non proprietary names Synonyms

Lecithin Soya lecithin, soyabean lecithin, phosphatidylcholine.

Chemical name and CAs registry no Empirical formula

Lecithin [8002-43-5]

CH2OCOR- - - CHOCOR- - - CH2OPOOOCH2CH2N (CH3)3

Functional category

Emollient; Emulsifying agent; Solubilizing agent

Description

They may vary in color from brown to light yellow, depending upon whether they are bleached or not or on the degree of purity.

SNIOP, PUSAD

2012-2013

Page 40

Chapter No.5

Drug Profile

when they are exposed to air, rapid oxidation occurs, also resulting in a dark yellow or brown color Solubility Insoluble but swells up in water and in Nacl solution forming a colloidal suspension, soluble in about 12 parts cold, absolute alcohol; soluble in chloroform, ether,

petroleum ether, in mineral oil and sparingly soluble in benzene Incompatibility Incompatible hydrolysis Regulatory status Gras listed included in the FDA inactive ingredient guide. included in nonparenteral and parenteral medicine licensed in UK Safety It is highly biocompatible and oral doses of up to 80 g daily have been given with esterase owing to

therapeutically in the treatment of Tardive dyskinesia.

SNIOP, PUSAD

2012-2013

Page 41

Chapter No.5

Drug Profile

5.2.2. Stearyl alcohol


5.2.2.1. Structure

Chemical Name Molecular formula Molecular weight Melting point Description

octadecyl alcohol or 1-octadecanol) CH3(CH2)16CH2OH 270.49 55-58C Freely soluble in chloroform and in ether; soluble in ethanol (95 per cent); practically insoluble in water

5.2.3. Bees wax


Structure

CAS No Chemical Name

8012-89-3 Bees wax

Molecular formula
SNIOP, PUSAD

C15H31COOC30H61
2012-2013 Page 42

Chapter No.5

Drug Profile

Molecular weight Melting point Description

415 62 to 64 C white,solid Insoluble in water, soluble in alcohol

Category

Cosmetic Ingredients & Chemicals

5.2.4. Carnauba wax


A coumpound is a pure substance and Carnuba is a mixture. Carnauba wax contains mainly esters of fatty acids (80-85 %), fatty alcohols (10-15 %), acids (3-6 %) and hydrocarbons (1-3 %). Specific for carnauba wax is the content of esterified fatty diols (about 20%), hydroxylated fatty acids (about 6 %) and cinnamic acid (about 10 %). Cinnamic acid, an antioxidant, may be hydroxylated or methoxylated. The major components of carnauba wax are aliphatic and aromatic esters of long-chain alcohols and acids, with smaller amounts of free fatty acids and alcohols, and resins. Another site claim common components as C30 alcohols and C26 acids.

CAS No Synonyms

8015-86-9 Carnuba;carnauba, brazil wax, carnubawax, carnaba wax, wax, carnaubawachs, carnauba wax yellow, carnauba wax flakes.

Molecular formula Molecular weight

----------

SNIOP, PUSAD

2012-2013

Page 43

Chapter No.5

Drug Profile

Melting point Description

81-86 C Soluble on warming in chloroform, in ethyl acetate and in xylene; practically insoluble in water and in ethanol

Category

Cosmetic Ingredients & Chemicals

SNIOP, PUSAD

2012-2013

Page 44

Chapter No.7

Experimental Work

7. Experimental work 7.1. Preformulation study


Identification test

7.1.1. Aceclofenac Organoleptic properties Aceclofenac was tested for organoleptic properties such as appearance, color, odor, taste, etc. 7.1.2. Melting point The melting point of the aceclofenac was determined by capillary method. 7.1.3. Solubility determination59 The approximate solubility of Pharmacopeial substance is indicated by descriptive terms in accompanying table. 7.1.4. Ultra-violet scanning The scanning of Aceclofenac was performed in 7.4 pH phosphate buffer saline and max was found to be 276 nm which was complies with the max reported in BP. 7.1.5. Fourier transforms infrared (FTIR) spectroscopy Procedure: Small quantity of aceclofenac 1 mg was placed on diamond ATR crystal then it was scanned between 4000-500 cm-1 range.

7.1.6. Lipids
7.1.6.1. Soyalecithin Saponification value60 About 35 g Potassium hydroxide was weighed, dissolved in 20 ml of water; sufficient ethanol was added (96 %) to produce 1000 ml and allowed to stand overnight. Clear liquid was poured off. About 2 gm of soya lecithin
SNIOP, PUSAD 2012-2013 Page 48

Chapter No.7

Experimental Work

was accurately weighed and taken into a 200 ml conical flask, 25 ml of ethanolic solution of KOH was added, boiled under a reflux condenser for 1 h with rotating the contents frequently. When the solution was still hot, the excess of alkali was titrated with 0.5 M Hcl using 1 ml of phenolphthalein solution as indicator. Blank determination was carried out excluding the substance being examined. The saponification value was calculated from expression Saponification value = 28.5 V/W Where, V- difference in ml between titration W- Weight in g of substance taken.

7.2. Preparation of standard calibration curve of aceclofenac


7.2.1. Preparation of phosphate buffer saline pH 7.4 About 2.38 g of disodium hydrogen phosphate (Na2Po4), 0.19 g of potassium dihydrogen phosphate (KH2PO4) and 8.0 g of sodium chloride (Nacl) was taken in volumetric flask and volume was made with water to produce 1000 ml. This solution had a pH of about 7.461. 7.2.2. Determination of max for aceclofenac Stock solutions of Aceclofenac was prepared by dissolving in 100 ml of phosphate buffer saline solution pH (7.4), solutions were further diluted and analyzed spectrophotometrically between 220 to 370 nm to determine max. 7.2.3. Preparation of standard calibration curve of aceclofenac The calibration curve was plotted within the concentration range of 2-10 g/ml. Appropriate dilutions were prepared and absorbance was measured for each solution at 276 nm since maximum absorbance was observed at this wavelength. Graph was plotted for absorbance vs. concentration.

SNIOP, PUSAD

2012-2013

Page 49

Chapter No.7

Experimental Work

y = 0.0109x

R = 0.994.

Correlation co-efficient value indicated the linear correlation between concentration and absorbance.62 Where Y is absorbance X is concentration R2 is coefficient of regression.

7.3. Formulation of liposphere by melt dispersion technique


Lipospheres (LS) were prepared by melt dispersion method. In this method different lipid materials (Carnauba wax, Bees wax and stearyl alcohol) along with the drug aceclofenac were used to form core material in the ratio 2:1 and 3:1, egg phosphatidylcholine and soy phosphatidylcholine were used as the coat material so as to give the core to coat ratio (cr/ct) of 2:1 and 3:1. The lipid core material was melted at 75 C using water bath and then added with the required amount of aceclofenac by dispersing it in the molten lipid. Separately 1000 mg of phospholipid was added in 100 ml of phosphate buffer saline, which was preheated at 75 C and the given mixture was homogenized using mechanical stirrer until a uniform dispersion was obtained. To this dispersion at 75 C the previously prepared drug was added to molten lipid at 75 C and dispersed with the help of mechanical stirrer at 4000 rpm speed. The homogenized milky solution was then rapidly cooled down to 10-20 C with continued stirring for another 5 min for formation of uniform dispersion of lipospehers63.

SNIOP, PUSAD

2012-2013

Page 50

Chapter No.7 Table 7: Formulation codes of lipospheres (F1-F12) Code Lipid core Material F1 F2 F3 F4 F5 F6 F7 F8 F9 F10 F11 F12 Stearyl alcohol Carnauba wax Bees wax Stearyl alcohol Carnauba wax Bees wax Stearyl alcohol Carnauba wax Bees wax Stearyl alcohol Carnauba wax Bees wax Lipid coat material Egg pc Egg pc Egg pc Soya pc Soya pc Soya pc Egg pc Egg pc Egg pc Soya pc Soya pc Soya pc Quantity of lipid core (mg) 2000 2000 2000 2000 2000 2000 3000 3000 3000 3000 3000 3000

Experimental Work

Quantity of lipid coat(mg) 1000 1000 1000 1000 1000 1000 1000 1000 1000 1000 1000 1000

Drug (mg) 500 500 500 500 500 500 1000 1000 1000 1000 1000 1000

Ratio (Cr/Ct) 2:1 2:1 2:1 2:1 2:1 2:1 3:1 3:1 3:1 3:1 3:1 3:1

Egg pc Egg phosphatidylcholine, soy pc-soya phosphatidylcholine Cr-core material, Ct-coat material

7.4. Evaluations of Lipospehers


7.4.1. Separation of Unentrapped Aceclofenac from the Prepared

Lipospheres Aceclofenac lipospheres were separated from free unentrapped aceclofenac by centrifugation at 20,000 rpm for 30 min. The pellets formed were washed with 10 ml phosphate buffered saline and recentrifuged again for 30 min64-66. The lipospheres were decanted and kept in the refrigerator for further investigations.

7.4.2. Determination of entrapment efficiency The entrapped drug concentration was determined by lysis of the lipospheres with absolute alcohol65-66. Accurately weighed amount of loaded lipospheres (50 mg) was dissolved in 10 ml absolute alcohol and covered well with aluminum foil to prevent evaporation. The solution was sonicated for 15 min to obtain a clear solution. An aliquot of 1 ml of this solution was added to 9 ml of absolute alcohol. The solution was sonicated for another 15 min. The concentration of aceclofenac in absolute alcohol was
SNIOP, PUSAD 2012-2013 Page 51

Chapter No.7

Experimental Work

determined spectrophotometrically at wavelength 276 nm after appropriate dilution. Each sample was analyzed in triplicate. The entrapment efficiency was calculated through the following relationship:

Entrapment efficiency percentage = Entrapped drug/ Total drug100

7.4.3. Photo microscopic analysis A drop of lipospheres preparation was placed on a slide for morphological examination under optical stereo microscope and photographed at a magnification of 200 by means of a fitted camera.

7.4.4. Particle size analysis The size of the prepared Lipospheres was measured by the optical microscopy method using a calibrated stage micrometer. It is carried out by using a compound microscope at 10 x lances. Dried lipospheres were first re-dispersed in distilled water and placed in a glass slide and the number of division of calibrated eye piece was counted by a micrometer using a stage micrometer 67.

7.4.5. Fourier transforms infrared (FTIR) spectroscopy FTIR spectra of pure Aceclofenac and the optimized liposphere formulation were recorded with a FTIR spectrophotometer. FTIR Spectroscopy: There is always a possibility of drug Lipid interaction in any formulation due to their intimate contact. The technique employed in the present study for this purpose is IR spectroscopy.IR spectroscopy is one of the most powerful analytical techniques, which offers the possibility of chemical identification.

SNIOP, PUSAD

2012-2013

Page 52

Chapter No.7

Experimental Work

7.4.6. Differential scanning calorimetry (DSC) Samples of aceclofenac, and drug loaded lipospheres of the selected formulation were submitted to DSC analysis using differential scanning calorimeter calibrated with indium. The analysis was carried out on 1 mg samples sealed in standard aluminum pans. Thermograms were obtained at a scanning rate of 10 C/min using dry nitrogen flow of (25 ml/min). Each sample was scanned between zero and 400 C.

7.4.7. Scanning electron microscopy (SEM) The detailed surface characteristics of the selected aceclofenac lipospheres formulation were observed using a scanning electron microscope. The lipospheres sample was attached to the specimen holder using a double coated adhesive tape and gold coated (~20 nm thickness) under vacuum using a sputter coater for 510 min at 40 mA and then investigated at 30 kV 68.

7.4.8. In vitro release of aceclofenac from lipospheres Prepared Lipospheres equivalent to 100 mg of aceclofenac lipospheres were accurately weighed and filled into gelatin capsules or tight in the Muslin cloth. Degassed 7.4 Phosphate buffer saline (PBS) medium (900 ml) was placed into the dissolution tester jars and the temperature was maintained at 37 0.5 C. A USP II (paddle) dissolution apparatus (Electrolab) at 100 rpm was used. Samples of 5 ml were drawn at time points of 30, 60, 120, 180, 240, 300, 360, 420 and 480 min and an equal amount of fresh dissolution medium was replaced each time. After suitable dilution, the samples withdrawn were analyzed spectrophotometrically at a wavelength 276 nm 69. Results were the mean of three runs. The amounts of drug present in the samples were calculated with the help of appropriate calibration curve constructed from reference standard. Percentage drug release was calculated.

SNIOP, PUSAD

2012-2013

Page 53

Chapter No.7

Experimental Work

7.5. Formulations of lipospheres based gel


Lipospheres based gel was prepared according to the formula (Table 9). The suitable lipospheres formulation for the topical delivery of aceclofenac was selected based on the evaluation of characteristics like: particle size, entrapment efficiency, and in vitro release. It was found that the formulation F7 and F8 were more suitable among the other formulations. Different gelling agents such as: carbopol 940 P, xanthan gum, chitosan, and hydroxypropyl methyl cellulose (HPMC) were used for the conversion of the lipospheres dispersion into the gel formulation. Based on the compatibility with the lipospheres dispersion and the ease of spreadability, carbopol was selected as the gelling agent. Appropriate quantity of carbopol 940P was soaked in water for a period of 2 h. Carbopol was then neutralized with triethanolamine (TEA) with stirring. Then specified amount of liposphere, glycerin and permeation enhancer (Mineral oil) was mixed. Solvent blend was transferred to carbopol container and agitated for additional 20 min. The dispersion was then allowed to hydrate and swell for 60 min, finally adjusted the pH with 98 % TEA until the desired pH value was approximately reached (6.8-7). During pH adjustment, the mixture was stirred gently with a spatula until homogeneous gel was formed. All the samples were allowed to equilibrate for at least 24 hours at room temperature prior to performing rheological measurements70-76.

SNIOP, PUSAD

2012-2013

Page 54

Chapter No.7 Table 8: Formulation of gel optimized Batch

Experimental Work

Ingredients (1.5%w/w)

Formulations Gels 50g F7 F8 0.5 0.75 5 q.s. --43.75 LF7 0.5 0.75 5 q.s. 0.5 43.18 LF8 0.5 0.75 5 q.s. 0.5 43.18 Blank Lipospheres gel 0.5 -5 q.s. --44.5

Carbopol 940P Lipospheres Glycerin Triethanolamine (TEA) Permeation enhancer Distilled water

0.5 0.75 5 q.s --43.7 5

7.6. Characterization of lipospheres based topical gel


7.6.1. pH pH of gel was determined using digital pH meter. About 1 g of gel was stirred in distilled water till a uniform suspension effected. The volume was made up to 40 ml and pH of the solution was measured 77. 7.6.2. Drug content78 Take 1g gel in a 100 ml of volumetric flask and dissolve with little amount of methanol and mixture was shaken till solution was affected. The volume was made up to 100 ml with methanol. The solution was filtered through Whatman filter paper (No. 41). Further dilute 5 ml to 50 ml with methanol. The absorbance of the solution was measured at 276 nm against reagent blank. 7.6.3. Viscosity79 Viscosity of the gel was determined by using Brookfield viscometer (Dial type). As the system is non-Newtonian spindle no. 4 is used. Viscosity is measured for the fixed time 2 min for 100 rpm.

SNIOP, PUSAD

2012-2013

Page 55

Chapter No.7

Experimental Work

7.6.4. Spreadability80-82 The spreadability of the gel was determined using the following technique: 0.5 g gel was placed within a circle of 1 cm diameter premarked on a glass plate over which a second glass plate was placed. A weight of 500 g was allowed to rest on the upper glass plate for 5 min. The increase in the diameter due to spreading of the gels was noted. It was calculated using formula, S = M. L / T Where, S = spreadability M = weight tied to upper slide L = length of glass slide T = time taken Shorter time interval, to cover distance of 6.5 cm, indicates better spreadability.

7.6.5. In vitro permeation of liposphere based gel83, 84


This study was carried out for the optimized batch selected, based on all above evaluation parameters. One of the batches among them was formulation prepared without permeation enhancer F8 and other was formulation prepared with permeation enhancer LF8 was studied through cellophane membrane using a fabricated dissolution testing apparatus. The dissolution medium used was artificial tear fluid freshly prepared (pH 7.4). Cellophane membrane, previously soaked overnight in the dissolution medium, was tied to one end of a specifically designed glass cylinder (open at both ends and of 5 cm diameter). A 5 ml volume of the formulation was accurately pipetted into this assembly. The cylinder was suspended in 50 ml of dissolution medium maintained at 37 C so that the membrane just touched the receptor medium surface. The dissolution medium was stirred at 50 rpm using magnetic stirrer. Aliquots, each of 1 ml volume, were

SNIOP, PUSAD

2012-2013

Page 56

Chapter No.7

Experimental Work

withdrawn at regular intervals and replaced by an equal volume of the receptor medium. The aliquots were suitablely diluted with the receptor medium and analyzed by UV-Vis spectrophotometer at 276 nm.

7.6.6. Skin irritation testing (Draize patch test)85


The lipospheres gel in comparison to marketed aceclofenac gel was evaluated by carrying out the Draize patch test on rabbits. Animal care and handling throughout the experimental procedure were performed in accordance to the CPCSEA guidelines. The experimental protocol was approved by the institutional animal ethical committee. White rabbits weighing 2.53 kg and were acclimatized before the beginning of the study. Animals were divided into four groups as follows: Group 1: No application (Control). Group 2: Marketed formulation (Audigel 1.5% w/w gel) Group 3: Lipospheres based gel containing LF7 (1.5% w/w) Group 4: Lipospheres based gel containing LF8 (1.5%, w/w) The back of the rabbits were clipped free of hair, 72 h prior to the application of the formulations. Formulations, 0.5 g, were applied on the hair free skin of rabbits by uniform spreading. Four rabbits will be use for the skin irritancy study. All the test formulation will apply on single rabbit, so total four surface areas will create on the skin of rabbit (3 cm3 cm) using hair depletion. To avoid biological variation, the study will perform on four rabbits. The respective test sample will apply on specified area for seven day and simultaneous observation for skin irritation such as redness, edema and skin rash. The results will interpret in the form of grading scale: A-no reaction; B-slight, patchy erythema; C-moderate but patchy erythema; D-moderate erythema, and E-severe erythema with or without edema.

SNIOP, PUSAD

2012-2013

Page 57

Chapter No.7

Experimental Work

7.6.7. In vivo anti-inflammatory study of lipospheres47


The anti-inflammatory activity of the selected lipospheres formulation (LF7 and LF8) applied and compared to the marketed product using the rat paw edema test. The protocol of the present work was approved by the experimental protocol was approved by the institutional animal ethical committee. Male Wistar rats (130150 g) were randomly divided into four groups of 3 rats each. Group I control application, group II for marketed formulation (AUDIGEL), group III for lipospheres based gel (LF7), group IV for lipospheres based gel (LF8) formula the volume of paw edema (milliliter) was measured in each animal using a plethysmometer to a precision of two decimal places. The rats were marked on the left hind paw just beyond the tibiotarsal junction, so that every time the paw was dipped in the electrolyte fluid column up to a fixed mark to ensure constant paw volume. The tested preparations were applied to the left hind paws of rats using an amount equivalent to 1 mg of aceclofenac. After 1 h of topical application, initial paw volume of the rats was measured by dipping the rat paw into the electrolyte column just before carrageenan injection and the increase in volume due to fluid displacement was noted from a digital display, followed by the injection of 0.1 ml of 1% (w/v) carrageenan solution in saline in the subplantar region of left hind paw of the rats. Measurement of paw volume was done after 1, 2, 3, 4, 5, 6, 7 and 8 h. The edema rate and inhibition rate of each group was calculated as follows: The edema rate and percentage inhibition of each group were calculated as follows: . The % inhibition of edema was calculated by formula: % inhibition= 1-(a-x/b-y)*100

SNIOP, PUSAD

2012-2013

Page 58

Chapter No.7

Experimental Work

Where, A= paw thickness of test animal after treatment X= initial paw thickness of test animal B=paw thickness of control animal after treatment Y= initial paw thickness of control animal.

7.8. Stability studies86


Stability study was performed as per ICH guideline. The purpose of stability testing is to provide evidence on how the quality of a drug substance or drug product varies with time under the influence of a variety of environmental factors such as temperature, humidity and light. Therefore, stability studies provide data to justify the storage condition and shelf-life of the drug product. For drug substance, such studies establish the retest date in addition to the storage condition of raw material. Initial formulation are packed in the aluminum lacquered tube and kept in the stability chamber, at different stability condition. All the selected formulations were subjected to a stability testing for three months as per ICH norms at a temperature of 40 c 2 c /75 % 5 % RH. All selected formulations (LF7 and LF8) were analyzed for the change in appearance, pH and drug content by procedure stated earlier.

SNIOP, PUSAD

2012-2013

Page 59

Chapter No.8

Results and Discussion

8. Results and Discussion


8.1 Identification test 8.1.1. Aceclofenac Physical characters of aceclofenac were found as
Table 9: Result of physical characters of aceclofenac drug

S.no. 1 2 3 4 Nature Color Odor Taste

Characters

Inference Amorphous powder white Odorless Slightly Bitter

Solubility5 In methanol In water In ethanol Soluble Practically insoluble Freely soluble

8.1.2. Melting point The melting point of the aceclofenac was determined by capillary method and found to 149-153 0C which compiled with melting point reported in BP. 8.1.3. Ultra-violet scanning The scanning of aceclofenac was performed in 7.4 pH phosphate buffer saline and max was found to be 276 nm which was compiles with the max reported in BP.

SNIOP, PUSAD

2012-2013

Page 60

Chapter No.8

Results and Discussion

Fig.10: UV Spectrum of aceclofenac

8.1.4. Fourier Transforms Infrared (FTIR) spectroscopy-Scanning was performed between 4000-500 cm-1 range.

Fig.11: FTIR of aceclofenac

SNIOP, PUSAD

2012-2013

Page 61

Chapter No.8 Table 10:- Interpretation of aceclofenac FTIR

Results and Discussion

Sr. No. 1

Functional Group Amino group, OH, Aliphatic and Aromatic CH Carboxylic acid salt Aromatic ring Aromatic ether Isopropyl group Aliphatic ether, sec.alcohol 1,4-disubstituted benzene

Wave Number (cm-1) 3600-2300

2 3 4 5 6 7 8.1.5. Lipids

1580 1580, 1515 1250, 1015 1180 1100 820

8.1.5.1. Soyalecithin Saponification value60 Saponification value = 28.5 V/W Where, V- Difference in ml between titration W- Weight in g of substance taken. Result: Saponification value was found to be 190-195 which complies with Merck Index76.

SNIOP, PUSAD

2012-2013

Page 62

Chapter No.8

Results and Discussion

8.2. Preparation of calibration curve of aceclofenac


Table 11: UV calibration curve reading of aceclofenac in phosphate buffer saline pH 7.4

Sr. No. 1 2 3 4 5 6

Concentration (g/ml) 0 2 4 6 8 10

Absorbance 0 0.0259 0.0452 0.0616 0.0896 0.1072

Result: The prepared dilutions obeys Beers Lamberts law in the entire concentration range selected and the coefficient of correlation was found to be 0.994.

Calibration curve
0.12 0.1 Absorbance 0.08 0.06 0.04 0.02 0 0 2 4 6 8

y = 0.0109x R = 0.9946

10

12

Concentration g/ml

Fig.12: UV Calibration curve of aceclofenac

From the scanning of drug in phosphate buffer pH 7.4, it was concluded that the drug had max of 276 nm. From the standard calibration curve of aceclofenac in pH 7.4 buffer (Figure), it was concluded that drug

SNIOP, PUSAD

2012-2013

Page 63

Chapter No.8

Results and Discussion

obeys Beer-Lamberts law in concentration range of 0-10 g/ml. The linear equation in pH7.4 Phosphate buffer was obtained as: y = 0.0109x R = 0.994.

Correlation co-efficient value indicated the linear correlation between concentration and absorbance.

8.3. Evaluations of lipospehers


Table 12: Results of entrapment efficiency Code Lipid core Material F1 Stearyl alcohol F2 Carnauba wax F3 Bees wax F4 Stearyl alcohol F5 Carnauba wax F6 Bees wax F7 Stearyl alcohol F8 Carnauba wax F9 Bees wax F10 Stearyl alcohol F11 Carnauba wax F12 Bees wax Entrapment SD (n=3) 96.350.941 70.42.1.38 76.352.92 101.650.99 32.862.52 88.841.19 78.070.97 96.922.51 55.480.59 120.22.40 50.291.73 49.381.81 Stabilizer (q.s.) Pectin Pectin Pectin ---------Gelatin PVA PVA PVA PVA PVA Gelatin Appearance DP DP AP DP DP DP DP DP AP DP DP AP

DP=dispersed particle, AP= aggregated particle, PVA Polyvinyl alcohol

Entrapment Efficiency

96.3

101.65 70.42 76.35 32.86

120.2 88.84 78.07 96.92

55.48

50.2 49.38

10

11

12

Fig.13: Result of entrapment efficiency of liposphere (F1-F12)

SNIOP, PUSAD

2012-2013

Page 64

Chapter No.8

Results and Discussion

8.3.1. Photomicroscopic analysis The photomicrographs of aceclofenac lipospehers of formula F8 was represented by Fig. 14 & 15. It reveals the uniform spherical shape of

lipospehers showing the solid lipid core and the phospholipid coat.

Fig.14

Fig. 15

Fig. Photomicrograph of optimized aceclofenac lipospehers prepared by melt method at a magnification of 60 X (Fig.14) and 200X (Fig.15)

8.3.2. Particle size analysis From Table 13 it was evident that the combination of carnauba wax (F8) are stearyl alcohol (F7) with the highest amount the smallest particles. Use of soy lecithin and egg phosphatidylcholine resulted in the smallest particle size but the lipospehers were found to be aggregated with soya lecithin when observed under microscope. Carnauba wax with egg phosphatidylcholine gives the smallest particle size was decreased. As the core/coat ratio increased, the particle size of lipospehers. This decrease in particle size was probably due to the availability of higher amounts of carrier material and emulsifier for the formation of discrete spherical particles.

SNIOP, PUSAD

2012-2013

Page 65

Chapter No.8

Results and Discussion

Table 13: Mean particle size of different batchs (F1-F12)

Formulation code Mean particle size (m) F1 F2 F3 F4 F5 F6 F7 F8 F9 F10 F11 F12 28.20.31 14.2.61 56.8.0.32 42.90.43 22.30.12 85.20.32 14.20.12 14.21.22 42.90.65 42.90.46 85.20.98 96.20.12

8.4. Drug lipid compatibility


8.4.1. Fourier transform infrared (FTIR) spectroscopy Drug-lipid compatibility in optimized lipospehers (F6, F7, and F8) was evaluated by FTIR and DSC analysis. FTIR spectra are shown in Fig. 16, 17, 18, 19. Interactions between the ingredients used in topical formulations, such as Aceclofenac, carnauba wax, bees wax, stearyl alcohol, Carbopol were studied using FTIR spectroscopy. Aceclofenac was identified by the presence of characteristic bands. The FTIR spectral analysis of aceclofenac alone (Fig. 9) showed that principle peaks were observed at wave numbers 3316 (N-H stretching vibration for amine), 2935 (aromatic C-H stretching), 1769 (carbonyl C=O stretching). The same peaks were observed at 3325, 2954, 1733, 1418, 1234, and 719 in F6 at 3302, 2955, 1471, 1299, and, 718 in F7 at 3320, 2954, 1733, 1467, 1257, 1260 and 719 cm1 in F8. All the principle characteristic bands of the drug were also observed in the respective formulations. The

SNIOP, PUSAD

2012-2013

Page 66

Chapter No.8

Results and Discussion

findings were suggestive of the absence of any major incompatibility between aceclofenac and the components employed in the preparation of dermatological bases.

Fig. 16: FTIR of pure drug

Fig. 17: FTIR of formulation (F6)

Fig.18: FTIR of formulation (F7)

SNIOP, PUSAD

2012-2013

Page 67

Chapter No.8

Results and Discussion

Fig. 19: FTIR of formulation F8

8.4.2. Differential scanning calorimetry (DSC)

Figure 20, 21 and 22 showed the results of the DSC analysis of aceclofenac, and the physical mixture of drug loaded liposphere formulation. From the DSC thermograms, it was observed that melting point depression occurred The DSC thermogram of aceclofenac showed an exothermic peak at 160.44 C, which is the reported melting point of the aceclofenac. Drugloaded liposphere showed a large endothermic peak at 89.07 C (F6) and 76.22 (F8). It was observed from the DSC thermogram that the exothermic peak of aceclofenac at about 160.4 C no longer exists in the DSC, traces of the drug-loaded liposphere. Taking into consideration the drug-crystal-free particle surface, it is apparent that aceclofenac is amorphously dispersed within the liposphere, which is preferable for a controlled release system. Furthermore, the inclusion of drug molecules in the lipid is normally accompanied by a depression in the lipids melting point.

SNIOP, PUSAD

2012-2013

Page 68

Chapter No.8

Results and Discussion

DSC mW

Thermal Analysis Result


File Name: Detector: Acquisition Date Acquisition Time Sample Name: Sample Weight: Annotation: Pure Drug .tad DSC60 13/03/01 14:49:51 Pure Drug 7.060[mg] Start End Peak Onset
0 151.40 x10 C 0 172.85 x10 C

Temp C 200.00

0.00

-10.00

160.44 0 C x10
0 154.10 x10 C 0 170.49 x10 C

150.00

-20.00

Endset Heat Height

-754.14 0 x10 mJ -106.82 0 x10 J/g -38.56 0 x10 mW

-30.00

100.00 -40.00

0.00

1.00

2.00 Time [min]

3.00

4.00

Fig.20: DSC of pure aceclofenac drug

DSC mW

Thermal Analysis Result


File Name: Detector: Acquisition Date Acquisition Time Sample Name: Sample Weight: Annotation: F8.tad DSC60 13/03/01 15:28:40 F8 7.310[mg]

Temp C

0.00

200.00

-5.00

Start End Peak Onset


-10.00

68.71

0 x10 C

0 101.32 x10 C 0 89.32 x10 C

76.22 0 x10 C
0 97.91 x10 C 0 -414.84 x10 mJ -56.75 0 J/g x10

Endset Heat Height

-13.58 0 x10 mW

100.00

-15.00

0.00

2.00

4.00 Time [min]

6.00

8.00

Fig.21: DSC of Drug loaded liposphere (F8)

SNIOP, PUSAD

2012-2013

Page 69

Chapter No.8

Results and Discussion

DSC mW

Thermal Analysis Result


File Name: Detector: Acq uisition Date Acq uisition Time Sample Name: Sample Weig ht: Annotation: F6 NEW.tad DSC60 13/03/01 15:43:15 F6 NEW 7.720[mg ]

Temp C

0.00

200.00

Start End
-5.00

0 71.47 x10 C 0 102.07 x10 C 0 89.07 x10 C 0 75.80 x10 C 0 98.04 x10 C

Peak Onset Endset Heat Heig ht

-316.77 0 x10 mJ -41.03 0 J/g x10


0 -9.92 x10 mW

100.00

-10.00

0.00

2.00

4.00 Time [min]

6.00

8.00

Fig.22: DSC of Drug loaded liposphere (F6)

8.4.3. Scanning electron microscopy


Figure 23, 24 illustrates the SEM of optimized F8 batch of prepared aceclofenac lipospehers. The particles are spherical in shape with irregular surface as usually obtained when egg phosphatidylcholine is used as the coat.

Fig.23: Scanning photomicrographs of aceclofenac lipospehers with carnauba wax: under high magnification (1000X)

SNIOP, PUSAD

2012-2013

Page 70

Chapter No.8

Results and Discussion

Fig.24: Scanning photomicrographs of aceclofenac lipospehers with Carnauba wax: under low magnification (350X)

The SEM photomicrographs in figure (23 and 24) showed that the lipid lipospheres of batch have a spherical morphology. In SEM photomicrographs, the lipid lipospheres were observed at different magnification value i.e. 350 X, 1000 X which showed surface texture of lipospehers. Surface texture of the lipospheres is rough because of presence of crystals of drug on the surface of lipid lipospheres.The lipospheres were found to be spherical, rounder, free flowing and of the monolithic matrix type. The SEM photomicrographs of drug-loaded lipospheres showed that the lipospheres were almost spherical in shape with rough and nonporous surface and it also indicated that the drug was dispersed at amorphous state in the lipid matrices.

SNIOP, PUSAD

2012-2013

Page 71

Chapter No.8

Results and Discussion

8.5.4. In vitro release of aceclofenac from lipospehers (F1-F4)


Table 14: Percentage drug release from various formulations of Liposphers

S.No. Time (min) 0 30 60 120 180 240 300 360 420 480 540 600

Cumulative percent drug release (%) F1 F2 F3 0 4.087 8.974 18.72 20.01 24.96 26.43 37.48 47.27 59.88 68.13 75.31 0 14.86 21.31 38.01 52.23 66.36 80.24 94.20 ----0 23.23 40.54 45.59 49.22 57.53 62.41 67.39 77.46 79.23 82.00 92.52 0 38.53 46.85 50.58 57.95 61.92 65.80 72 75.11 79.48 82.16 83.81

F4

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12.

(Mean, n=3)
100 90 Cumulative % drug Release 80 70 60 50 40 30 20 10 0 0 100 200 300 400 500 600 700 Time (min) F1 F2 F3 F4

Fig.25: in vitro drug release of formulation batches F1, F2, F3, F4

SNIOP, PUSAD

2012-2013

Page 72

Chapter No.8

Results and Discussion

8.5.5. In vitro release of aceclofenac from lipospehers (F5-F8)


Table 15: Percentage drug release from various formulations of liposphers

S.No. Time (min) 0 30 60 120 180 240 300 360 420 480 540 600

Cumulative percent drug release (%) F5 F6 F7 0 29.96 33.82 38.75 42.78 43.94 51.33 67.47 69.23 72.33 80.57 82.88 0 25.57 27.60 31.02 32.76 37.69 48.96 49.96 56.28 58.97 69.05 81.11 (Mean, n=3) 0 16.94 32.76 38.02 47.27 55.34 66.36 75.36 83.81 91.68 --0 18.58 29.96 38.35 52.14 66.36 75.28 83.72 91.68 92.52 ---

F8

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12.

100

90
Cumulative % drug Release

80 70 60 50 40 30 20 10 0 0 100 200 300 400 500 600 700 F5 F6 F7 F8

Time (min)

Fig.26: in vitro drug release of formulation batches F5, F6, F7, F8

SNIOP, PUSAD

2012-2013

Page 73

Chapter No.8

Results and Discussion

8.5.6. In vitro release of aceclofenac from lipospehers (F9-F12)


Table 16: Percentage drug release from various formulations of liposphers

S.No. Time (min) 0 30 60 120 180 240 300 360 420 480 540 600

Cumulative percent drug release (%) F9 F10 F11 0 8.52 16.92 18.88 27.29 37.73 46.76 55.00 66.04 75.28 80.07 83.64 0 0 12.72 13.47 17.1 16.92 27.93 25.33 33.81 42.22 43.91 43.94 50.63 51.33 59.13 67.47 67.38 69.23 68.22 72.33 79.23 83.01 82.60 83.90 (Mean, n=3)

F12

1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12.


90 80 70 60 50 40 30 20 10 0 0

0 17.16 25.41 31.10 36.94 40.54 42.23 50.64 59.04 62.32 73.63 83.89

F9 F10 F11 F12

100

200

300

400

500

600

700

Fig. 27: in vitro drug release of formulation batches F9, F10, F11, and F13

8.6. Drug release kinetics The cumulative percent drug release curve of the drug loaded lipospehers. it is obvious that with all lipospheres prepared using egg phosphatidylcholine, the percentages of drug released after 8 h (T8hr) were overall significantly
SNIOP, PUSAD 2012-2013 Page 74

Chapter No.8

Results and Discussion

lower than with those prepared with soybean phosphatidylcholine under the same conditions. In addition, soybean phosphatidylcholine membranes are known to be more fluid than egg phosphatidylcholine membranes resulting in faster drug release. The highest T 8h value was obtained with carnauba wax, formulation F8 (92.52 %) and F7 (91.68 %) formulation, with stearyl alcohol. The lowest T 8h value was obtained with Bees wax, formulation F6 (58.97 %). The optimized batchs F8 and F7. Drug release kinetics for formulations F1-F12 was shown in table (18) shows First Order, Higuchi's and Peppas Korsmeyer's plot respectively.
Table 17: drug release kinetics for the various formulations of lipospheres

R2 Formulation Code Zero First Korsmeyer n Higuchis order order Peppas equation equation equation equation 0.9852 0.9929 0.7947 0.4777 0.8383 0.8855 0.9396 0.9440 0.9934 0.9743 0.9673 0.9364 0.9321 0.8748 0.9682 0.9515 0.9695 0.9303 0.9712 0.9578 0.9641 0.9790 0.9783 0.9329 0.9557 0.9571 0.9389 0.8751 0.9591 0.9425 0.9891 0.9925 0.9855 0.9935 0.9917 0.9592 0.9866 0.9970 0.9787 0.9892 0.9464 0.9287 0.9881 0.9960 0.9874 0.9964 0.9908 0.9771 0.9364 0.7696 0.4105 0.2745 0.3677 0.3847 0.5719 0.6054 0.7842 0.6625 0.6732 0.4947

F1 F2 F3 F4 F5 F6 F7 F8 F9 F10 F11 F12

SNIOP, PUSAD

2012-2013

Page 75

Chapter No.8

Results and Discussion

Result: The correlation coefficient (R2) values showed that formulations follow Korsmeyer peppas model for drug release. 8.6.1. Zero order, First order, Higuchis equation,and Korsmeyer Peppas equation plot of drug release Optimized batch of Lipospheres F7, F8 (a, b)

120 100
% Drug Released

Release Profile

80 60 Zero

1st 40
20 0 0 100 200 300 Time 400 500 600 Matrix Peppas

Hix.Crow.

a) Fig.28: F7

140 120
% Drug Released

Release Profile

100 80 Zero 60 40

1st
Matrix Peppas

20
0 0 100 200 300 Time 400 500

Hix.Crow.
600

b) Fig.29: F8
SNIOP, PUSAD 2012-2013 Page 76

Chapter No.8

Results and Discussion

The drug release kinetics showed in (Table 17), where majority of the batches governed by peppas model. All the batches showed release up to 8 h and above 40-90% of drug released with each formulation. Formulation F8 (92.52%) showed maximum release while other formulation showed less amount of drug release in 8 h. Formulation of F7, F8 and F11 has highest correlation coefficient (R2=0.9970, 0.9960, 0.9908) respectively and follows drug release by peppas model. The drug release from lipospheres depends on many factors including the composition of lipospheres, the type of drug encapsulated and nature of the cell. Once released, drug that normally crosses the membrane of a cell will enter the cell. The drug is released from lipospheres by one of the possible mechanism i.e., Endocytosis, fusion and adsorption. Orally administered lipospheres release the drug by endocytosis as gut epithelial cells take up intact lipospheres by absorptive endocytosis.

8.7. Formulations of lipospheres based gel


Lipospheres based gel was prepared according to the formula (Table 8).

8.8. Characterization of lipospheres based topical gel


Table 18: Result of pH, Viscosity, Drug content and Spreadability

S. No. 1 2 3 4

Formulation pH

F7 F8 LF7 LF8

6.5 6.9 6.9 7.0

Viscosity Drug (Cps) content (%) 29600 83.33 30200 96.51 29200 100.317 31000 115.73

Time(sec) Spredability (g.cm/sec) 20 20 20 20 47.27 37.14 40 34.66

SNIOP, PUSAD

2012-2013

Page 77

Chapter No.8

Results and Discussion

Fig.30: Lipospheres based gel of optimized batch

All the Lipospheres based topical gel formulations were having good feel and showed no clogging and lumps which indicate good texture of system. pH of lipospshere based topical gel was around the neutral pH and in the range of 6.5-7.0. All the formulations showed no significant skin irritation on intact skin (Fig 32).Thus, indicating skin acceptability of these formulations for topical application. Viscosity is an important parameter for characterizing the gels as it affects the spreadibility, and release of the drug. Viscosity of formulations was ranging between 29000-32000 cps. Easy spreadability is one of the important characteristics of any topical preparation as far as patient compliance is concerned. Liposphere based gel is considered to be good if it takes minimum time to spread on the surface. Among the various gels studied LF8 aceclofenac liposphere gel was find to show better spreadability. The values of spreadability indicated that the gel is easily spreadable by small amount of shear. Drug content uniformity of all formulations were observed and F7, F8 without permeation enhancer and LF7,LF8 with permeation enhancer batch showed the 83.33 %, 96.51 %, 100.317 % and 115.73 % drug content respectively. Finally selected formulations were subjected to a stability testing for three months and drug content of batch F7, F8, LF7 and LF8 was found to be 83.33 %, 96.51 %, 100.317 % and 115.73 % respectively. Depending upon different evaluation
SNIOP, PUSAD 2012-2013 Page 78

Chapter No.8

Results and Discussion

parameters made on all formulations, batch LF8 was declared as an optimized batch.

8.8.1. In vitro permeation of liposphere based gel


The release profile of a drug predicts how a delivery system might function and gives valuable insight into its in vivo behavior. The optimized (F8 and LF8) were subjected to in vitro release studies. These in vitro release studies were carried out using simulated tear fluid (STF) of pH 7.4 as the dissolution medium. The drug release data obtained for formulations shows the cumulative percent drug released. It was found that cumulative percent drug release was 82.42 %, 91.68 % for F8 and LF8 formulation respectively.
Table 19: Result of cumulative percent (%) drug release of optimize batch (F8, LF8)

S.NO.

Cumulative percent (%) drug release Time (hr) Batch F8 0 25.33 33.74 43.85 55.69 64.12 71.12 79.32 82.42 0 24.84 35.03 47.32 58.49 66.02 75.40 79.23 91.68 Batch LF8

1 2 3 4 5 6 7 8 9

0 1 2 3 4 5 6 7 8

SNIOP, PUSAD

2012-2013

Page 79

Chapter No.8
100 90 80 70 60 50 40 30 20 10 0 0 2 4 time (hr) 6 8

Results and Discussion

cumulative Percent drug release

F8 LF8

10

Fig.31: in vitro release profile of optimized formulation F8 and LF8

8.8.2. Skin irritation testing (Draize patch test)


The results of the skin irritation study revealed that following 72 h application of LF7, LF8 and marketed preparation (AUDIGEL) and blank gel, there was no reaction found on the skin. Therefore, it can be assured that the gel formulation can be used for topical application.

Fig.32: The skin irritation results of lipospheres based topical gel to skin of four rabbits (number A,B,C,D) after administration of 72 h (1) No application (2) Marketed aceclofenac gel (AUDIGEL) (3) LS based topical gel (LF7) (4) LS based topical gel (LF8)

SNIOP, PUSAD

2012-2013

Page 80

Chapter No.8

Results and Discussion

8.8.3. In vivo anti-inflammatory study of lipospheres


Table 20: Result of Percentage inhibitions for anti-inflammatory activity

Edema degree (ml) at different movement (h)

Group

0 0.700.12

1 1.280.48

4 1.460.266

1.320.043 1.33.09

Control 0.770.22 Marketed LF7 LF8 0.890.24 0.980.36 1.080.37 1.120.3 1.060.40 0.90.40 0.910.41 0.860.4 0.840.46

0.650.077 0.820.075 0.840.077 1.020.19 0.720.36

Edema degree (ml) at different movement (h)


Group 5 6 7 8 % Inhibition after 8 hr Control 0.700.12 1.280.48 1.320.043 1.33.09 ----

Marketed 0.770.22

0.90.40

0.910.41

0.860.4

65%

LF7 (III) LF8 (IV)

0.890.24

0.980.36

1.080.37

1.120.3

81%

0.650.077 0.820.075 0.840.077 1.020.19 96.78%

Data were expressed as meanS.D and statistically assessed by one way analysis of variance (ANOVA). Values for edema rate percentage for lipospheres were compared to the saline control and the differences were determined statistically using Dunnetts t test. P<0.05 was considered significant.

SNIOP, PUSAD

2012-2013

Page 81

Chapter No.8

Results and Discussion

The anti-inflammatory activity of the optimized formulation was evaluated by the carrageenan-induced hind paw inflammation method on wistar rats. The percentage inhibition value of LF7 and LF8 was compared to marketed gel. Both of the formulations LF7 and LF8 not only decreased the inflammation by a larger magnitude, but also sustained the effect for a prolonged period. After 8 h, the percent edema for LF7 and LF8 was found to be 81 % and 96.78 %, respectively (as shown in Table 20). While in case of marketed gel, percentage edema inhibition were found to be 65 %. Hence the lipospheres based gel formulation of aceclofenac remained superior to the marketed product in its ability to suppress edema and sustained the antiinflammatory activity.

8.9. Stability studies


All the selected formulations were subjected to a stability testing for three months as per ICH norms at a temperature of 40 C 2 C / 75% 5% R H. All selected formulations (LF7 and LF8) were analyzed for the change in appearance, pH and drug content by procedure stated earlier. (Table: 21 stability studies)
Table 21: Result of stability study of optimized batch

S No. 1

Batches

LF7

LF8

Months Month Appearance Drug Appearance pH pHDrug content (%) content (%) Initial white 6.9 100.317 1 white 6.8 100.2 2 white 6.9 99.5 3 white 7.0 99.30 Initial white 7.0 115.73 1 white 6.9 105.21 2 white 6.8 106.2 3 white 7.0 102.1

SNIOP, PUSAD

2012-2013

Page 82

Chapter No.10

Future scope

10. Future scope we can Increase drug loading efficiency We can improve the bioavailability of poorly water soluble drug by lipid drug delivery system. Increasing physical and chemical storage stability Minimizing overall costs

SNIOP, PUSAD

2012-2013

Page 86

Chapter No.9

Summary and Conclusion

9. Summary
The lipospheres system is a newly introduced lipid-based carrier system developed for parenteral and topical drug delivery of bioactive compounds. Lipospheres consist of water-dispersible solid microparticles of particle size between 0.2500 m in diameter and composed of a solid hydrophobic fat core stabilized by one monolayer of phospholipid molecules embedded in their surface which is a potential group of penetration enhancers. Both egg and soybean phosphatidylcholine contain unsaturated fatty acids which may be responsible for the penetration enhancement. The packing nature of unsaturated fatty acids disrupts the stratum corneum lipid structure and enhances the percutaneous penetration of drugs, also they strongly raise the fluidity of the stratum corneum. In addition, lecithin has a high affinity for epidermal tissue and even seems to improve skin hydration. Being biodegradable and composed of natural body constituents, topically administered phospholipids can be generally considered as safe. Lipospheres like solid lipid nanoparticles are one of the carriers of choice for topically applied drugs because their lipid components have an approved status or excipients used in commercially available topical cosmetic or pharmaceutical preparations. The small size of the lipid particles ensures close contact to the stratum corneum and can increase the amount of the drug penetrating into the mucosa or skin. Due to their solid lipid matrix, controlled release from these carriers is possible which is important to supply the drug over a prolonged period of time and to reduce systemic absorption, increased drug stability can be achieved and finally lipospheres possess a film forming ability leading to occlusive properties.

The purpose of this study was to prepare lipospheres containing aceclofenac intended for topical skin delivery with the aim of exploiting the favorable properties of this carrier system and developing a sustained release formula to overcome the side effects resulting from aceclofenac oral
SNIOP, PUSAD 2012-2013 Page 83

Chapter No.9

Summary and Conclusion

administration. Lipospheres were prepared using different lipid cores (carnauba wax, bees wax, steryl alcohol) and phospholipids coats (egg phosphatidylchoine and soya phosphatidylcholine) by melt dispersion technique. Characterization of the prepared lipospheres formulation carried out through photomicroscopy, scanning electron microscopy (SEM), particle size analysis, diffential scanning caorimetry (DSC), and in vitro drug release and stability study. It was uniformly dispersed after suitably gelled by Carbopol 940 preparation. The characterization of the prepared lipospheres based topical gel rheological study, pH, Spreadability, drug content, skin irritation test. No oedema and erythema were observed after administration of lipospheres based aceclofenac gel on rabbit skin, the anti-inflammatory effect of liposphere systems was assessed by the rat paw edema technique and compared to the marketed product. Results revealed that liposphere systems were able to entrap aceclofenac at very high levels (101.65 %). The particle size of liposphere systems was well suited for topical drug delivery. DSC revealed the molecular dispersion of aceclofenac when incorporated in lipospheres. Lipospheres were very stable after 3 months storage at 28 C. Liposphere topical gel was found to possess superior antiinflammatory activity compared to the marketed product.

SNIOP, PUSAD

2012-2013

Page 84

Chapter No.9

Summary and Conclusion

Conclusion
This present work indicates that the Lipospheres based aceclofenac gel could be successfully prepared by the melt dispersion technique. The melt dispersion method produced smaller particles. This study also indicates that the amount of lipids and lecithin significantly affects the particle size as well as entrapment efficiency. It can be concluded that the optimized lipospheres gel exhibit faster onset and prolonged action as compared to the marketed product. Further, in vivo anti-inflammatory and skin irritation studies are necessary to assess the improvement of therapeutic efficacy of the LS gel compared to the marketed product. Findings of this investigation suggest that lipospheres can be considered a promising delivery system for topical aceclofenac delivery. Lipospheres were able to entrap the drug at very high levels and sustained its release over a prolonged time. Lipospheres possessed a suitable size for topical route and being based on non irritating and non toxic lipids, lipospheres seemed to be well suited for use on damaged or inflamed skin. Furthermore, lipospheres possessed a very high stability as well as superior anti-inflammatory activity compared to the marketed product.

SNIOP, PUSAD

2012-2013

Page 85

Chapter 12

Errata

ERRATA __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________
SNIOP, PUSAD 2012-2013 Page 98

Chapter 12

Errata

__________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ __________________________________________________________ _________________________________________________________
SNIOP, PUSAD 2012-2013 Page 99

Chapter No.11

References

11. References 1. Domb, A.J. et al., Degradable polymers for site-specific drug delivery, Polymer Adv. Technol, (1992):3, 279. 2. Barke, S.A. and Khossravi, D., Drug delivery strategies for the new millennium, Drug Del. Technol, (2001):6, 75. 3. Mizushima, Y., Lipid microspheres (lipid emulsions) as a drug carrier: an overview, Adv. Drug Del. Rev, (1996):20, 113. 4. Ravi Kumar, M.N.V., Nano and microparticles as controlled drug delivery devices, J. Pharm. Pharmaceut. Sci, (2000):3, 234. 5. Gombotz, W. et al., Prolonged release of GM-CSF, United States Patent, (1995):5, 253. 6. Wake, M.C. et al., Effects of biodegradable polymer particles on rat marrowderived stromal osteoblasts in vitro, Biomaterials, (1998):19, 1255. 7. Cortesi, R. et al., Preparation of liposomes by reverse-phase evaporation using alternative organic solvents, J. Microencapsul,( 1999):16, 251. 8. Vyas, S.P., Singh, R. and Dimitrijevic, D., Development and characterization of nifedipine lipospheres, Pharmazie, (1997):52, 403. 9. Jenning, V., Thnemann, A.F. and Gohla, S.H., Characterization of a novel solid lipid nanoparticle carrier system based on binary mixtures of liquid and solid lipids, Int. J. Pharm,(2000):199, 167. 10. Schwarz, C. and Mehnert, W., Solid lipid nanoparticles (SLN) for controlled drug delivery. II. Drug incorporation and physicochemical characterization, J. Microencapsul (1999):16, 205.
SNIOP, PUSAD 2012-2013 Page 87

Chapter No.11

References

11. Mller, R.H., Mder, K. and Gohla, S., Solid lipid nanoparticles (SLN) for controlled drug delivery: a review of the state of the art, Eur. J. Pharm. Biopharm,( 2000):50, 161. 12. Takenaga, M., Application of lipid microspheres for the treatment of cancer, Adv. Drug Del. Rev, (1996):20, 209. 13. Yang, S.C., et al., Body distribution of camptothecin solid lipid nanoparticles after oral administration, Pharm. Res, (1999): 16, 751. 14. Inoue, K. et al, ex vivo anti-platelet effects of isocarbacyclin methyl ester incorporated in lipid microspheres in rabbits, Arzneim-Forsch/Drug. Res, (1995):45, 980. 15. Mller, R.H. et al., Solid lipid nanoparticles (SLN) as potential carrier for human use: interaction with human granulocytes, J. Control. Rel, (1997):47, 261. 16. Schwarz, C. et al., Solid lipid nanoparticles (SLN) for controlled drug delivery: production, characterization and sterilization, J. Control. Rel, (1994): 30, 83. 17. Bodmeier, R., Chen, H. and Bhagwatwar, H., Polymer and wax microspheres prepared by emulsification techniques, Bull. Tech. Gattefoss, (1990):83. 18. zur Mhlen, A., Schwarz, C. and Mehnert, W., Solid lipid nanoparticles (SLN) for controlled drug delivery: drug release and release mechanism, Eur. J. Pharm. Biopharm, (1998):45, 149.

SNIOP, PUSAD

2012-2013

Page 88

Chapter No.11

References

19. Luxenhofer R, Schulz A, Roques C, Li S, Bronich TK, Batrakova EV, et al. Doubly amphiphilic poly (2-oxazoline) s as high-capacity delivery systems for hydrophobic drugs. Biomaterials, (2010):31(18), 4972-4979. 20 Jang JH, Shea LD. Intramuscular delivery of DNA releasing microspheres: microsphere properties and transgene expression. J Control Release. (2006):112(1), 120-128. 21. Dey N, Majumdar S, Rao M. Multiparticulate drug delivery systems for controlled release. Trop. j. pharm. Res, (2008):7(3), 1067-1075. 22. Wichert B, Rohdewald P. A new method for the preparation of drug containing polylactic acid microparticles without using organic solvents. J Control Release,(1990):14(3), 269-283. 23. Fricker G, Kromp T, Wendel A, Blume A, Zirkel J, Rebmann H, et al. Phospholipids and Lipid-Based Formulations in Oral Drug Delivery. Pharm Res (2010):1-18. 24. Muller RH, Mader K, Gohla S. Solid lipid nanoparticles (SLN) for controlled drug delivery-a review of the state of the art. Eur J Pharm Biopharm (2000):50(1), 161-177. 25. Bangham AD. Techniques in life Science, lipid membrane biochemistry. Elsevier Scientific Publications. Ireland, (1982):25. 26. Lachman L, Lieberman HA, Kanig JL. In, The Theory and Practice of Industrial Pharmacy. Varghese Publishing House. Bombay, (1987):417. 27.Vyas SP, Khar RK. Targeted and controlled Drug Delivery, Novel Carrier system, CBS Publishers and Distributors, (2002): 263-65.

SNIOP, PUSAD

2012-2013

Page 89

Chapter No.11

References

28. Datta S, Karki R. Liposomes encapsulated Rifampicin and Isoniazid. Indian J Pharm Sci (2000): 62(5), 384. 29. Ramana MV, Chaudhari AD. An approach to minimize pseudo membranous colitis caused by Clindamycin through liposomal formulation. Indian J Pharm Sci (2007), 69(3), 390. 30.Qui L, Jing N, Jin Y. Preparation and invitro evaluation of liposomal chloroquinediphophate loaded by a transmembrane pH-gradient method. Int J Pharma,(2008), 56-63. 31. Claudio Nastruzzi, editor. Lipospheres in Drug Targets and Delivery Approaches, Methods, and Applications. New York: Washington, 2005. 32. Domb, Abraham J, Maniar, Manoj, Lipospheres for Controlled Delivery of Substances. European Patent: EP0502119. 33. Mu L, Feng S. Fabrication, characterization and in vitro release of paclitaxel (Taxol) loaded poly (lactic-co-glycolic acid) microspheres prepared by spray drying technique with lipid/ cholesterol emulsifiers. J Control Release, (2001), 76(3), 239-254. 34.Toongsuwan S, Li LC, Erickson BK, Chang HC.Formulation and characterization of bupivacaine lipospheres. Int J Pharm.,(2004):280(1-2), 5765. 35. Chakraborty S, Shukla D, Mishra B, Singh S. Lipid-An emerging platform for oral delivery of drugs with poor bioavailability. Eur J Pharm Biopharm., (2009):73(1), 1-15. 36. Wong HL, Rauth AM, Bendayan R, Manias JL, Ramaswamy M, Liu Z, et al. A new polymerlipid hybrid nanoparticle system increases cytotoxicity of
SNIOP, PUSAD 2012-2013 Page 90

Chapter No.11

References

doxorubicin against multidrug-resistant human breast cancer cells. Pharm Res., (2006):23(7), 1574-1585. 37. zur Mhlen A, Schwarz C, Mehnert W. Solid lipid nanoparticles (SLN) for controlled drug delivery-drug release and release mechanism. Eur J Pharm Biopharm, (1998): 45(2), 149-155. 38. Cortesi R, Esposito E, Luca G, Nastruzzi C. Production of lipospheres as carriers for bioactive compounds. Biomaterials, (2002): 23(11), 2283-2294. 39. Bhatia A, Singh B, Rani V, Katare O. Formulation, characterization, and evaluation of benzocaine phospholipid-tagged lipospheres for topical

application. J Biomed Nanotechnol, (2007):3(1), 81-89. 40. Sznitowska M, Janicki S, Gajewska M, Kulik M. Investigation of diazepam lipospheres based on Witepsol and lecithin intended for oral or rectal delivery. Acta Pol PharmDrug Res, (2000):57, 6164. 41. Bekerman T, Golenser J, Domb A. Cyclosporin nanoparticulate lipospheres for oral administration. J Pharm Sci ,004;93(5):1264-1270. 42. Sandipan dasgupta et al, nanostructured lipid carriers (NLC) based gel for the topical delivery of aceclofenac: preparation, characterization, and in vivo evaluation, scientia pharamaceutica 2012, 7-8. 43. Esimone et al, formulation and evaluation of goat fat and shea butter based lipospheres of benzyl penicillin, international journal of

pharmaceutical sciences and research, (2012):4, 1022-1027. 44. Abraham J. Domb et al, cyclosporin pro-dispersion liposphere formulation, journal of controlled release, (2012):160,406-407.

SNIOP, PUSAD

2012-2013

Page 91

Chapter No.11

References

45. Vignesh muruganandam, formulation development & characterization of ofloxacin microspheres,indo global journal of pharmaceutical sciences,( 2012):2(2),130-141. 46. Sanming Li et al,Nanostructured lipid (NLC) based topical gel of flurbiprofen :Design characterization and in Vivo evaluation, international Journal of pharmaceutics, (2012):439, 349-357. 47. Sateesh Babu N et al, formulation design and in vitro evaluation of azithromycin loaded lipospheres using melt dispersion technique, journal of pharmacy research, (2011):4(11), 4069-4071. 48. Loganathan Veerappan et al, formulation development and evaluation of flurbiprofen Lipospheres,International Journal for the advancement of science & Arts, (2010): Vol.1 (1). 49. Kamal dua et al,aceclofenac topical dosage forms:In vitro and In vivo characterization ,Acta pharm, (2010):60, 467-478. 50. Maha Nasr et al, Lipospheres as carriers for topical delivery of aceclofenac:preparation characterization and In vivoevaluation AAPs pharmscitech, (2008):19 (1), 154-162. 51. Jia you fang et al,a study of the formulation design acoustically active lipospheres as carriers for drug delivery,European Journal of pharmaceutics and Biopharmaceutics, (2007):67, 67-75. 52. Hagalavadi Nanjappa shivkumar et al, design and statistical optimization of glipizide loaded lipospheres using response surface methology, Actapharm, (2007):57, 269-285.

SNIOP, PUSAD

2012-2013

Page 92

Chapter No.11

References

53. vandana B. Pataravale et al, solid lipid nanoparticles (SLN) of tretinoin: Potential in topical delivery, international journal of pharmaceutics, (2007):345, 163-171. 54. http://www.tzzy.com/lh6.htm. 55. http://www.chemyq.com/En/xz/xz9/83172xpppl.htm. 56. http://www.medindia.net/doctors/drug_information/aceclofenac.htm. 57.Indian Pharmacopoeia, Ministry of Health and family welfare, govt. of India, Published by the Drug Controller of India, New Delhi, (2007): vol. II, 6364. 58. Dufour P, Vuillemard JC, Laloy E. Characterization of enzyme immobilization in liposomes prepared from proliposomes. J

Microencapsulation, (1996): 13(2), 185-94. 59. Indian pharmacopoeia, Government of India, Ministry of Health & Family Welfare, The controller of Publications, New Delhi, (2007): Vol-II, 143. 60.Indian Pharmacopoeia, Ministry of Health and family welfare, govt. of India, Published by the Drug Controller of India, New Delhi, (2007): Vol-II, 143.

61. Perrett S, Golding M, Williams PW.A simple method for the preparation of liposome for pharmaceutical applications: Characterization of the liposomes. J of Pharmacol,(1991): 43, 154-61. 62. Al-Angry AA, Bayomi MA, Khidr SH. Characterization stability and in vivo targeting of liposomal formulations containing Cyclosporine.Int. J of Pharmaceutics, (1995): 114, 221-225.

SNIOP, PUSAD

2012-2013

Page 93

Chapter No.11

References

63. A. J. Domb, L. Bergelson, and S. Amselem. Lipospheres for controlled delivery of substances Microencapsulation, methods and industrial applications, Marcel Dekker, New York, NY, (1996), 377410. 64. D. B. Masters and A. J. Domb. Liposphere local anesthetic timedrelease for perineural site application. Pharm. Res, (1998): 15, 10381045. 65. V. Iannuccelli, N. Sala, R. Tursilli, G. Coppi, and S. Scalia. Influence of liposphere preparation on butyl-methoxydibenzoyl methane photostability. Eur. J. Pharm. Biopharm, (2006): 63,140145. 66. R. Tursilli, A. Casolari, V. Iannuccelli, and S. Scalia. Enhancement of melatonin photostability by encapsulation in lipospheres. J. Pharm. Biomed. Anal. (2006): 40:910914. 67. F. Coi, D. Cun, A. Tao, M.Yang , Y.K.Shi and Y.Guan. Preparation and characterization of melittin- loaded poly (DL-lactic acid) or poly (DLlactic co- glycolic acid )microspheres made by double emulsion method, Journal of Controlled release, (2005):107 (2), 310-319. 68. I. El Gibaly, and S. K. Abdel-Ghaffar. Effect of hexacosanol on the characteristics of novel sustained release allopurinol solid lipospheres (SLS): factorial design application and product evaluation. Int. J. Pharm, (2005):294, 3351. 69. J. J. Sheng, P. J. Sirois, J. B. Dressman and G. L. Amidon, Particle diffusional layer thickness in a USP dissolution apparatus II: a combined function of particle size and paddle speed, J. Pharm. Sci, (2008): 4815 4829.

SNIOP, PUSAD

2012-2013

Page 94

Chapter No.11

References

70 Arul B, Sathyamurthy D. Formulation and evaluation of ketorolac tromethanmine gels.TheEastern Pharmacist (1998):135-6. 71 Chowdary KPR, Kumar PA. Formulation and evaluation of topical drug delivery systems of ciprofloxacin. Ind J Pharm Sci,(1996): 58 (2),47-50. 72 Sanghavi NM, Mahalaxmi D. Determination in vitro release of clobetasol propionate from topical bases.Indian Drugs, (1993): 30 (8),36470. 73 Mura P, Faucci MT, Bramanti G, Corti P. Evaluation of transcutol as a clonazepam transdermal penetration enhancer from hydrophilic gel formulations. Eur J Pharm Sci (2002): 9, 365-372. 74 Arellano A, Santoyo S, Martin C, Ygartua P. Influence of propylene glycol and isopropyl myristate on the in vitro percutaneous penetration of diclofenac sodium from carbopol gels. Eur J Pharm Sci, (1998): 7, 129-35. 75 Islam MT, Rodriguez-Hornedo N, Ciotti S, Ackermann C. Rheological characterization of topical carbomer gels neutralized to different pH. Pharm Res (2004): 21, 1192-1199. 76. Mohamed MI. Optimization of chlorphenesin emulgel formulation. The AAPS Journal, (2004):6 (3), 1-7. 77. Tas C, Ozkan Y, Savaser S, Baykara T. In vitro release studies of chlorpheniramine maleate from gels prepared by different cellulose derivatives,IL Farmaco, (2003): 58, 605-11.

SNIOP, PUSAD

2012-2013

Page 95

Chapter No.11

References

78. Gendy AME, Jun HW, Kassem AA. In vitro release studies of flurbiprofen from different topical formulations, Marcel Dekker, New York, Drug Dev Ind Pharm, (2002):48:823-31. 79. Aejaz A, Azmail , Sanaullah S, Mohsin A.A., Formulation and invitro evauluation of aceclofenac solid dispersion incorporated gels,International Journal of Applied Pharmaceutics,(2010): 2(1), 8-12. 80. Nagariya K., Jadon P.S, Naruka P.S., Chauhan C.S., Formulation development and characterization of aceclofenac gelusing poloxamer 407, J. Chem. Pharm. Res, (2010): 2(4), 357-363. 81. Mutimer M.N., Riffikin C., Hill J.A., Marry E., Cry N.G., Gliekman G.J., Am Pharm, Assoc. Sci,(1956):45, 212. 82. Thorat S.P., Rane S.I., Formulation and in vitro evaluation of lecithin (soya and egg) based aceclofenac organogels, Journal of Pharmacy Research, (2010): 3(6), 1438-1441. 83. Sirish Vodithala; Sadhna Khatry; Nalini Shastri; M Sadananda. International Journal of Current Pharmaceutical Research, (2010): 2(3), 33. 84. Basavaraj K. Nanjwade; Manjappa AS; Murthy RSR; Yuvaraj D Pol. Asian Journal of Pharmaceutical Sciences, (2009):4(3), 189. 85. Draize, J., Woodard, G., Calvery, H., Methods for the study of irritation and toxicity of substances topically applied to skin and mucous membranes. J. Pharmacol. Exp. Ther, (1944): 82, 377390. 86. www.ICH/stability.

SNIOP, PUSAD

2012-2013

Page 96

Chapter No.11

References

87. Al-Meshal MA, Khidr SH, Bayomi MA. Oral administration of liposomes containing Cyclosporine: a pharmacokinetic study. Int. J of Pharmaceutics (1998): 168, 163-168.

SNIOP, PUSAD

2012-2013

Page 97

You might also like