274
i Formulation and In-vitro/In-vivo Evaluation of Oral Diclofenac sodium and Diclofenac potassium Liposomal Dosage Forms A Dissertation Submitted in partial fulfilment of the requirements for the degree of Doctor of Philosophy (Pharmaceutics) by Muhammad Asadullah Madni B. Sc., B. Pharm., M. Phil. Department of Pharmacy Faculty of Pharmacy & Alternative Medicine The Isl he Isl he Isl he Islamia University of Bahawalpur amia University of Bahawalpur amia University of Bahawalpur amia University of Bahawalpur-Pakistan Pakistan Pakistan Pakistan 2010

Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

i

Formulation and In-vitro/In-vivo Evaluation of

Oral Diclofenac sodium and Diclofenac

potassium Liposomal Dosage Forms

A Dissertation Submitted

in partial fulfilment of the requirements for the degree

of

Doctor of Philosophy (Pharmaceutics)

by

Muhammad Asadullah Madni B. Sc., B. Pharm., M. Phil.

Department of Pharmacy

Faculty of Pharmacy & Alternative Medicine

TTTThe Islhe Islhe Islhe Islamia University of Bahawalpuramia University of Bahawalpuramia University of Bahawalpuramia University of Bahawalpur----Pakistan Pakistan Pakistan Pakistan 2010

Page 2: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

ii

In the name of Allah, the Most Merciful, the Most Kind

In the name of Allah, the Most Merciful, the Most KindIn the name of Allah, the Most Merciful, the Most Kind

Page 3: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

iii

"Whether you conceal what is in your hearts or bring

it into the open, Allah knows it: for He knows all that

is in the heavens and all that is on earth; and Allah

has the power to will anything."

(Al-Qur’an: 3:29)

Page 4: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

iv

Page 5: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

v

Page 6: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

vi

DECLARATION

I, Mr. Asadullah Madni, PhD scholar (Pharmaceutics) of the Department of Pharmacy,

the Islamia University of Bahawalpur, hereby declare that The research work entitled

“Formulation and In-vitro/In-vivo Evaluation of Oral Diclofenac sodium and Diclofenac

potassium Liposomal Dosage Forms” is done by me. I also certify that this dissertation

does not incorporate any material previously submitted for any degree in any university

without acknowledgment; and to the best of my knowledge and belief that it does not

contain any material previously published or written by any other person where due

reference is not made in the text.

Muhammad Asadullah Madni

Page 7: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

vii

DEDICATIONDEDICATIONDEDICATIONDEDICATION

All Family Members especially my Father and

Mother for their efforts in putting their children to

the pursuit of knowledge. Throughout my life, they

have actively supported me in my determination to

find and realise my potential, and to make this

contribution to our world. Thanks for their

unconditional love and support throughout the

course of this thesis.

AND

Mrs. Ameen Fatima and Mehboob Ahmad Bhatti for

their constant prayers and support through out my

professional education and especially in PhD studies.

AND

MS. Atezaz Ahmad, Rumaisa Asad, Umama Asad

and Muhammad Qasim for their love, patience,

sincerity and sacrifices.

AND

Amina Saeed and Hafiz Abdul Mannan for their

constant prayers and love.

Page 8: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

viii

ACKNOWLEDGEMENTS

I wish to acknowledge my deepest sense of gratitude to the Allah Almighty who is

the creator of the mankind and enabled me to complete this research successfully.

All respects for the Holy Prophet Muhammad (Peace be Upon Him) who guided

us to recognize our creator and whose way of education is unchallengeable.

I am much thankful to Prof. Dr. Mahmood Ahmad, Dean Faculty of Pharmacy

and Alternative Medicine, the Islamia University of Bahawalpur, who provided

guidelines and continuous encouragement to complete this research. He has been

generous for enough to spare his precious time for inspirational discussions

through out this project.

I am obliged to Dr. Nisar-ur-Rehman, Chairman and Associate Professor,

Department of Pharmacy, Dr. Naveed Akhtar, Associate Professor and Principal

College for Conventional Medicine, the Islamia University of Bahawalpur, Dr.

Muhammad Iqbal Chadhary, Chairman TUF, for precious advises during the

study to facilitate my research work and helped me in arranging the materials

required for the conduct of this project.

Special thanks to Prof. Dr. Muhammad Aleem, Chairman, Department of

Statistics for his support in statistical analysis of data and precious advises for

completion of this research project.

I am greatly indebted to Dr. Nasir Ali, Assistant Professor, Department of

Orthopedics, Quaid-e-Azam Medical College Bahawalpur for his constant

inspiring help and support in completing this research.

I pay special thanks to my friends especially Mr. Muhammad Usman, Mr. Mr.

Saleem Khalid, Mr. Abubakar Munir, Mr. Saleem Qureshi, Mr. Abdul Latif, Mr.

Qamar-uz-Zaman, Mr. Zafar Iqbal, Mr. Shaheq-uz-Zaman and Mr. Mahtab

Page 9: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

ix

Ahmad Khan for their constant support in conducting research and entertainment

through out this project.

I also appreciate help and support of Dr. Ghulam Murtaza, Mr. Shujat Ali Khan,

Mr. M. Naeem Aamir and Ms. Fatima Rasool.

I am grateful to my colleagues Dr. Riffat-uz-Zaman, Dr. Rao Saeed Ahmad, Dr.

Irshad Ahmad, Dr. Qaisar Jabeen, Mr. Waheed Asghar, Mr. H. M. Shoaib Khan,

Mr. Gulzeb Aziz, Mr. Muhammad Atif, Mr. Muhammad Akhtar, Ms. Gulfishan,

Mr. Rizwan Ahmad, Mr. Fahad Pervaiz, Mr. Qazi Adnan, Mr. Jaffar Shirazi, Mr.

Muhammad Khan Sarfraz, Mr. Sajid-ur-Rehamn, Mr. M. Younas and Ms. Nuzhat

Khanum for the collaboration and prayers.

Deep gratitude is extended to my friends Mr. Ahsan Ali, Mr. Nadeem Ahmad,

Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant

prayers, help, co-operation and entertainment during the study.

It will be injustice if I don’t pay thanks to Mr. Muhammad Abbas, Manzoor

Ahmad Baloch, Hafiz Muhammad Saleem, Muhammad Abdullah and all other

clerical and office staff for their prayers and co-operation.

I should not forget the efforts of Mr. Muhammad Shafique (store keeper) and

other laboratory assistants for their cooperation.

Muhammad Asadullah Madni

Page 10: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

x

CONTENTS Title ------------------------------------------------------------------------------------------i

Bismillah Sharif ----------------------------------------------------------------------------ii

Translation of Ayat from Holy Qur’an -------------------------------------------------iii

Approval Certificate------------------------------------------------------------------------iv

Approval Certificate after Open Defense------------------------------------------------v

Declaration----------------------------------------------------------------------------------vi

Dedication ----------------------------------------------------------------------------------vii

Acknowledgment-------------------------------------------------------------------------viii

List of Contents ----------------------------------------------------------------------------x

List of Tables -----------------------------------------------------------------------------xvii

List of Figures -----------------------------------------------------------------------------xx

List of Important Abbreviations -------------------------------------------------------xvii

Study Plan --------------------------------------------------------------------------------xix

Objectives --------------------------------------------------------------------------------xxx

ABSTRACT ---------------------------------------------------------------------------------1

CHAPTER: 1.

1. INTRODUCTION------------------------------------------------------------------------5

CHAPTER: 2.

LITERATURE REVIEW--------------------------------------------------------------------7 2.1. Structure and properties of liposomes -----------------------------------------11

2.2. Structural components of Liposomes -------------------------------------------13

2.2.1. Soya Lecithin --------------------------------------------------------------13

2.2.2. Cholesterol ------------------------------------------------------------------16

2.2.3. Poly Ethylene Glycol (PEG)-----------------------------------------------17

2.3. Liposomal formulations of different drugs using different techniques-----19

Page 11: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xi

2.4. Application of Liposomes in various Diseases---------------------------------31

2.4.1. Cancer ----------------------------------------------------------------------- 31

2.4.2. Acquired Immuno Deficiency Syndrome---------------------------------33

2.4.3. Dermatology -----------------------------------------------------------------34

2.4.4. Liver Diseases ---------------------------------------------------------------36

2.4.5. Lungs Diseases --------------------------------------------------------------38

2.4.6. Malaria -----------------------------------------------------------------------39

CHAPTER: 3. 3.1. Introduction ------------------------------------------------------------------------41

3.2. Experimental-----------------------------------------------------------------------45

3.2.1. Materials ---------------------------------------------------------------------45

3.2.2. Chromatographic system and applied conditions ----------------------45

3.2.3. Preparation of standard solutions ----------------------------------------46

3.2.4. Determination of analgesics in marketed brands ----------------------46

3.2.5. Determination of analgesics in drug delivery systems -----------------47

3.2.6. Determination of NSAIDs in human plasma ----------------------------47

3.3. Results and Discussions----------------------------------------------------------47

3.3.1. Choice of chromatographic separations---------------------------------47

3.3.2. Determination of analgesics in pharmaceutical formulations --------48

3.3.3. Determination of analgesics in human plasma--------------------------51

3.3.3.1. Linearity ---------------------------------------------------------51

3.3.3.2. Extraction efficiency--------------------------------------------53

3.3.3.3. Assay accuracy and precision in plasma--------------------54

Page 12: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xii

3.3.3.4. Stability----------------------------------------------------------56

3.4. Conclusion-------------------------------------------------------------------------58

CHAPTER: 4.

4.1. Introduction ------------------------------------------------------------------------59

4.2. Experimental----------------------------------------------------------------------- 62

4.2.1. Materials and solvents ---------------------------------------------------- 62

4.2.2. Chromatographic system and optimized conditions of Separation---62

4.2.3. Preparation of stock and working standard solution ----------------- 62

4.2.4. Preparation of liposomes-------------------------------------------------- 63

4.2.5. Sample preparation of liposomes, marketed brands and human

plasma ----------------------------------------------------------------------- 63

4.2.6. Method validation ---------------------------------------------------------- 64

4.2.7. Recovery--------------------------------------------------------------------- 64

4.2.8. Stability studies------------------------------------------------------------- 65

4.3. Results and discussion----------------------------------------------------------- 66

4.3.1. Screening and optimization------------------------------------------------66

4.3.1.1. Selection of detection wavelength----------------------------66

4.3.1.2. Choice of mobile phase composition ------------------------66

4.3.2. Method validation-----------------------------------------------------------68

4.3.2.1. Linearity----------------------------------------------------------68

4.3.2.2. Limit of detection and quantification-------------------------70

4.3.2.3. Precision and Accuracy----------------------------------------71

4.3.3. Recovery ---------------------------------------------------------------------73

Page 13: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xiii

4.3.4. Stability-----------------------------------------------------------------------75

4.4. Application of present method --------------------------------------------------78

4.5. Conclusions------------------------------------------------------------------------79

CHAPTER: 5.

5.1. Introduction ------------------------------------------------------------------------80

5.2. Materials and Methods-----------------------------------------------------------81

5.2.1. Materials ---------------------------------------------------------------------81

5.2.2. Preparation of liposomes---------------------------------------------------82

5.2.3. Entrapment Efficiency (EE) ---------------------------------------------- 84

5.2.4. Particle Size measurement and size distribution------------------------85

5.3. Study Design for liposomal formulations-------------------------------------- 85

5.4. Results and Discussion ---------------------------------------------------------- 89

5.4.1. Model validation ----------------------------------------------------------- 90

5.4.2. Response surface (contour) plots----------------------------------------- 91

5.4.3. Response optimization -----------------------------------------------------93

5.5. Comparison of different methods of preparations of liposomes------------ 93

5.6. Conclusion ------------------------------------------------------------------------ 99

CHAPTER: 6.

6.1. Introduction-----------------------------------------------------------------------100

6.2. Experimental ---------------------------------------------------------------------103

6.2.1. Materials -------------------------------------------------------------------103

6.2.2. Preparation of liposomes ----------------------------------------------- 104

6.2.3. Particle Size measurement and size distribution---------------------- 105

Page 14: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xiv

6.2.4. Morphology of liposomes ------------------------------------------------ 105

6.2.5. Differential Scanning Calorimetric Analysis-------------------------- 106

6.2.6. X-ray diffractometry (XRD)---------------------------------------------- 106

6.2.7. FT-IR studies -------------------------------------------------------------- 107

6.2.8. Entrapment Efficiency (EE) ----------------------------------------------107

6.2.9. Stability studies of liposomal formulations of diclofenac-------------108

6.2.10. In vitro release study -----------------------------------------------------108

6.2.10.1. In-vitro Kinetics and modeling----------------------------109

6.2.11. In-vivo Studies------------------------------------------------------------ 109

6.2.11.1. Subject selection, clinical design, drug administration

and sampling schedule--------------------------------------109

6.2.11.2. Pharmacokinetic analysis --------------------------------- 110

6.2.11.3. Statistical analysis------------------------------------------ 111

6.3. Results and Discussion --------------------------------------------------------- 112

6.3.1. Fabrication, Morphology, Size and Size Distribution of

Liposomes ----------------------------------------------------------------- 112

6.3.2. Measurement of Zeta potential (ζ)-------------------------------------- 120

6.3.3. Differential scanning calorimetric (DSC) evaluation of liposomes

formulation -----------------------------------------------------------------124

6.3.4. X-ray Diffraction (XRD) studies ---------------------------------------- 127

6.3.5. FT-IR studies -------------------------------------------------------------- 129

6.3.6. Encapsulation efficiency ------------------------------------------------- 131

6.3.7. Stability studies of liposomal formulations --------------------------- 132

Page 15: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xv

6.4. In-vitro Studies ------------------------------------------------------------------ 138

6.4.1. Profile of Liposomal Formulations of Diclofenac-------------------- 139

6.4.1.1. Diclofenac liposomes prepared with Purified Soya

Lecithin ------------------------------------------------------ 139

6.4.1.2. Liposomes prepared with Soya Lecithin and PEG------140

6.4.1.3. Diclofenac liposomes prepared with

Purified Egg Lecithin --------------------------------------141

6.4.1.4. Liposomes prepared with Egg Lecithin and PEG-------142

6.4.2. Discussion of In-vitro studies--------------------------------------------------- 156

6.5. Similarity factor determination (f-2) ----------------------------------------------162

6.5.1. Comparison of release profile of Liposomal formulation of diclofenac

sodium and Phlogin-SR®

Brooks Pharmaceuticals (Pakistan) as

Reference Brand --------------------------------------------------------------- 162

6.6. In-vivo Evaluation of Liposomal Formulations of Diclofenac------------ 165

6.6.1. Plasma concentration profile of liposomal formulations of DFS -- 166

6.6.1.1. Liposomes prepared with Soya Lecithin (LP-05) and

Egg Lecithin (LP-11) ------------------------------------------- 166

6.6.2. Plasma Concentration of Liposomal formulations of DFP---------- 173

6.6.2.1. Liposomes prepared with PSL (LP-17) and

Egg Lecithin (LP-23) --------------------------------------173

6.6.3. Pharmacokinetic parameters of Liposomal formulations------------190

6.6.3.1. Liposomes of DFS prepared with PSL (LP-05) and

Egg Lecithin (LP-11) --------------------------------------190

Page 16: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xvi

6.7. Plasma Level Time Profile of Liposomal Formulation of DFS and DFP-----

--------------------------------------------------------------------------------------199

6.7.1. Plasma level time profile of diclofenac sodium (DFS) with purified

soya lecithins (PSL) and purified egg lecithins (PEL)----------------199

6.7.2. Comparison of plasma concentration of Liposomal formulations of

diclofenac sodium with Phlogin-SR®------------------------------------199

6.7.3. Plasma level time profile of diclofenac potassium (DFP) with

purified soya lecithins (PSL) and purified egg lecithins (PEL)----200

6.7.4. Comparison of Plasma Concentration of Liposomal Formulations of

Diclofenac Potassium with Caflam®

-Novartis Pharmaceuticals----201

6.8. Pharmacokinetics Profile of Liposomal Formulation of Diclofenac Sodium and

Diclofenac potassium -------------------------------------------------------------------202

6.8.1. Pharmacokinetics Profile of Liposomal Formulations of Diclofenac

Sodium------------------------------------------------------------------------------202

6.8.2. Pharmacokinetics Profile of Liposomal Formulations of Diclofenac

potassium---------------------------------------------------------------------------211

6.9. Conclusion-------------------------------------------------------------------------------215

Page 17: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xvii

LIST OF TABLES

Table

No. Contents

Page

No.

3.1 With-in-Day Accuracy and Precision of Analgesics in Drug Solutions

49

3.2 With-in-Day Accuracy and Precision of Analgesics in Standard Drug

50

3.3 Percentage Contents Found in Liposomes, Microcapsules and Marketed Brands

50

3.4 Standard Curve Parameters of Analgesics in Plasma 53 3.5 Extraction Efficiency of Analgesics in Plasma 54 3.6 Between-the-Day Accuracy and Precision in Plasma 55 3.7 Between-the-Day Accuracy and Precision in Plasma 55

3.8 Long Term Stability of Analgesics in Standard Drug Solutions

56

3.9 Freeze-Thaw Stability of DFS in Plasma 57 3.10 Freeze-Thaw Stability of FLP in Plasma 57 3.11 Freeze-Thaw Stability of TMD in Plasma 58

4.1 Standard curve parameters of DP (a) and MLX (b) in plasma samples

70

4.2 Inter-day Precision and Accuracy of DP and MLX in human Plasma

72

4.3 Intra-day Precision and Accuracy of DP and MLX in human Plasma

73

4.4 Recovery (Percent Extraction Yield) of DP (a) and MLX (b) in plasma samples

74

4.5 Freeze-thaw stability DP (a) and MLX (b) in human Plasma

76

4.6 Bench top stability DP (a) and MLX (b) in human Plasma 77 4.7 Bench top stability DP (a) and MLX (b) in human Plasma 78

4.8 Application of method in analysis of liposomes, marketed brands and human plasma

78

5.1 The composition and observed response from randomized runs in central composite design (Coded values)

87

5.2 The composition and observed response from randomized runs in central composite design (Actual values) 87

5.3 Coded level values for X1 and X2 88 5.4 Standardized main effects of variable on the responses 88

5.5 Summary of results of model analysis and lack of fit for measured responses

89

5.6 Comparison of different methods of preparation for evaluation of characteristics of liposomes (n=3)

95

6.1 Composition of liposomal formulations of DFS and DFP (n=24)

116

Page 18: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xviii

6.2 Characterization of (n= 24) Liposomal Formulations of DFS and DFP (Mean ±SEM)

117

6.3 Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at initial time of stability studies

135

6.4 Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at one month period of stability studies

135

6.5 Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at two month period of stability studies.

136

6.6 Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at three month period of stability studies

136

6.7 Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at six month period of stability studies

137

6.8 In-vitro release parameters of liposomal formulations of diclofenac sodium and diclofenac potassium

161

6.9 f2-values of liposomal formulations calculated from drug release profile and compared with reference drug product Phlogin-SR®

164

6.10 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05) administered in the oral dose of 100 mg in healthy subjects (n=18)

173

6.11 Plasma level time profile of oral formulation of Liposomes of DFS (LP-11) administered in the oral dose of 100 mg in healthy subjects (n=18)

174

6.12 Plasma level time profile of sustained release marketed brand of DFS (Phlogen SR®) administered in the oral dose of 100 mg in healthy subjects (n=18)

175

6.13

Comparison of plasma level time profile of oral formulation of liposomes (LP-05, LP-11) and marketed brand of DFS (Phlogen-SR® Brooks Pharma) administered in the oral dose of 100 mg to healthy subjects (n=18)

177

6.14

Plasma level time profile of oral formulation of Liposomes of Diclofenac potassium (LP-17) administered in the oral dose of 50 mg to healthy subjects (n=18

185

Page 19: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xix

6.15 Plasma level time profile of oral formulation of Liposomes of Diclofenac potassium sodium (LP-23) adminis-tered in the oral dose of 50mg to healthy subjects (n=18)

186

6.16

Plasma level time profile of oral marketed brand of Diclofenac potassium (CAFLAM® Novartis Pharma) adm-inistered in the oral dose of 50 mg to healthy subjects (n=18)

187

6.17

Comparison of plasma level time profile of oral formulation of liposomes (LP-17, LP-23) and marketed brand of Diclofenac potassium(Caflam®-Novartis Pharma) adminis-tered in the oral dose of 50 mg to healthy subjects (n=8)

189

6.18 Bioavailability and pharmacokinetic parameters of oral formulation of Liposomes of DFS (LP-05) administered in the oral dose of 100 mg to healthy human subjects (n=18)

191

6.19 Bioavailability and pharmacokinetic parameters of oral formulation of Liposomes of DFS (LP-11) administered in the oral dose of 100 mg to healthy human subjects (n=18)

192

6.20 Bioavailability and pharmacokinetic parameters of Phlogin®-Brooks Pharmaceuticals, Pakistan) administered in the oral dose of 100 mg to healthy human subjects (n=18)

193

6.21

Comparison of (mean ± SEM) of Bioavailability and Pharmacokinetics of oral formulation of liposomes (LP-05, LP-11) and marketed brand of Diclofenac sodium (Phlogen-SR® Brooks Pharma) administered in the oral dose of 100 mg

194

6.22

Bioavailability and pharmacokinetic parameters of oral formulation of Liposomes of Diclofenac potassium (LP-17) administered in the oral dose of 50 mg to healthy human subjects (n=18)

195

6.23

Bioavailability and pharmacokinetic parameters of oral formulation of Liposomes of Diclofenac potassium (LP-23) administered in the oral dose of 50 mg to healthy human subjects (n=18)

196

6.24

Bioavailability and pharmacokinetic parameters of marketed brand of Diclofenac potassium (Caflam® Novartis Pharmaceuticals, Pakistan) administered in the oral dose of 50 mg to healthy human subjects (n=18)

197

6.25

Comparison of (mean ± SEM) of Bioavailability and Pharmacokinetics of oral formulation of liposomes (LP-05, LP-11) and marketed brand of Diclofenac potassium (Caflam® Novartis pharmaceuticals) administered in the oral dose of 50 mg to healthy subjects (n=18)

198

Page 20: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xx

LIST OF FIGURES

Fig. No. Contents Page No.

2.1 Types of liposomes based on surface modifications 12

2.2 Types of liposomes depending upon size and type of lamellae

13

2.3 Structural formula of Phosphatidyl Choline (major component of lecithin)

15

2.4 Structure of Cholesterol 16

2.5 Structure of poly ethylene glycol (PEG) 18 3.1 Structure of (a) DFS (b) TMD (c) FLP 44

3.2 Chromatograms showing (a) separation of analgesics in drug solution (b) plasma samples

48

3.3 Standard curve of (a) DFS (b) FLB and (c) TMD in plasma 51-52 4.1 Structural formula for DP (a) and MLX (b) 61

4.2 UV Scan of DP and MLX drug solutions in Diode Array Detector of HPLC

67

4.3 Separation of DP and MLX plasma samples on C 18 Column 67

4.4 Standard Curve of DP (a) and MLX (b) in spiked human plasma

68-69

5.1 Linear correlation plots between Predicted vs. Actual Response values for response Y

91

5.2 Contour curve plot showing the influence of Cholesterol and Soya lecithin on response Y (Percent drug entrapment)

92

5.3 Response Surface plot showing the effect of Cholesterol and Soya lecithin on response Y (Percent drug entrapment)

92

5.4 Comparison of entrapment efficiencies obtained with different method of preparation of Liposomes.

96

5.5 Size range of the selected liposomal formulation (09) prepared with MEE method (Zetasizer, Malvern, UK)

96

5.6 Size range of the selected liposomal formulation (09) prepared with TLH method (Zetasizer, Malvern, UK

97

5.7 Size range of the selected liposomal formulation (09) prepared with REV method (Zetasizer, Malvern, UK)

97

5.8 Size range of the selected liposomal formulation (09) prepared with REV method (Zetasizer, Malvern, UK)

98

5.9 Zeta potential of the selected liposomal formulation (09) prepared with MEE method (Zetasizer, Malvern, UK)

98

5.10 Zeta potential of the selected liposomal formulation (09) Prepared with REV method (Zetasizer, Malvern, UK) 99

Page 21: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxi

6.1 Size of the liposomal formulation of DFS with PSL (LP-01) measured by Zetasizer (n=3) 118

6.2 Size of the liposomal formulation of DFS with PEL (LP-07) measured by Zetasizer (n=3)

118

6.3 Size of the liposomal formulation of DFP with PSL (LP-13) measured by Zetasizer (n=3)

119

6.4 Size of the liposomal formulation of DFP with PEL (LP-19) measured by Zetasizer (n=3)

119

6.5 Scanning electron microscopic images of selected liposomal formulation of DFS (LP-01)

120

6.6 6.5: Zeta potential of the liposomal formulation of DFS with PSL (LP-01) measured by Zetasizer (n=3)

122

6.7 6.6: Zeta potential of the liposomal formulation of DFS with PEL (LP-07) measured by Zetasizer (n=3)

122

6.8 6.7: Zeta potential of the liposomal formulation of DFP with PSL (LP-07) measured by Zetasizer (n=3)

123

6.9 6.8: Zeta potential of the liposomal formulation of DFP with PEL (LP-19) measured by Zetasizer (n=3)

123

6.10 DSC of selected formulations of DFS liposomes 126 6.11 XRD of selected formulations of DFS liposomes 128 6.12 FT-IR of selected formulations of DFS liposomes 130

6.13 Zero order release model of oral liposomal formulations of diclofenac sodium

143

6.14 First order release model of oral liposomal formulations of diclofenac sodium.

143

6.15 Higuchi release model of oral liposomal formulations of diclofenac sodium.

143

6.16 Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

144

6.17 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

144

6.18 Zero order release model of oral liposomal formulations of diclofenac sodium.

144

6.19 First order release model of oral liposomal formulations of diclofenac sodium

145

6.20 Higuchi release model of oral liposomal formulations of diclofenac sodium 145

6.21 Hixon Crowell release model of oral liposomal formulations of diclofenac sodium

145

6.22 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

146

6.23 Zero order release model of oral liposomal formulations of diclofenac sodium

146

6.24 First order release model of oral liposomal formulations of diclofenac sodium 146

Page 22: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxii

6.25 Higuchi release model of oral liposomal formulations of diclofenac sodium

147

6.26 Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

147

6.27 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

147

6.28 Zero order release model of oral liposomal formulations of diclofenac sodium.

148

6.29 First order release model of oral liposomal formulations of diclofenac sodium

148

6.30 Higuchi release model of oral liposomal formulations of diclofenac sodium

148

6.31 Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

149

6.32 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium

149

6.33 Zero order release model of oral liposomal formulations of diclofenac potassium

149

6.34 First order release model of oral liposomal formulations of diclofenac potassium.

150

6.35 Higuchi release model of oral liposomal formulations of diclofenac potassium

150

6.36 Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

150

6.37 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac potassium

151

6.38 Zero order release model of oral liposomal formulations of diclofenac potassium

151

6.39 First order release model of oral liposomal formulations of diclofenac potassium.

151

6.40 Higuchi release model of oral liposomal formulations of diclofenac potassium.

152

6.41 Hixon Crowell release model of oral liposomal formulations of diclofenac potassium

152

6.42 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

152

6.43 Zero order release model of oral liposomal formulations of diclofenac potassium.

153

6.44 First order release model of oral liposomal formulations of diclofenac potassium.

153

Page 23: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxiii

6.45 Higuchi release model of oral liposomal formulations of diclofenac potassium.

153

6.46 Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

154

6.47 Pappas release model (first 60% drug release) of oral liposomal formulations of diclofenac sodium.

154

6.48 Zero order release model of oral liposomal formulations of diclofenac potassium.

154

6.49 First order release model of oral liposomal formulations of diclofenac potassium.

155

6.50 Higuchi release model of oral liposomal formulations of diclofenac potassium

155

6.51 Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

155

6.52 Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

156

6.53 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogin SR® administered in the oral dose of 100 mg in Subject No. 1.

167

6.54 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 2

167

6.55 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 3.

167

6.56 Plasma level time profile of oral formulation of Liposome of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100mg in Subject No. 4

168

6.57 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP- 11) and Phlogin SR® administered in oral dose of 100 mg in Subject No. 5.

168

6.58 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogin SR® administered in oral dose of 100 mg in Subject No. 6.

168

6.59 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 7.

169

6.60 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 8.

169

6.61 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP- 11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 9.

169

6.62 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 10.

170

Page 24: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxiv

6.63 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No.11

170

6.64 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 12.

170

6.65 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 13

171

6.66 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 14.

171

6.67 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 15.

171

6.68 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in the oral dose of 100 mg in Subject No. 16

172

6.69 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in the oral dose of 100 mg in Subject No. 17.

172

6.70 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogen SR® administered in oral dose of 100 mg in Subject No. 18

172

6.71 Plasma level time profile of oral formulation of Liposomes of DFS (LP-05) administered in the oral dose of 100 mg in healthy subjects (n=18)

176

6.72 Plasma level time profile of oral formulation of Liposomes of Diclofenac sodium (LP-11) administered in oral dose of 100 mg in healthy subjects (n=18)

176

6.73 Plasma level time profile of sustained release marketed brand of DFS (Phlogin SR®) administered in the oral dose of 100 mg in healthy subjects (n=18)

176

6.74

Comparison of plasma level time profile of oral formulation of liposomes (LP-05, LP-11) and marketed brand of DFS (Phlogen-SR® Brooks Pharma) administered in the oral dose of 100 mg to healthy subjects (n=18)

177

6.75 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 1.

179

6.76 Figure 6.76: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 2

179

6.77 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 3.

179

Page 25: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxv

6.78 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 4

180

6.79 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 5.

180

6.80 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 6.

180

6.81 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 7

181

6.82 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP 23) and Caflam® administered in oral dose of 50 mg in Subject No. 8.

181

6.83 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 9.

181

6.84 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 10.

182

6.85 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 11.

182

6.86 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 12.

182

6.87 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP- 23) and Caflam® administered in oral dose of 50 mg in Subject No. 13

183

6.88 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 14.

183

6.89 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 15

183

6.90 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 16.

184

6.91 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 17.

184

6.92 Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 18.

184

Page 26: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxvi

6.93 Plasma level time profile of oral formulation of Liposomes of DFP (LP-11) administered in the oral dose of 50 mg to healthy subjects (Sum, n=18)

188

6.94 Plasma level time profile of oral formulation of Liposomes of DFP (LP-23) administered in the oral dose of 50 mg to healthy subjects (Sum, n=18)

188

6.95 Plasma level time profile of oral marketed brand of DFP (CAFLAM® Novartis Pharma) (LP-05) administered in the oral dose of 50 mg to healthy subjects (Sum, n=18)

188

6.96

Comparison of plasma level time profile of oral formulation of liposomes (LP-17, LP-23) and marketed brand of DFP (CAFLAM® Novartis Pharma) administered in the oral dose of 50 mg to healthy subjects (n=18)

189

Page 27: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxvii

LIST OF IMPORTANT ABBREVIATIONS

S. No. Term Description

1 DFS Diclofenac sodium 2 DFP Diclofenac potassium 3 PSL Purified soya lecithin 4 PEL Purified egg lecithin 5 FLP Flurbiprofen 6 TMD Tramadol HCl 7 MLX Meloxicam 8 FDA Food and drug administration 9 ICH International conference on harmonization 10 RSM Response surface methodology 11 CH Cholesterol 12 LP Liposomal formulation 13 CCD Central composite design 14 MEE Micro-emulsification evaporation 15 SEM Scanning electron microscopic studies 16 DSC Differential scanning calorimetry 17 XRD X-ray diffractometry 18 FT-IR Fourier transform infra-red spectroscopy 19 RP Reversed phase 20 HPLC High performance liquid chromatography 21 ODDS Oral drug delivery system 22 NSAIDs Nonsteroidal anti-inflammatory drugs 23 PEG Poly ethylene glycol 24 PC Phosphotidyl choline 25 PE Phosphatidylethanolamine 26 PI Phosphotidylinositol 27 ZTO Zedoary turmeric oil 28 FD Freeze-drying 29 BSA Bovine serum albumin 30 PIN Pinacyanol chloride 31 MLV Multilamellar vesicles 32 SCID Combined immuno-deficient 33 RES Reticulo endothelial system 34 BPA Boronophenylalanine

35 DOPE Dioleoyl phosphatidyl ethanolamine

Page 28: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxviii

36 ODNs Oligo deoxy nucleotides 37 TPLs Total polar lipids 38 SEC Size exclusion chromatography 39 PCS Photon correlation spectroscopy 40 hLf Human lactoferrin 41 NIR Near infra red spectroscopy 42 MPS Mononuclear phagocyte system 43 5-A.L.A 5-amino levulinic acid 44 HMM Hexa methyl melamine 45 NaDC Sodium deoxycholate 46 TMC N-trimethyl chitosan 47 HGF Hepatocyte growth factor 48 SSL Sterically stabilized liposomes 49 NPC Non parenchymal cells 50 HSC Hepatic satellite cells 51 DRV Dried re-hydrated vesicles 52 ELISA Enzyme linked immuno sorbent assays 53 ODS Octa decyl silane 54 SPE Solid phase extraction 55 PEY Percent extraction yield 56 TLH Thin layer hydration method 57 REV Reverse phase evaporation method 58 LOQ Limit of quantification 59 LOD Limits of detection 60 LDDS Liposomal drug delivery systems 61 BCS Biopharmaceutics classification system 62 DLS Dynamic light scattering 63 Cmax Maximum concentration 64 Tmax Time for maximum concentration 65 AUC Area under the plasma concentration–time curve 66 AUMC Area under the first moment curve 67 MRT Mean residence time 68 T1/2 Half life 69 Ke Elimination rate constant 70 VD Volume of distribution 71 CLT Total body clearance 72 ANOVA Analysis of variance

Page 29: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

xxix

Formulation and In-vitro/In-vivo Evaluation of Oral Diclofenac sodium and Diclofenac potassium Liposomal Dosage Forms

Study Plan

New Simultaneous HPLC Method for determination of NSAIDs and Opioid Analgesic in advanced drug delivery system and human plasma

HPLC Method Development and Validation for the simultaneous determination of DFP and MLX in human Plasma

Optimization of Liposomal Formulations using Response Surface Methodology (R.S.M.)

Formulation and In-vitro/In-vivo Evaluation of Oral Diclofenac sodium and Diclofenac potassium Liposomal Dosage Forms

1. Tested for DFS, FLB and TMD. 2. Optimization studies were

performed to select chromatographic conditions for separation of drugs.

3. Validation studies were performed. 4. Stability studies were also

performed.

1. Tested for DFP and MLX. 2. Optimization studies were

performed to select chromatogram-phic conditions for separation of drugs.

3. Validation studies were performed. 4. Stability studies were also

performed.

1. Three methods were applied for preparation of liposomes as MEE, TLH and REV using formulation pattern of central composite design (C.C.D).

2. Two independent variables as Cholesterol (X1) and soya lecithin (X2) were selected at five levels of α=1.267 against the dependant variable (Y) taken as percent entrapment.

3. Validation of Model. 4. Optimization of Model.

1. Formulation development by MEE followed by freeze drying

2. Characterization of liposomes

3. Determination of entrapment efficiency

4. Stability studies

5. In-vitro evaluation 6. In-vivo Evaluation

1. Linearity (r2) was 0.996, 0.998 and 0.995 for DFS, FLB and TMD, respectively.

2. Extraction efficiency was 99.60, 99.80 and 99.63 for DFS, FLB and TMD, respectively.

3. With-in-day Precision & Accuracy was 98.82, 99.53 and 99.43 for DFS, FLB and TMD, respectively.

4. Between-the-day Precision and Accuracy was 98.33, 99.64 and 99.60 for DFS, FLB and TMD, respectively.

5. Stability data showed that method was stable in long term and freeze-thaw studies with values of %CV were 0.35, 0.13 and 0.15at LQC for DFS, FLB and TMD, respectively.

6. %CV values of Freeze-Thaw studies were 1.82, 0.77 and 0.18 at HQC for DFS, FLB and TMD, respectively.

7. Applied in In-vitro and In-vivo studies of DFS liposomes.

1. Linearity (r2) was 0.9989 (DFP) and 0.9979 (MLX).

2. Extraction efficiency was 99.67 and 99.48 at HQC for DFP and MLX, respectively.

3. Inter-day Precision and Accuracy was 98.26 and 99.39 at HQC for DFP and MLX, respectively.

4. Intra-day Accuracy and Precision at was 99.22 and 99.25 for DFP and MLX, respectively.

5. Freeze-thaw stability values of %CV were 1.821 and 1.566 at LQC for DFP and MLX, respectively.

6. %CV values of Bench top stability values were 0.071 and 0.021 at HQC for DFP and MLX, respectively.

7. Long term stability values of %CV were 0.302 and 0.192 at HQC for DFP and MLX, respectively.

8. Applied in In-vitro and In-vivo studies of DFP liposomes

1. MEE method was found superior than TLH and REV highest entrapment using C.C.D.

2. ANOVA and estimated regression coefficient analysis was applied for determination of response (Y) based on two independent variables as Cholesterol (X1) and soya lecithin (X2).

3. Experimental results and predicted values were calculated using polynomial equation and plotted in linear correlation plots.

4. Two dimensional (contour) and three dimensional (Response surface) plots were obtained to show the optimum concentration of X1 and X2 as 25 and 75 for getting maximum entrapment of 82.56.

1. Twenty four formulations were developed successfully using fix amount of drug and varying concentration of lipids in ratio of (1:1), (1:2) and (1:3).

2. 5, 10 and 15% of PEG was added in selected formulations.

3. Particle size, PI and ZP were determined by Zetasizer and results were found satisfactory.

4. Entrapment studies were performed using developed analytical methods efficiency.

5. DSC, XRD and FTIR studies were performed on selected formulations

6. Selected formulations were tested for stability for analyzing percent content and time for 60% release.

7. All formulations were subjected to dissolution for In-vitro evaluation.

8. Selected formulations were evaluated in 18 human subjects for determining plasma conc. and PK parameters.

Page 30: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

0

OBJECTIVES OF THE RESEARCH

The following aims and objectives were made for this research

1. To develop oral liposomal formulations of diclofenac sodium and diclofenac

potassium.

2. Optimization and validation of formulations of liposomes by using statistical

method i.e. Response surface methodology (R. S. M).

3. To develop and validate new HPLC method for analysis of diclofenac sodium

in liposomal formulation and human plasma,

4. To develop and validate new HPLC method for analysis of diclofenac

potassium in liposomal formulation and human plasma

5. To evaluate physicochemical properties of developed liposomes using

different techniques like SEM, Zeta Potential, DSC, XRD and FTIR.

6. To evaluate stability of developed liposomal formulations.

7. To evaluate the release profile using dissolution studies of different developed

liposomal formulations and comparison with available brands in markets.

8. To evaluate and compare the plasma level profiles by In-vivo studies of

different developed formulations and comparison with available brands in

markets.

9. To evaluate and compare the pharmacokinetics and bioavailability parameters

of different developed formulations and comparison with available brands in

markets by In-vivo evaluation in human subjects.

10. To enhance the patient compliance and reduce site effects by administering

more bioavailable formulations.

xxx

Page 31: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

1

Abstract

The current study was aimed to develop stable and reproducible liposomal

formulations of Diclofenac sodium (DFS) and Diclofenac potassium (DFP)

using purified soya lecithin (PSL) and purified egg lecithin (PEL) for oral

delivery.

For the accomplishment of analysis task of DFS and DFP in In-vitro/In-vivo

evaluation as well as for entrapment studies, two simultaneous methods were

developed and validated. In one study for analysis of DFS, an isocratic system

was employed for the flow of mobile phase consisting of 10 mM sodium

dihydrogen phosphate buffer and acetonitrile in molar ratio of 67: 33 with

adjusted pH of 3.2. The stationary phase was hypersil ODS column (C18,

250×4.6 mm i.d., 5 µm) with controlled temperature of 30˚C. DFS in

liposomes, microcapsules and marketed drug products was determined in range

of 99.76-99.84%. FLP and TMD in microcapsules and brands formulation were

99.78 - 99.94 % and 99.80 - 99.82 %, respectively. Single step liquid-liquid

extraction procedure using combination of acetonitrile and trichloroacetic acid

(TCA) as protein precipitating agent was employed. The detection limits (at

S/N ratio 3) of quality control solutions and plasma samples were 10, 20, and

20 ng.mL-1 for DFS, FLP and TMD, respectively. The Assay was acceptable in

linear dynamic range. All other validation parameters were found in limits of

FDA and ICH method validation guidelines. The proposed method for DFS

analysis was found as sensitive, accurate and precise and applied for

Page 32: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

2

dissolution studies as well as in human plasma samples for bioequivalence and

pharmacokinetics studies.

For analysis of DFP, a new, easy and consistent reversed-phase high-

performance liquid chromatographic method with diode array detection was

developed and validated for DFP and MLX (Meloxicam. The optimized mobile

phase was used in the molar ratio of 20:20:60 (v/v/v) mixture of acetonitrile,

methanol and 20 x 10-3 M potassium dihydrogen phosphate buffer (pH 3.7),

pumped at an optimized flow rate of 1.0 mL.min-1. The linearity was performed

in the concentration range of 15 ng.mL−1 to 10µg.mL-1 with r2 values of

0.9989 ± 0.13 and 0.9979 ± 0.11 (n = 6) for DP and MLX, respectively. The

assay was repeatable at concentration levels of 10 ng.mL-1, 1 µg.mL-1 and 10

µg.mL-1 with coefficient of variation of 0.168 - 0.603% for 10 ng.mL-1 (DP),

15 ng.mL-1 (MLX) and 1 µg.mL-1 &10 µg.mL-1 for DP and MLX. The LOD

values were 0.3 and 0.5 ng.mL−1, while values of LOQ were 10 ng.mL-1 and

15 ng.mL-1, for DP and MLX. The present method was applied in advanced

drug delivery formulations (Liposomes), In-vitro and In-vivo studies.

An important part of study was development of an optimized liposomal

formulation of diclofenac sodium (DFS) of most suitable concentration of

formulating ingredients, soya lecithin (SL) and Cholesterol (CH) with

maximum entrapment efficiency. For this purpose, response surface

methodology (RSM) was used to optimize formulation variable. Cholesterol

was selected as independent variable 1 and designated as X1 while soya lecithin

was independent variable 2 designated as X2. The response was the entrapment

Page 33: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

3

of drug and designated as dependent variable Y. The two formulation

ingredients were ranged with central composite rotatable design (CCD) and

quadratic model at five levels (α=1.267) was followed for blending the

liposomal formulation. It was observed that cholesterol (variable 1) may

decrease the entrapment of DFS in the order of increasing concentration while

soya lecithin (variable 2) was found to increase entrapment (dependant

variable, Y) with increasing concentration. The central composite design has

resulted in an optimized formulation (Formulation No. 9) with an optimum

concentration of cholesterol and soya lecithin (ratio of 25:75) with maximum

of entrapment of 82.56%. The study was also extended to compare different

methods employed for the preparation of liposomes using optimized

formulation by RSM. It was concluded that formulation prepared by micro-

emulsification evaporation (MEE) followed by freeze drying method showed

maximum entrapment of DFS.

A comprehensive study was conducted for development of liposomal

formulations of DFS and DFP with variable concentrations of purified soya

lecithin (PSL) and purified egg lecithin (PEL) employing micro-emulsification

evaporation method (MEE) followed by freeze drying. The prepared liposomes

were free flowing and of uniform particle size distribution in the rage of 1.01 ±

0.011 to 1.80 ± 0.008 µm for DFS liposomal formulations while the mean size

of (mean ± SEM) 1.94 ± 0.008 µm for diclofenac potassium (DFP). The

selected liposomal formulations of DFS and DFP were also characterized by

using scanning electron microscopic studies (SEM), differential scanning

Page 34: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

4

calorimetry (DSC), x-ray diffractometry (XRD) and fourier transform infra-red

spectroscopy (FT-IR). Drug entrapment efficiency was above 82%. The

entrapping efficiency and in-vitro release of DFS and DFP of all liposomal

formulations were determined by reversed phase high-performance liquid

chromatography (RP-HPLC). Different kinetics models of in-vitro were

applied and release of DFS and DFP from liposomal formulations of DFS and

DFP and it was concluded that release followed higuchi model and relatively

zero order release, calculated on the basis of r2 value of straight line fit

equation. A sustained release was observed for 16-24 hours from all range of

liposomal formulations. The selected formulations after similarity factor (f-2)

were subjected to in-vivo evaluation in eighteen healthy human subjects.

Present study results in new formulation of DFS and DFP using PSL and PSL

for oral delivery, which was found stable, reproducible and sustained release by

using modified micro-emulsification evaporation method (MEE) followed by

freeze drying which was found a probable and better to produce liposomes for

oral drug delivery system (ODDS).

Keywords: Liposomes; Phospholipids; Diclofenac sodium; Diclofenac potassium; Validation; Response surface Methodology (RSM), micro-encapsulation vesicle method (MEE); In-vitro Release; Kinetics Models; Higuchi Model; In-vivo studies.

Page 35: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

5

Chapter No. 1

1. Introduction

Modern research is aimed at development of drug delivery system with

maximum therapeutic benefits for safe and effective management of disease(s).

The concept is based on controlled drug delivery, biotechnology and polymer

sciences which surpass all the barriers of a disease. A probe in current research

on drug delivery systems to maximize therapeutic performance eliminates

undesirable side effects of drugs and increases patient compliance. The ideal

drug delivery system delivers the drug at a rate dictated by the needs of body

over the period of treatment and it channels the active entity to the site of

action (William et al., 2010; Dey et al., 2008).

The idea of a drug carrier system with target specificity has fascinated

scientists and tremendous efforts have been made to achieve this goal. Paul

Ehrlich in 1906, initiated era of development for the drug delivery mechanism

that would target to the diseased cell. Since then numerous attempts have been

made to devise clinically effective drug delivery systems. Numbers of carriers

were utilized to carry the drugs at target tissues/organs which include

liposomes (Zuzanna and Agata, 2010), nanoparticals (Wim and Paul, 2008;

Svetlana et al, 2005 micro-beads (Sunita and Lalet, 2008; Zecheru et al., 2006),

erythrocytes (Mehrdad and Hosnieh, 2003; Ulrich and James, 1997), niosomes

(Dubey et al., 2010; Cosco et al., 2009; Gyatri et al., 2000), microcapsules

(Sajeev et al., 2002), microspheres (Wang et al., 2007; Fischer et al., 2006),

pharmacosomes (Semalty et al., 2009), multiple emulsions (Naveed et al.,

Page 36: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

6

2010) and nanosuspension (Jiraporn, 2007; Barrett., 2004). The evaluation of

targeted drug delivery is at a primordial stage nevertheless some practical

applications have emerged.

Amongst various carriers, few drug carriers reached the stage of clinical trials

where phospholipids based vesicles (Liposome) showed strong potential for

effective drug delivery to the site of action. These are biologically inert in

nature, devoid of any antigenic, pyrogenic and allergic reactions and their

components can be utilized as the components of biological membrane. Drugs

incorporated in liposomes are not inactivated under physiological conditions

and donot produce unfavourable side effects as well (Zuzanna and Agata, 2010;

Kshirsagar et al., 2005; Martin, 1995).

Liposomes are simple microscopic vesicles in which aqueous volume is

entirely enclosed by a membrane composed of lipids. Various amphipathic

molecules have been use to form liposomes. The drug molecules are either

encapsulated in aqueous space or intercalated into the lipid bilayer. The exact

location of the drug will depend upon physicochemical properties and

composition of lipids (Tomoko and Ishii., 2005; Moreira et al., 1997).

Drugs of the analgesic, antipyretic and anti-inflammatory class includes a

heterogeneous group of compounds, referred to as the nonsteroidal anti-

inflammatory drugs (NSAIDs). Many of these agents affect pain, fever and

inflammation and consequently, they are used widely for minor aches and

pains, headaches, the general illnesses, and to alleviate symptoms of rheumatic

fever, arthritis, gout, and other musculoskeletal disturbances (Sweatman, 2009;

Page 37: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

7

Laurence, 2007). Diclofenac is NSAID, used mainly as the sodium and

potassium salts for the relief of pain and inflammation in various conditions

like musculoskeletal and joint disorders such as rheumatoid arthritis,

osteoarthritis and ankylosing spondylitis. Peri-articular disorders such as

bursitis and tendonitis, sift-tissue disorders such as sprains and strains and other

painful conditions such as renal colic, acute gout, dysmenorrheal and following

some surgical procedures. It has also been used in some countries for the

management of fever (Arcelloni et al., 2001).

The present study was aimed to formulate oral liposomal dosage forms of

diclofenac sodium (DFS) and diclofenac potassium (DFP) using two types of

phospholipids named as purified soya lecithin (PSL) and purified egg lecithins

(PEL). In the first phase of study, new analytical methods using sensitive and

latest techniques of analysis (HPLC) in pharmaceutical dosage forms, advanced

delivery systems and human plasma were developed and validated. Two

separate studies were conducted for development of analytical assays of DFS

and DFP. The developed methods were found appropriate to achieve higher

sensitivity and accuracy for accomplishment of set tasks. These methods can

also be applied for routine quality control operations and in clinical conditions

for evaluation of bioavailability, pharmacokinetics and therapeutic drug

monitoring. To accomplish the task of formulation of oral liposomes, a primary

study was conducted by applying statistical design known as central composite

rotatable design (C.C.D) of response surface methodology (R.S.M) in order to

optimize the concentration of formulating ingredients of liposomes. This part

Page 38: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

8

of the study resulted out in selection of appropriate concentration of

formulation variables purified soya lecithin (PSL) and cholesterol (CH) after

developing thirteen formulations and analyzing the entrapment efficiency. It

was observed that formulation showed maximum response (entrapment of

DFS) having an appropriate concentration ratio of (75:25) for both (PSL) and

(CH), respectively. In this primary study, different methods for preparation of

liposomes were also applied and compared for achieving maximum entrapment

of DFS in liposomes. It was concluded that micro-emulsification evaporation

method (MEE) followed by freeze-drying was found most appropriate method

for preparation of liposomes of desired physicochemical characteristics and

maximum entrapment of drugs.

The extensive study was than conducted for preparation of liposomes of both

analgesic drugs (DFS and DFP) using both of phospholipids PSL and PEL. The

increasing concentration of PSL and PEL was also studied. The study was

further extended to note the effect of poly ethylene glycol (PEG) on stability

and residence time of drug in body. Twenty four formulations were developed

for this part of study. All formulations were subjected to evaluate

physicochemical properties like particle (vesicle) size, morphology,

polydispersity index, zeta potential and entrapment efficiencies using latest

techniques and updated methods. Various techniques like differential scanning

calorimetry (DSC), x-rays diffractometry (XRD) and fourier transform infra-

red spectrophotometery (FT-IR) for evaluation of interaction between drugs

and phospholipid contents of liposomes. It was observed that liposomal

Page 39: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

9

formulations were stable and having no interactions between formulation

components. The entrapment efficiencies were also determined in all

formulations. It was observed that PEL formulations have relatively higher

entrapment efficiencies than PSL formulations. Stability studies were also

conducted and it was observed that liposomes formulations were stable over

the entire experimental conditions.

An important part of the study was in-vitro evaluation of liposomal

formulations. Twenty four formulations were subjected to dissolution studies

for evaluation of release profile in experimental conditions to predict and

ensure its performance in biological system. The data of release profile was

gathered using developed methods for DFS and DFP. It was concluded from

these studies that liposomal formulations followed zero order (approximate)

and higuchi model of release kinetics.

The last part of the study was in-vivo evaluation of selected liposomal

formulations. After the results of f-2 analysis (similarity determination) with

market brand of DFS (Phlogin-SR®-Brooks Pharmaceutical, Pakistan), DFS

liposomal formations (LP-05 and LP-11) were selected for in-vivo studies. LP-

05 was formulated with (150:50) ratio of purified soya lecithin (PSL) and

cholesterol (CH), 100 mg of DFS and 10 % (30 mg) of PEG where as LP-11

was formulated with (150:50) ratio of purified egg lecithin (PEL) and

cholesterol (CH), 100 mg of DFS and 10 % (30 mg) of PEG.

In-vivo studies were performed in eighteen healthy human volunteers after

completing all requirements of inclusion/exclusion criteria. Written informed

Page 40: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

10

consent was taken from all subjects and administered three formulations

according to the Latin-Square design. HPLC method for both DFS was used to

calculate plasma levels of each subject at all data point. Pharmacokinetics

parameters were calculated according to the non-compartmental method of

analysis using pharmacokinetics software Kinetica®-version 4.4. Similarly,

DFP liposomal formulations were also evaluated in 18 human subjects and

compared with drug product (Caflam® Novartis Pharmaceutical, Pakistan) as

reference brand. HPLC method which was developed and validated for DFP

was used to determine plasma levels of each subject at all data points.

Pharmacokinetics parameters were calculated according to the non-

compartmental method of analysis. It was found that liposomal formulations of

DFS showed relatively greater rate and extent of drug absorption than reference

sustained release market brand (Phlogin-SR® Brooks Pharmaceutical,

Pakistan). The DFP liposomal formulations were also having higher rate and

extent of drug absorption and significant higher bioavailability than non

sustained release market brand (Caflam®-Novartis Pharmaceutical, Pakistan).

The liposomal formulations of DFS and DFP showed higher residence time in

the body, ensuring the effect of PEG on increased circulation of liposomes.

Page 41: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

11

2. Chapter No. 2

2.1 Structure and properties of liposomes

Liposomes are structural vesicles consisting of one or more phospholipid

bilayer surrounding a water space for entrapping water soluble and insoluble

compounds. These are also employed for improving solubility of hydrophobic

drug as well enhancing stability (Paola et al., 2000). The diameter of liposome

varies from 0.02 to 10 µm. Vesicle formulations are usually based on natural

and synthetic phospholipids and cholesterol. The structure may also possess

lipoproteins (Ulrich, 2002). The physicochemical properties of liposomes can

be modified by changing:

i. the types of lipids;

ii. the composition and proportions of lipids in the liposomal formulation;

iii. the size of liposome;

iv. the charge of liposomal surface: positive, negative, or neutral;

v. pH sensitivity;

vi. temperature sensitivity;

vii. the fluidity of liposomal membrane: rigid and fluid liposomes

(Zuzanna, 2010; Martin, 1995).

Liposomes as carriers have certain advantages:

1. These formulations increase the solubility and/or stability of drugs in

vivo thereby improving physicochemical characterization and

subsequent mounting in-vivo kinetics and biodistribution. An example is

Page 42: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

12

the increased solubility and bioavailability of low soluble drug (e.g.

silymarin) by formulating in liposomal drug delivery system.

2. This delivery system is meant for reduced unwanted or toxic effects by

decreasing toxicity of drugs. An example is the liposomal delivery of

gentamicin which has neurotoxicity and nephrotoxicity.

3. Liposomes can be targeted at any site in the body with different

formulation variables such as addition of PEG, antibodies, making pH

sensitive and temperature sensitive drug delivery systems.

4. Liposomes can show enhanced activity against extra cellular pathogens

to overcome bacterial drug resistance.

The variety of liposomal formulations allows the design of effective antibiotic

forms and subsequent therapeutic success (Zuzanna et al., 2010; Cristina, 1996;

El-Samaligy, 2006)

Figure 2.1: Types of liposomes based on surface modifications Source: Zuzanna et al., (2010)

Page 43: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

13

Figure 2.2: Types of liposomes depending upon size and type of lamellae (Source: Amarnath and Uma., 1997)

2.2 Structural components of Liposomes

2.2.1 Soya Lecithin

Lecithin is a complex mixture of phospholipids and other materials. The

chemical name as given in some literature is 1, 2-diacyl-sn-glycero- 3-

phosphocholine (trivial chemical name, phosphatidylcholine). The composition

of lecithin (and hence also its physical properties) varies largely based upon the

origin of lecithin and the extent of purification. Egg lecithin, for example,

contains 69% phosphatidylcholine and 24% phosphatidyl-ethanolamine, while

soybean lecithin contains 21% phosphatidylcholine, 22%

phosphatidylethanolamine, and 19% phosphatidylinositol, along with other

components. Lecithins are soluble in hydrocarbons, halogenated hydrocarbons,

mineral oil and fatty acids. They are practically insoluble in cold vegetable and

animal oils, polar solvents, and water. When mixed with water, lecithins

Page 44: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

14

hydrate to form emulsions. The lecithins have wide applications in the field of

cosmetics, pharmaceuticals and food products (Raymond et al., 2006).

Physical properties are variable, from viscous semi-liquids to powders, based

upon the free fatty acid content. There is variability in color from brown to

light yellow, depending upon whether they are bleached or unbleached or on

the degree of purity. When they are exposed to air, rapid oxidation occurs, also

resulting in a dark yellow or brown color. Lecithins have practically no odor.

Those derived from vegetable sources have a bland or nutlike taste, similar to

that of soybean oil.

Soya lecithin is composed of several kinds of phospholipids including

Phosphotidyl choline (PC), phosphatidyl ethanolamine (PE) and phosphatidyl

inositol (PI). It is largely extracted from soybean oil. The soya bean are placed

at optimum temperature and moisturized environment for about 7 to 10 days

and then subjected to pressurized pressing so that the flakes are obtained giving

rise to soy bean oil. Now, soya lecithin can be purified by steam precipitation

or by adding water and then centrifuging the oil to yield required product. The

concentration of lecithin is variable based upon the type of formulation. 0.15 in

aerosol preparation, 0.3- 2.3% in IM injections and about 0.25 – 10.0 % in oral

suspension are the examples (Rousset et al., 2010; Paz et al., 2010; Raymond et

al., 2006).

Soya lecithin has wide spread application in food and pharmaceutical industry.

It is best known in pharmaceutical industry due to its emulsifying properties. It

is widely used in targeting the drug to its active site due to its lamellar

Page 45: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

15

arrangement forming vesicle structures. It has been investigated as a solubilizer

for enzymes acyltransferase to estimate the lipoprotein metabolism in animal

models by Rousset et al., (2010).

Its potential utilization as an extender/ release retardant was evaluated by Paz et

al., (2010). Soya lecithin can also act as a stabilizing agent when its effect were

studied on mechanical properties of HPMC film, the thickness and deformation

curves were remained unaffected by incorporation of lecithin (Bajdik et al.,

2009). Only a slight variation was seen when lecithin content were exceeded

than 5%. Similarly, the biocompatibility of soya lecithin nanoparticles was also

studied using cholesterol and modified lecithin (Chuah et al., 2009).

P O

O

O

HO

N

H2C

CH

O

C O

CH2

O

CO

Figure 2.3: Structural formula of Phosphatidyl Choline (major component of lecithin)

Page 46: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

16

2.2.2 Cholesterol

Cholesterol (mol. formula C27H45OH and mol. wt. 386.65) is white or faintly

yellow, almost odorless powder or granules. It attains yellow to tan color when

exposed to light or air for longer period of time. The boiling point of

cholesterol is about 360 ºC with a melting point in range of 147-150 ºC.

Cholesterol is practically insoluble in water but highly soluble in acetone and

chloroform (Raymond et al., 2006). The structure of cholesterol is given in

Figure 2.4.

HO

Figure 2.4: Structure of Cholesterol

Cholesterol is widely used in liposomal formulation as a hardening agent

because it imparts structural integrity to the lamellar membrane of liposome.

Commercially it is produced by extracting spinal cord of cattle’s with

petroleum ether, but also a sufficient amount can be obtained from wool fat.

Repeated bromination leads to highly pure drug. Now synthetic mode of

preparation is also very much accountable. Cholesterol also has its major role

as a sterol in living organisms, being present in all body tissues, in brain and

spinal cord. It may also provoke the formation of gallstones. Unilamellar

liposomes when formulated using cholesterol demonstrate an increase in the

trans membrane movement of hydrophobic drugs (Edward and Efraim, 1989;

Page 47: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

17

Raymond et al., 2006). The presence of cholesterol may alter the particle size

and physical stability of liposomal membranes. Therefore, optimum cholesterol

along with other membrane forming ingredients should be selected for best

characteristics of liposomal membranes.

It is also mentioned in literature that cholesterol can be incorporated into

phospholipid membranes in very high concentration up to 1: 1 or even 2:1

molar ratios to PC but does not itself form bi-layers structure. Cholesterol

oriented towards the aqueous surface through hydroxyl group & the aliphatic

chain aligned parallel to acyl chains in the center of bi-layers. A cholesterol

incorporation increase the separation between choline head groups & reduce

the values of zeta potential by plummeting the normal electrostatic & hydrogen

bonding interactions. The high solubility of cholesterol in phospholipid

liposomes has been attributed to both hydrophobic & specific head group

interaction (Horwitz et al., 1999; Yeagle et al., 1990; Ladbrook et al., 1968)

2.2.3 Poly Ethylene Glycol (PEG)

The synonyms of PEG are Carbowax; Carbowax Sentry; Lipoxol; Lutrol E.

Chemically, it is Hydro-o-hydroxypoly (oxy-1,2-ethanediyl). The empirical

formula is HO-CH2-(CH2-O-CH2)m-CH2-OH where “m” represents the average

number of oxy-ethylene groups. Alternatively, the general formula H-(O-CH2-

CH2)n-OH may be used to represent polyethylene glycol, where n is a number

of oxy ethylene groups (Raymond et al., 2006).

Poly Ethylene Glycol (Figure 2.5) is abundantly used in many pharmaceutical

preparations including parenteral, topical, ophthalmic, oral and rectal

Page 48: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

18

preparations. It has also been used as biodegradable polymers for controlling

the release rate of drug.

C CH2HO

H

H

O CH2 C

H

OH

H

n

Figure 2.5: Structure of poly ethylene glycol (PEG)

In liposomal formulations, PEG acts as a stabilizing agent (stealth liposomes)

and also imparts flexibility to the bilayer membranes of the phospholipids

(Wang et al., 2010). The grades of the PEG may range from 200 – 8000 based

upon poly oxy ethylene units attached with basic structure. Grades in range of

200-600 are liquids, while grades above 1000 are solids. Liquid grades occur as

clear colorless or these may be slightly yellowish viscous liquids. Solid grades

have great variations in color ranging from white to off-white with physical

appearance ranging from pastes to waxy flakes. PEGs grades above 6000 are

present as free flowing finely divided powders (Parampal and Khuller, 1997;

Sakae et al., 1995; Raymond et al., 2006). Melting points ranges from 37 to 63

ºC . Melting point of PEG 2000 is given as (45-50 ºC). All grades of PEGs are

soluble in water, acetone, alcohols, benzene, glycerin and glycols. PEGs were

extensively studied for the potential delivery of anticancer drugs to liver by

covering the liposome with PEG-lipids and with glactosylated lipids. Proper

Page 49: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

19

proportions of various grades of liposomes can impart a smart role in

modulation of required characteristics in a formulation (Manconi et al., 2007).

2.3 Liposomal formulations of different drugs using different techniques

Zhiwen et al., (2010) formulated liposomes of zedoary turmeric oil (ZTO) as

new formulation of a water-insoluble oily drug by freeze-drying (FD) using

TBA/water cosolvent systems. The TBA/water cosolvent systems were used to

investigate a feasible method with sugar/lipid mass ratio and TBA content

(concentration with efficient and less time consuming process leaving behind

very less concentration of solvent). Transmission electron microscopic studies

revealed smooth and spherical liposomes with an average particle size of 0.457

± 0.078 µm and the values of zeta potential were more than 3.65 MV. In-vivo

experimentation was conducted in rabbits and plasma level–time profiles with

superior sustained-release properties were accomplished while using oral route

with bioavailability of 257.7%. The results indicated that liposomes

formulations have highly significant improved bioavailability. It was concluded

that ZTO liposome vesicles have shown improved and enhanced bioavailability

via oral route.

In a study conducted by Cui-Tao et al., (2010) demonstrated a comparison of

encapsulation efficiency and ultrasound-triggered release for protein from

microbubbles and liposomes prepared bovine serum albumin (BSA). The

formulation ratio between BSA and HPC in microbubbles and liposomes was

1:5, 1:7 and 1:10, respectively. Encapsulation efficiency and ultrasound-

stimulated release profile were detected. The mean size of loaded microbubbles

Page 50: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

20

and liposomes was 3.4 mm and 1.7 mm, respectively. An inverse liaison was

observed between BSA and HPC for encapsulation efficiency of microbubbles.

More than 90% of the entrapped BSA was released microbubbles where as it

releases less than 5% liposomal formulations (BSA).

Jukui et al., (2010) prepared sodium deoxycholate (NaDC) liposomes by

reverse phase evaporation method for oral delivery. As a model drug (Hexa

methyl melamine, HMM) was chosen having anti-tumor properties. Particle

sizes, entrapment efficiency of pinacyanol chloride (PIN) spectral

characteristics with various molar ratio of NaDC/PC, as well as the vesicle

stability measurements were found in acceptable range (9.76 & 1.21 fold). It

was concluded that sodium deoxycholate (NaDC) liposomes can be used as

potential carrier for oral drug administration.

Martin et al. (2009) investigated the feasibility of preparing liposomes that are

coated with the multifunctional polymer poly (acrylic acid)-cysteine (PAA-

Cys). Cationic multilamellar vesicles (MLV) as well as cationic submicron-

sized liposomes were prepared and coated with polymer poly (acrylic acid)-

cysteine. The change of initial positive zeta potential value to the negative side

after coating pointed out the successful coating process. It was observed that in

the multi lamellar and submicron sized liposomes, the quantity of free thiol

groups was comparable to that of PAACys solution of the same concentration.

The entrapment of hydrophilic marker fluorescence-isothiocyanate 4 kDa (FD4)

was higher in PAA-Cys liposomes in comparison to non-coated liposomes but

found lower when compared with coating of unmodified poly (acrylic acid)

Page 51: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

21

(PAA). It was concluded in the study that coating with PAA-Cys and PAA on

liposomal formulations did not influence the release profile of FD4 and CF,

whereas the release profile was affected by the molecular mass of the marker

and the liposome size. The feasibility of liposomes as novel carrier system

accomplished an essential necessity for employing in oral drug delivery.

Maha et al., (2008) described vesicular delivery systems as local depot for

sustained action of drugs. Multilamellar liposomes and niosomes of

aceclofenac were prepared and evaluated. A comparative study was performed

for the evaluation of entrapment efficiency, particle size, shape, differential

scanning calorimetry and in vitro drug release. It was found that formulations

were stable for three months when vesicles were stored at (2-8 ºC). The results

of anti-inflammatory effect showed entrapment efficiency and in-vitro release

of aceclofenac from vesicles can be controlled by varying the cholesterol,

surfactant as well as the type of charge. The both vesicular systems showed

significant sustained anti-inflammatory activity compared to the marketed

product, with niosomes being superior to liposomes as manifested by both

oedema rate and inhibition rate percentages suggesting their effectiveness as

topical anti-inflammatory delivery systems.

According to the study of Danbo et al., (2006), a well-characterized novel

lyophilized, sterile and easy-to-use nimodipine liposomal formulation

composed of PC/CHOL 20:3 and co-surfactant poloxamer 188/ sodium

deoxycholate/ PC/ 3:0.3:5 was developed for further studies. After

lyophilization, 89.9% entrapment efficiency and particle size of 200 nm was

Page 52: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

22

obtained. After oral & IV administration in rabbits, liposomal formulations

produced noticeable differences compared to the commercial brands with an

increased value of AUC, Cmax & T (1/2). It was observed that values of liposomal

formulations of nimodipine were larger compared to the commercial brands. It

was concluded that liposomal formulation of nimodipine can be used as

substitute to the commercial brand of nimodipine.

Carla et al., (2004) studied the interactions of anti-inflammatory drugs

indomethacin and acemetacin between the bulk aqueous phase and the

membrane of egg phosphatidylcholine vesicles using Isothermal titration

calorimetry. It was observed that significant electrostatic binding exists

between charged drugs and the membrane which can be calculated using Gouy-

Chapman theory. The binding/partition mechanism was calculated using

partition coefficient, and/or the equilibrium constant. The electrostatic effect

was determined by the mathematical terms to find out partition coefficients and

binding constants. Calorimetric titrations conducted under a lipid/drug ratio

[100:1] lead to a constant heat release and were used to directly calculate the

enthalpy of the process, DH, and indirectly, DG and DS. It was observed that

as the lipid/drug ratio decreased, simple partition was no longer valid and

interaction phenomenon was interpreted in terms of binding isotherms. Another

important finding was the unique phenomenon of interactions in all studied

regimen at varied concentration of lipid/drug ratio, the extent of interaction, as

well as binding stoichiometry, is affected by the lipid/drug ratio. It was

concluded that the change in these parameters reflects the biphasic behavior of

Page 53: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

23

interaction-possibly the consequence of a modification of membrane’s physical

properties as it becomes saturated with the drug.

Gohel and Amin, (1997) developed lipospheres of diclofenac sodium by melt

dispersion technique using triple pressed stearic acid. Free flowing lipospheres

were obtained by congealing the micro-emulsion. The amount of water, Tween

20 (surfactant) and butyl alcohol (co-surfactant) were identified as the key

variables affecting the formation of discrete spherical lipospheres. More than

70 percent of the isolated lipospheres were of the size range 180-250 µm. The

amount of drug entrapped in the lipospheres was found to be dependent on

lipid to drug ratio and drug loading was futher increased by using caranuba

wax coated particles of diclofenac sodium. The in vitro drug release study was

conducted in phosphate buffer “pH 7.2” and dissolution of entrapped drug was

greatly retarded.

Els Etten et al., (1998) studied the clearance of liposomes from the circulation

to a substantial extent by phagocytic cells of the mononuclear phagocyte

system (MPS). At first, the effect of administration of three types of empty

liposomes (devoid of drug) resulted in difference in residence time and

clearance from blood was studied in mice. Classical liposomes designated as

(LIP A) and placebo liposomes with lipid composition as in AmBisome (LIP

B) or as in Doxil (LIP C) were administered intravenously as a single dose. The

blood clearance capacity of the MPS was monitored at different time points

after the last liposome injection. It was revealed that the carbon blood clearance

capacity of MPS was ruined only at a high lipid dose of empty classical

Page 54: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

24

liposomes. The bacterial blood clearance capacity was never impaired, not even

after prolonged treatment with AmBisome administered in a clinically relevant

regimen.

Naoto et al., (1993) described that liposomes are bio-degradable and essentially

non-toxic vesicles that can encapsulate both hydrophilic and hydrophobic

materials and are utilized as drug carriers for drug delivery systems (DDS).

Recent progress in gene technology provides a novel modality of therapy for

various diseases with a variety of newly developed cationic liposomes.

Delivery of agents to the reticulo endothelial system (RES) is easily achieved

since most conventional liposomes are trapped by the RES. For the purpose of

delivery of agents to target organs other than RES, long-circulating liposomes

have been developed by modifying the liposomal surface. The in-vivo

dynamics of liposome-carried agents is required to evaluate the bioavailability

of drugs encapsulated in liposomes. In-vivo trafficking of liposomes was

visualized by positron emission tomography (PET) and discusses the

characteristics of liposomes that affect the targeting of drugs in-vivo.

Franca et al., (2000) developed and stabilized the boronophenylalanine (BPA)-

loaded conventional liposomes by reversed phase evaporation method to treat

liver metastases using boron neutron capture therapy. Conventional vesicles

were composed of phosphatidylcholine and cholesterol, molar ratio 1:1. To

obtain stealth liposomes, GM or PEG was included in the lipidic bilayer at a

concentration of 6.67 or 5 mol%, respectively. Large unilamellar vesicles were

formulated encapsulating BPA in the liposome aqueous compartment as a

Page 55: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

25

complex with fructose; BPA free base also was embedded into the lipidic

bilayer. In vivo experiments were carried out after intravenous injection of

liposome suspensions in BD-IX strain rats in which liver metastases had been

induced. Alpha particle spectroscopy associated with histological analysis was

performed to visualize boron spatial distribution in liver. Simultaneously, tissue

boron concentrations were determined using inductively coupled plasma-mass

spectroscopy. Results showed that PEG-modified liposomes accumulated

boron in therapeutic concentrations (>30 µg boron/g tissue) in metastatic tissue.

The PEG-liposomes could be further explored in enhancing boron delivery to

tumor cells.

Kisoon et al., (2002) formulated a novel cationic cholesterol derivative, 3g [N-

(N', N’, N’-trimethyl amino propane) -carbamoyl] cholesterol iodide (Chol-Q)

with equimolar amounts of dioleoyl phosphatidyl ethanolamine (DOPE) into

stable unilamellar liposomes up to 100 nm in size for DNA delivery into

mammalian cells. When compared with similarly constituted liposomes

containing the tertiary analogue 3g [N-(N', N'-dimethyl amino propane)-

carbamoyl] cholesterol (Chol-T) in a band shift assay, liposomes displayed

similar DNA binding affinities and appeared to afford complete protection to

plasmid DNA against serum nuclease catalysed degradation at liposome: DNA

ratios (w/w) of 2.5:1, 5:1, and 10:1 in incubation mixtures containing 5% fetal

bovine serum at 37 ºC for 90 min. Chol-Q liposomes were found markedly less

toxic to cells in culture over a wide range of concentrations with cells

numbering 76% of untreated controls at 37.5 wg/mL complete medium in the

Page 56: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

26

human hepatocellular carcinoma line HepG2 and 75% at 30 wg/mL in cervical

carcinoma HeLa cells. At these levels of Chol-T liposomes, cell numbers were

37% and 15%, respectively. Gene transfer experiments with pSV2CAT and

pRS VCAT plasmids in HepG2 cells showed maximum efficiency at a Chol-Q

liposome: DNA ratio of 5:1 (w/w) and at a Chol-T liposome: DNA ratio of

10:1. In HeLa cells, both liposome preparations performed best at a ratio of

2.5:1. Differences in transfection efficiencies over the liposome range of 5-20

wg.mL were rather less pronounced with Chol-Q lipoplexes suggesting a

greater versatility of this system.

Abdelwahab et al., (2003) developed novel formulation called Archaeosomes,

the liposomes made from polar ether lipids of archaea show promise for

vaccine and drug delivery applications. The potential toxicity of intravenously

(14, 70, or 140 mg/kg/day for 5 consecutive days) and orally (given at doses of

55, 275, or 550 mg/kg/day for 10 consecutive days) administered as

unilamellar archaeosomes, prepared from total polar lipids (TPLs) extracted

from several species of archaea was assessed in female BALB/c mice. Control

groups of mice were administered as 0.1 ml phosphate-buffered saline (PBS)

by either route. Animals were monitored at least once daily for temperature,

body weight, and clinical signs of adverse reactions. One day after the last

dose, the mice were sacrificed. Blood was collected for selected

biochemical/enzyme analyses, and the major organs (heart, lungs, liver, spleen,

kidneys) were weighed and examined macroscopically where as the spleens

were scrutinized histologically. There was no significant difference observed at

Page 57: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

27

the two lower dosages of intravenously administered vesicles. At the highest IV

dose of 140 mg/kg/day, archaeosomes prepared from TPL of the extreme

halophiles, Halobacterium salinarum and Natronobacterium magadii, specify

potential toxicity, as shown by the symptoms of drop in body temperature, and

loss in body weight. Spleens from mice administered some archaeosomes

types, primarily at the highest intravenous dose tested, were enlarged, had

increased organ weight, and microscopic examination revealed mild to

moderate expansion of the red pulp with increased numbers of hematopoietic

cells, but no changes in the white pulp. There were similar clinical signs at one

or more of the higher oral doses of the ester liposomes and some of the

archaeosome types; however, no other apparent toxicity was observed. It was

concluded that archaeosomes were found well tolerated when administered via

intravenous or oral dosing at dosages indicated for study.

Moein et al., (2001) described that liposomes and polymeric nanospheres are

detected by Kupffer cells and such carriers have applications in site specific

drug delivery by passive as well as active targeting, release profile and as well

as transfusion medicine. The analysis illustrated the translation and fabrication

to design a wide range of particulate carriers (nanospheres, liposomes,

micelles, oil-in-water emulsions) with prolonged circulation and/or target

specificity.

Sihorkar and Vyas., (2001) emphasized that carbohydrate mediated liposomal

formulations interact with the target cells via ligands, such as glycoproteins,

glycolipids, viral proteins, polysaccharides, lipo-polysaccharides and other

Page 58: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

28

oligosaccharides. The polysaccharide anchored liposomal system has been

potential in drug delivery, targeting and immunization over an extended times.

Various schemes were developed with coating of liposomal surface with

natural or hydro-phobized polysaccharides. The polysaccharide (s) coat tends

vesicular constructs physicochemically stable in bio-environments and site-

specific. The polysaccharide anchored liposomes were used for improving the

physical and biochemical stability and ability to target liposomes to specific

organs and cells including lung therapeutics, targeted chemotherapy, cellular

targeting, cellular or mucosal immunity and macrophage activation.

Protein transduction domains (P.T.Ds) have been utilized to enhance cellular

uptake of drugs, proteins, polynucleotides or liposomes (Marty et al., 2004). In

the present study, functionalized antennapedia and HIV Tat peptides were

joined to small unilamellar liposomes via thio-maleimide connection. Uptake

into a panel of cell lines including tumor and dendritic cells with modified

liposomes has been enhanced. At slightest, 100 PTD molecules per SUV

liposome (100 ± 30 nm) were required for efficient translocation into cells.

Modified liposomes were found 15- to 25-fold augmented compared to non

modified liposomes and were repressed by preincubation of liposomes with

heparin. Glycosaminoglycan- deficient CHO cells showed dramatically

reduced cell association of PTD-modified liposomes, confirming the important

role of heparin sulfate proteoglycans in PTD-mediated uptake. ANTP-

liposomes used as carriers of the cytotoxic drug N (4)-octadecyl -1-β-D-

arabino furanosyl cytosine -(5'-5')-3'-C-ethinyl cytidine showed a reduction of

Page 59: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

29

the IC (50) by 70% on B16-F1 melanoma cells compared with unmodified

liposomes. PTD functionalized liposomes, particularly ANTP-liposomes,

represent an interesting novel carrier system for enhanced cell-specific delivery

of a large variety of liposome-entrapped molecules.

Ingebrigtsen and Brandl, (2002) studied that small liposomes obtained by high-

pressure homogenization were fractionated according to their particle sizes by

size exclusion chromatography (SEC). The sub fractions were analyzed by

photon correlation spectroscopy (PCS) as well as enzymatic

phosphatidylcholine (PC) assay for their particle sizes and lipid contents

respectively. For small egg PC-liposomes, a size range of 15 to 60 nm was

found, with 80% of the vesicles being smaller than 30 nm in size. This is in

contradiction to a mean size of 85 ± 32 nm as indicated by PCS without

fractionation. The PCS technique appears to underestimate very small particles

below 30 nm if (few) bigger particles are present. The PCS particle size

analysis of unfractionated hydrogenated egg PC/cholesterol-liposomes (2:1)

mole/mole by PCS did not yield any significant results. On fractionation,

however, a particle size range of 40 nm to 120 nm was determined in a

reproducible manner. The results indicate that a combination of size exclusion

fractionation with subsequent photon correlation spectroscopic particle size

analysis and enzymatic PC assay can give both more detailed and more reliable

insight into the particle size distribution of small liposomes than PCS alone.

Mihaela Trif et al., (2001) prepared liposomes from naturally occurring

biodegradable and nontoxic lipids as these materials are considered good

Page 60: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

30

candidates for local delivery of therapeutic agents. Treatment of arthritis by

intra-articular administration of anti-inflammatory drugs encapsulated in

liposomes prolongs the residence time of drug in joint. In a previous study, it

has been shown that intraarticular injection of human lactoferrin (hLf), a

glycoprotein that possesses anti-inflammatory and antimicrobial activities, into

mice with collagen-induced arthritis reduces inflammation. It is possible of

using liposome-entrapped of hLf as a delivery system to prolong hLf retention

at sites of local inflammation such as the rheumatoid joint. Entrapment of hLf

in negatively charged liposomes enhanced its accumulation in cultured human

synovial fibroblasts from rheumatoid arthritis (RA) patients, compared with

positively charged formulations or free protein. However, in the presence of

synovial fluid, positively charged liposomes with entrapped hLf were more

stable than the negatively charged formulations. In vivo experiments in mice

with collagen-induced arthritis showed that the positive liposomes were more

efficient in prolonging the residence time of hLf in the inflamed joint as

compared with other liposomes. Thus, the amount of hLf retained in the joint

after 2 hr was 60% of the injected dose in case of positive liposomes and only

16% for negative pH-sensitive liposomes. The results suggest that entrapment

of hLf in positively charged liposomes may modify its pharmacodynamics

profile and be of therapeutic benefit in the treatment of RA and other local

inflammatory conditions.

Page 61: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

31

2.4 Application of Liposomes in various Diseases

2.4.1 Cancer

The liposomes used for delivery of anticancer drugs can be broadly classified

into three groups based on the surface charges. Liposomes with no net charge,

having positive charge and liposomes with negative surface charge (Dass.,

2008). Through the utilization of dual liposomes containing 400 µg

dipyridamol/ml and 6mM perifosine, produced by extrusion method, the

metastatic activity of carcinoma cells was found to be suppressed (Wenzel et

al. 2009). Effective delivery to the androgen positive carcinoma patients can be

coupled by forming liposomes containing testosterone as a ligand. Using this

technique, the 5-flourouracil can be targeted to the organs consisting androgen

receptors positive tumors (Mishra et al. 2009).

Zidan et al., (2010) employed NIR spectroscopy for the detection of individual

components of tenofovir in liposomal formulation. By applying the thin film

method, 5 batches were prepared with varying concentrations of stearylamine

as a positive charge imparting agents. It was concluded that drug entrapment

efficiency, zeta potential and particle size of all batches were with in acceptable

range. The increasing concentration of stearylamine resulted in higher

entrapment of drug with retardation of drug release.

Liposomal formulations of Cilostazol inhibit the metastasis in a murine 4-T1

breast cancer model by preventing tumor cell-platelet interactions. Liposomes

were prepared and evaluated in-vitro for their anti-metastatic potency in a

spontaneous metastasis model of 4T1 breast cancer. it was observed that

Page 62: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

32

pulmonary metastasis was reduced to 55% when mice were treated i.v with 100

nmol Cil-L 6 h before tumor cell inoculation and then daily for 2 weeks.

Finally it was observed that Cilostazol can prevent interaction between the

circulating 4-T1 tumor cells and platelets by limiting the agreeability of platelet

cells (Wenzel et al., 2010).

Krieger et al., (2010) investigated ways of overcoming cisplatin resistance of

ovarian cancer cells by targeted liposomes in vitro. PEGylated cisplatin-

containing liposomes were prepared with a mean diameter of 110nm and were

labeled with transferring receptors for easy uptake by cells. The free cisplatin

entry was reduced after 24 hrs in resistance but the liposomal cisplatin was

shown to have equal uptake in both resistant and normal cell lines. Hence, it

was suggested that the liposomal delivery can play an important role in active

targeting of cisplatin. Anti cancer drugs loaded into the flexible liposomal

carrier system can act as a highlighting strategy for developing therapeutic

tools for the management of cancer in children (Federman and Denny., 2010).

Reduced systemic toxicity, better Pka profile and tendency to govern the release

rate of the drug can be gained by delivering the active anticancer drugs through

liposomes. The commercially available liposomal preparation can lead toward

the striking role of this delivery system in chemotherapy (Malam et al., 2009)

In the research conducted for a period of about 15 years, it was concluded that

anticancer drugs encapsulated into the antibody targeted liposomes can be used

as a potentially safe and effective way of drug delivery in chemotherapy

(Torchilin, 2008).

Page 63: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

33

Vinorelbine and parthenolide showed to cause complete inhibitory action on

cancer stem cells in the breast cancer patients. The predisposition of liposomes

for eradication of cancer stem cells along with the cancerous cells was

demonstrated by Liu et al. (2008). The liposomes prepared by natural lipids

showed remarkable reproducibility and these were tested for delivery of

aqueous drugs to the carcinoma cells. The fate of liposomes within the

cancerous cells was depicted by the same group of authors (Gupta et al. 2008).

It is a well established fact that stealth stabilized liposomes offer new era of

interest relating production of delivery vehicles for active targeting of disease,

including Targeted liposomes and genetic material vehicle with cationic

stabilized liposomes (Lasic et al. 1999).

2.4.2 Acquired Immuno Deficiency Syndrome

Liposomes of anti retroviral drugs can be formulated as a drug core surrounded

by the lipid bilayer for the enhanced absorption and effective delivery to the

active site. Example of HIV-1 replicon liposomes inhibits growth by protease

inhibitor enclosed in gp-120 targetted liposomes. The immuno-liposomes were

preferentially taken up by HIV-1 infected cells. It was concluded in the study

that amount and time of stay of drug was moderately superior to free drug

(Clayton et al., 2009).

Liposomes formulated with dimyristoyl phosphatidylcholine and poly oxy

ethylene (n) dodecyl ether for their selective inhibitory effect on HIV-1 type

infected cells. An increase effect was observed in viral production in the

Page 64: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

34

latently HIV infected cells after treatment with hybrid liposomes (Ueoka et al.,

2008)

Mononuclear phagocyte system (MPS) contains lecithin receptors on their

surface as well as on cells are targeted by HIV. In-vitro binding capacity was

observed using mannose binding protein concanavalin A. The entrapment

efficiency was found as 47.2 ± 1.57%. It was taken up in maximum

concentration. Possible differences were observed between free drug,

mannosylated liposomes and uncoated liposomes with an enhanced drug

delivery to the cancerous cells through mannosylated liposomal preparations.

The chances to develop leucocytopenia and anaemia which were commonly

observed in free drug treatment were also decreased using mannosylated

liposomes with improved pharmacological effects (Garg et al., 2006)

Active targeting of HIV drugs to the lymphoid tissues was carried out by

Desormeaux et al., (2005). HIV cells accumulate and replicate into the

lymphoid tissues. Thus, it was concluded through proper experiments that the

active drug targeting can be obtained by optimum ligand coupling with

sterically stabilized liposomes.

2.4.3 Dermatology

Using the amphipathic character of liposomes, both hydrophilic and lipophilic

drugs can be incorporated into them for their potential applications in

dermatology along with the ability of some drugs to cross the skin and reach

the circulation which are otherwise unable to cross the skin. The disorders

related with the hair follicles can be subsided with reduced irritation to the skin.

Page 65: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

35

The level of photodynamic therapy can be enhanced by 5-A.L.A encapsulated

liposomes (de Leeuw et al., 2009).

The ethanolic liposomes were formulated by incorporating Indinavir liposomes

for transdermal delivery. This system resolved the problem of short half life

and erratic absorption due to altered pH when the drug administered via oral

route. It was reported that the absorption trough human cadaver skin was

greater in case of formulated liposomes in comparison with drug solution in

ethanol and conventional liposomes. It was found that liposomes can be

considered as a potential technique for delivery of Indinavir (protease inhibitor)

via skin (Dubey et al., 2010).

Four potential routes of drug delivery are possible for skin; these include hair

follicles, skin pores, columnular spaces and lipid water matrix. The liposomes

can play an important role by retaining the sequester compounds on to the skin

which are otherwise very difficult to be retained in contact with the skin where

they can act as light absorbents, sunscreens or as a reservoir for controlled

release preparations. Flexible liposomes are best absorbed by water gradients

driving force. Stratum cornium and skin functions disabilities can be overcome

by localized liposomes in particular areas onto the body. The lipophilic drugs

can be incorporated into liposomal membranes and can be effectively delivered

to the skin via method that escapes the lysosomal pathway. This method can be

effectively used to deliver active DNA repair enzymes from liposomes into

epidermal cells (Yarosh, 2001).

Page 66: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

36

Lipophilic and hydrophilic drugs can be simultaneously incorporated into the

liposomes and through this system proteinous substances can be delivered to

their active site of delivery. Similarly, lysine and hydro lysine containing

liposomes are acting as an adhesive covering linked enzymatically thus

increasing the contact time of active drugs to the skin (Westerhof, 1985).

A liposomal preparation of betamethasone dipropionate seems better to a

conventional marketable preparation in eczema but not in psoriasis vulgaris.

However, one cannot rule out the follicular pathway as an alternate route.

Many questions must be resolved before a complete understanding of

liposomes as a drug carrier system in dermatology can be reached. However,

examinations performed so far indicate that liposomes might be useful as

vehicles for topical drug delivery in various diseases of the skin (Schmid and

Korting, 1994).

Hexa methyl melamine (HMM) an anticancer agent, was tried to be delivered

through liposomes containing sodium deoxycholate (NaDC), the relative AUC

was found to be higher (1.21 fold) compared with HMM solution and HMM

lip, proving that NaDC LIP can be used as a potential way of delivery for oral

drugs (Sun et al., 2010).

2.4.4 Liver Diseases

By using anionic liposomes, S-adenocyl methionine (SAMe) can be actively

targeted to the liver cells. 1, 4-butanedisulfonate salt (SD-4) containing

liposomal preparation can lead to conclusion that the liposome can serve as an

important way for liver disease (Wagner et al., 2009). Hepatocellular

Page 67: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

37

carcinoma (HCC) may be produced due to NF-kappa B cells activation which

intern is dependent on presence of hepatitis B virus. Liposomes were

formulated containing beta-sitosterol glucoside that can target the liver cells

and hence NF-kappa B inducing kinase (NIK) activity can be suppressed

leading to spatial delivery of siRNA to the liver (Cho et al., 2009).

Similarly, N-trimethyl chitosan (TMC) coated multivesicular liposomes can be

used for oxymatrine (OM) oral delivery in liver problems specially hepatitis B.

The TMC coated multivasicular liposomes were 1.96 fold efficient as per

pharmacokinetic properties compared with non coated liposomes and OM

solution (Cao et al., 2009). Vitamin A coupled liposomes were used for

delivery of small interfering RNA (siRNA) in liver cirrhosis against

glycoprotein-46 in experimental animals through 5 preparations resulting in

possible positive favor for improving the liver cirrhosis (Sato et al., 2008).

The possible enhancements were observed in treating liver cirrhosis in rats by

incorporation of hepatocyte growth factor (HGF) into stabilized liposomes

(SSL) containing cyclic Arg-Gly-Asp (RGD) peptides which are combined

with maleimide. The rate of liver cirrhosis cure was higher in case of RGD-

SSL-HGF injection as compared to SSL –HGF alone, HGF-RGD-SSL and

saline, hence it was concluded that HGF encapsulated liposomes can be used

for better remission of liver cirrhosis by increasing collagen fiber digestion (Li

et al., 2008).

Galactosylated cationic liposomes were used for possible delivery of small

interfering RNA (siRNA) into the liver parenchymal cells. Higher stability was

Page 68: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

38

observed in case of galactosylated liposomes complexed with siRNA then

naked siRNA in the plasma, along with prevention from nuclease digestion. It

was also observed that maximum concentration was within the PC cells, while

naked siRNA was largely subjected to non parenchymal cells (NPC) (Sato et

al., 2007).

Hepatic satellite cells (HSC) are the major cause of liver fibrosis and they can

lead to liver cirrhosis in advance stages, active delivery of drugs were tried to

HSC for possible prevention of liver fibrosis. It was concluded that the

bioactive compounds can be delivered to these cells using liposomes as a

delivery vehicle for prevention of liver fibrosis. (Adrian et al., 2007).

2.4.5 Lungs Diseases

Various model drugs were tried for their Potential delivery to the lungs through

their possible encapsulation into the liposomes because of great versatility of

liposomes nature to carry drugs and capability of carrying various drugs.

Liposomes after being coated with chitosan (CHT) were compared for their

delivery to lungs with empty liposomes of varying compositions. It was

concluded that the CHT coated liposomes due to their mucoadhesive nature

were better for their potential delivery to the lungs by nebulization compared

with non CHT coated liposomes. Similarly, the toxicity to the lungs epithelium

was lower in case of CHT coated liposomes then observed in free drugs

delivered to the lungs by nebulization (Zaru et al., 2009). Similar study was

conducted revealing the possible usage of chitosan aprotonin covered

Page 69: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

39

liposomes for possible delivery of polypeptides by oral route of administration

(Werle, 2009).

Liposomes containing 9-nitrocamptothecin (9-NC) were tried for pulmonary

delivery to determine severity of damage in lungs of mice and distribution of

drug into the available compartment of test animal. Phosphate buffer saline

solution was used to measure the in-vitro release of 9-NC and then distribution

of drug was determined after administration of liposome via pulmonary route.

The lungs damage by the liposomal preparation was less severe than that

caused by free solution or non-liposomal preparation, and drug release with

sustained effects was observed with liposomes delivery (Zhang et al., 2008)

Liposomes were studied for active targeting of drugs to the lungs by

nebulization. Rifampicin (RIF) containing multilamellar vesicle (MLV)

liposomes and dried re-hydrated vesicles (DRV) composed of various lipids

were prepared and characterized (Zaru et al., 2007).

2.4.6 Malaria

Gel core liposomes containing acrylic acid polymers, were prepared by reverse

phase evaporation for the delivery of malarial vaccines by Tiwari et al., (2009)

by injecting into the muscles. The ability of vaccines to stimulate the immune

system was studied against Pfs25 and found to be higher in case of gel core

liposomes containing vaccine than conventionally administered drug.

In a similar study, chloroquine was formulated into the liposomes and its anti-

malarial activity against drug resistant rodent malaria was checked in animal

models (Rats) after making complex between liposomes and Fab Fragment on

Page 70: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

40

monoclonal antibody of mouse. The liposomes coated with Fab fragments were

recognized by malarial red cells both in vivo and in vitro studies (Owais et al.,

1995). Primaquine was also formulated in liposomal preparation for its

effectiveness in animals induced with malaria (Pirson et al., 1980).

Anti-malarial antigens were studied for their appearance on the surface of cells

after ingestion of antibody captured into liposomes by phagocytes. ELISA test

was used for the determination of proteins appeared onto the cell surface. The

relative residence time of liposomes was determined by pulse-chase

experiments. Amount of protein captured by the liposomes was directly linked

with rate and amount of protein exposed onto the surface of cells. Hence, it was

concluded that the antigenic proteins can be expressed safely by using

liposome as a potential vehicle (Verma et al., 1991).

Page 71: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

41

Chapter No. 3

3.1 Introduction

Pain management in current treatment scenario is assumed to be more

important than ever before. Combination of NSAIDs and Opoid analgesics

(Figure 3.1) are being prescribed for the relief of pain in different situations of

cancers, orthopedics, dental pain, post operative pain management,

osteoarthritis, rheumatoid arthritis and related conditions (Srinivasa and

Jennifer, 2005; Raffa, 2001; Singla et al., 2005; Perrrot et al., 2006 ). The

simultaneous measurement of these NSAIDs in biological samples is required

in therapeutic monitoring, pharmacokinetic and bioavailability studies, as well

as in clinical and toxicological screening. Furthermore, it is also very important

to precisely quantify these analgesics in advance drug delivery systems and

marketed brands for quality control operation (Mohammed et al., 2001; Lee et

al., 2000; Kuhlmann et al., 1998; Herrai et al., 1997).

Diclofenac sodium is an aryl propionic acid derivative belongs to non-steroidal

anti-inflammatory drugs (NSAIDs), which have analgesics, antipyretics and

anti-inflammatory activities. Mechanism of action is related to inhibition of

cyclooxygenase (COX) and reduces prostaglandin biosynthesis at the site of

inflammation (Arcelloni et al., 2001). Flurbiprofen is a propionic acid-derived

non-steroidal anti-inflammatory drug (NSAID) used widely in the treatment of

rheumatism and non-arthritic pain and its use is also approved in fever and

platelet aggregation (Panusa et al., 2007). Tramadol hydrochloride (trans-2-

[(dimethyl-amino) methyl] -1-(methoxypheny) cyclohexanol, centrally acting

Page 72: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

42

opoid agonist provide analgesic effect by blocking µ-receptors (Taburet et al.,

1995).

Multiple separation and analysis techniques have been developed for the

simultaneous and single entity determination of different NSAIDs in

formulations and biological fluids. But no method is available for simultaneous

determination of DFS, FLP and TMD in drug products and biological fluids.

The available methods for different combination of analgesics mainly

employed spectroscopic and micro colorimetric assay (Gun and Ismail., 2001;

Soledad et al., 1998; Kugtrin et al., 1997; Mayer et al., 2003),

spectrofluorimetry (Damiani et al., 2008), HPLC with crocheted ETEF post

column photo derivatization (Kuhlmann et al., 1997), HPLC with ultraviolet

(Lee et al., 2000; Herrai et al., 1997; Arsalloni et al., 2001; Panusa et al., 2007;

Taburet et al., 1995; Carson., 2001; Jankowski et al., 2006; Sun et al., 2003)

electrochemical detection (Kuhlmann et al., 1998), HP-TLC method (Lala et

al., 2002), potentiometric analysis (Shamsipur et al., 2005), thermal and raman

spectroscopic method (Sipos et al., 2008; Muzerek and Szustak., 2006.

Spectrophotometric, fluorometric, potentiometric, thin layer chromatographic

method and thermal and raman methods are slow and difficult methods

lacking sensitivity and selectivity required for multidrug analysis in different

fluids, HPLC analysis being fast, sensitive, specific and selective for multidrug

analysis replacing these methods. While mass spectrometric (MS) with

different assemblies are being costly and cannot be employed for routine

analysis of drugs in formulations and biological samples. Elessia et al., (2007)

Page 73: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

43

used UV and ESI-MS detection employing liquid chromatography for eight

different drugs in formulations, although the technique is very sensitive but

peaks of ketoprofen and naproxen interfered with each other. The technique is

not available in most of the laboratories therefore, making the method

expensive and cannot be employed in biological fluid analysis having large

number of samples. Sun et al, (2003) presented HPLC method for determining

seven kinds of NSAIDs simultaneously in pharmaceutical formulations and

human plasma. Although the method was simple but separation was

compromised, as naproxen and fenoprofen were not sufficiently separated from

endogenous peaks and the chromatograph was not clear. Compared to method

developed by Toshio et al, (1997) employing SPE for determination of twelve

drugs that was quite time consuming, expensive, inappropriate separation of

drugs shown in chromatograms and requires large volumes of solvents for

sample preparation resulted in less sensitivity compared to present method

(Herai et al., 1997). In this study, we have employed variable UV detector with

no special assembly which proved to be convenient, inexpensive and suitable

for routine work, an isocratic separation of analgesic drugs only using reversed

phase column was also found better for the separation of these drugs. In spite

of analyzing more drugs, it is better to analyze and separate less number of

drugs to avoid the problem of interference and peak symmetry. The proposed

method was successfully applied for the determination of each drug

simultaneously in drug delivery systems, commercial brands as well as in

human plasma samples.

Page 74: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

44

NH

O

Cl ONa

(a)

OH

O

HClHN

(b)

O

OH

F

(c)

Figure 3.1: Structure of (a) DFS (b) TMD (c) FLP

Page 75: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

45

3.2 EXPERIMENTAL

3.2.1 Materials

Diclofenac sodium (DFS), Flurbiprofen (FLP) and Tramadol hydrochloride

(TMD) were donated by Novartis Pharmaceutical (Pakistan), Pfizer

Pharmaceutical (Pakistan) Ltd. and Getz Pharmaceuticals (Pakistan) Ltd,

respectively. Analytical grade Sodium dihydrogen phosphate, phosphoric acid,

methanol, acetonitrile and trichloroacetic acid were purchased by Merck

(Darmstadt, Germany). Double distilled water was prepared in laboratory

(Pharmacy Department, the Islamia University of Bahawalpur-Pakistan).

3.2.2 B. Chromatographic system and applied conditions

The Sykam GmbH (Germany) HPLC system consisted of S-2100 solvent

delivery system (Germany), VE 3210 variable wavelength UV/VIS detector,

Rheodyne (Sykam GmbH, Germany) sample injector with a 20-µL loop.

Clarity DataApex® software (Sykam GmbH, Germany) was used as interface

modulator for data processing. Chromatographic separations were carried out

on a hypersil ODS column (250 mm × 4.6 mm i. d., 5µm particle size of

internal packing, Germany) preceded by guard column. S 4011 Column

Thermo Controller (Sykam GmbH, Germany) was used to control and maintain

column temperature at 30 ˚C. Isocratic separations were carried out with

optimized composition of mobile phase. After several trials, a mobile phase of

composition acetonitrile-sodium dihydrogen phosphate buffer (pH 3.5; 10m

M)-acetonitrile (33:67, v/v) with an optimized flow rate of 1.0 mL.min-1. The

effluent was monitored at 242 nm for 0–5min, 274 nm 5-11 minutes.

Page 76: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

46

3.2.3 Preparation of standard solutions

100 µg.mL-1 standard stock solutions of DFS, FLB and TMD were prepared by

dissolving 100 mg of each drug in 100 mL of acetonitrile. These stock

solutions were further diluted to make working dilutions to encompass linearity

range of each drug. Standard dilutions were analyzed in HPLC. Standard

dilutions of DFS were made in the range of 10 ng.mL-1 to 20 µg.mL-1, FLB and

TMD were made in standard dilutions of 20 ng.mL-1 to 10 µg. mL-1,

respectively. Same dilutions were prepared in human plasma after spiking

known drug solution concentrations with plasma. Liquid-Liquid Extraction

procedure was adopted for sample preparation HPLC system.

3.2.4 Determination of analgesics in marketed brands

Ten tablets of DFS, FLB and TMD and 10 capsules of DFS and FLP in

sustained release marketed brands were finely ground. An accurately weighed

powdered sample containing the labeled amount of each drug was transferred

to a 100 mL volumetric flask. The volume was adjusted with acetonitrile and

the resultant solution was sonicated for 5 min. A portion of the solution was

then filtered through a 0.45 µm filter. 1mL of filtrate was diluted to 50 mL and

a portion of 20 µL was injected to HPLC system.

Page 77: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

47

3.2.5 Determination of analgesics in drug delivery systems

Liposomes were prepared in laboratory by modified microencapsulation vesicle

method by Tomokio and Fumiyoshi, (2005). Microcapsules of DFS, TMD and

FLP were prepared by coacervation based on temperature change technique

used by Sajeev et al., (2002). The formed liposomes and microcapsules

equivalent to one dose were weighed and dissolving in acetonitrile and vortex

mixed, centrifuged, and filtered through 0.45 µm filter & 1 mL of filtrate was

diluted to 50 mL and injected through 20 µL loop into HPLC system.

3.2.6 Determination of NSAIDs in human plasma

Blood samples were withdrawn and centrifuged to separate plasma and kept in

ultra low freezer (-20°C) until analysis performed. LLE was performed by

taking 250 µL of human plasma in glass tube and 200 µL precipitant was added

and vortex mixed for 1 minute and centrifuged for 5 minutes. The clear

supernatant was separated and injected into HPLC column for separation.

3.3 RESULTS AND DISCUSSIONS

3.3.1 Choice of chromatographic separations

The isocratic elution system was found easier and economical for separation of

analgesics in formulations and plasma samples using an optimized composition

of mobile phase, mixture of acetonitrile-10 mM sodium dihydrogen phosphate

buffer (pH 3.2). The value of retention factor (k′) of each drug was reduced as

the value of pH of mobile phase reaches above 3.5, while diminutive changes

were found up to pH 3.5; the pH of 3.2 was chosen as the best condition. The

proposed separation condition was applied to the analysis of NSAIDs in

Page 78: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

48

pharmaceutical formulations. Though, TMD interfered with modifier retention

and peak of plasma by using methanol which was selected at initial stages but

with adjustment of pH of buffer to pH 3.2 and slight adjustment of composition

in mobile phase (33:67 acetonitrile and phosphate buffer) a complete separation

entities in plasma was made possible (Fig. 3.2). Each drug contains different

chromophores (light absorbing units) for variation in maximum wavelength of

ultraviolet absorbance. Variable wavelength detector was set at different λmax

i.e. 242 nm for 0-5min, 274 nm 5-11 min.

(a)

(b)

Figure 3.2: Chromatograms showing (a) separation of analgesics in drug solution (b) plasma samples

3.3.2 Determination of analgesics in pharmaceutical formulations

The Accuracy is a measure of deviation of mean from the true value as

determined by the replicate assay. The accuracy of the present method was

Page 79: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

49

found in the range of 99.76 to 99.99 for drug solution and 99.65- 99.88 for drug

solutions samples for the drug concentrations of LQC, MQC and HQC (Table

3.1). The measure of the amount of agreement among the observed results

when method is applied repeatedly and reproducibly is called precision. The

precision (% C.V) for all the studied components in different fluids were found

in the recommended limits and summarized in Table 3.2. The results of

analysis of liposomes, microcapsules and marketed brands are shown in Table

3.3.

Table 3.1: With-in-Day Accuracy and Precision of Analgesics in Drug Solutions

Curve Code LQC (ng.ml

-1) MQC (µg.ml

-1) HQC (µg.ml

-1)

DFS FLP TMD DFS FLP TMD DFS FLP TMD

Batch 01 9.86 19.94 19.28 0.995 0.998 0.992 9.98 19.98 19.92

Batch 02 9.91 19.98 19.34 0.992 0.986 0.994 9.94 19.96 19.84

Batch 03 9.88 19.92 19.81 0.998 0.979 0.996 9.881 19.96 19.9

Batch 04 9.84 19.96 18.96 0.969 0.996 0.995 9.96 19.94 19.96

Batch 05 9.87 19.98 18.81 0.996 0.998 0.988 9.61 19.97 19.81

Batch 06 9.85 19.95 19.72 0.994 0.994 0.996 9.92 19.98 19.88

Mean 9.87 19.96 19.32 0.99 0.99 0.99 9.88 19.97 19.89

S.D. 0.02 0.02 0.40 0.01 0.01 0.00 0.14 0.02 0.05

Nominal 10 20 20 1 1 1 10 20 20

%CV 0.25 0.12 2.06 1.09 0.78 0.31 1.39 0.08 0.27

%Bias -1.32 -0.22 -3.40 -0.93 -0.82 -0.65 -1.18 -0.18 -0.57

%Accuracy 98.68 99.78 96.60 99.07 99.18 99.35 98.82 99.83 99.43

Page 80: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

50

Table 3.2: With-in-Day Accuracy and Precision of Analgesics in Standard Drug

Solutions

Table 3.3: Percentage Contents Found in Liposomes, Microcapsules and Marketed

Brands

Formulations Drug/Brands Stated Found Percentag

e

Liposome DFS 50 49.86 99.72

Microcapsule

DFS 50 49.93 99.86 FLP 100 99.95 99.95 TMD 100 99.72 99.72

Marketed

brands

Dicloran® Sami 50 49.88 99.76

Voltral ® Novartis 50 49.92 99.84

Froben® Abbott 100 99.94 99.94

Ansaid® Pfizer 100 99.78 99.78

Tramal® Searle 50 49.9 99.8 Traumanil® Howards 50 49.91 99.82

Curve Code DFS FLP TMD

LQC MQC HQC LQC MQC HQC LQC MQC HQC

Units ng. ml-1

µg. ml-1

µg. ml-1

ng. ml-1

µg. ml-1

µg. ml-1

ng. ml-1

µg. ml-1

µg. ml-1

Day-01

9.98 19.99 19.96 0.996 2.49 2.48 9.96 19.98 19.98 9.99 19.95 19.94 0.995 2.49 2.49 9.98 19.96 19.96 9.99 19.98 19.97 0.998 2.48 2.49 9.92 19.97 19.97

Day-02

9.98 19.97 19.96 0.992 2.48 2.48 9.96 19.98 19.94 9.95 19.96 19.98 0.991 2.49 2.47 9.94 19.92 19.95 9.95 19.98 19.94 0.996 2.48 2.48 9.98 19.96 19.94

Day-03

9.97 19.96 19.94 0.990 2.49 2.49 9.92 19.98 19.96 9.96 19.94 19.94 0.988 2.48 2.48 9.96 19.94 19.97 9.95 19.94 19.92 0.992 2.49 2.48 9.95 19.96 19.98

Mean 9.97 19.96 19.95 0.99 2.49 2.48 9.95 19.96 19.96 S.D. 0.017 0.018 0.02 0.003 0.005 0.07 0.02 0.02 0.02

Nominal 10 20 20 1 2.5 2.5 10 20 20 %CV 0.17 0.09 0.09 0.33 0.21 0.27 0.22 0.10 0.08 %Bias -0.31 -0.18 -0.25 -0.69 -0.58 -0.71 -0.48 -0.2 -0.19

%Accuracy 99.69 99.82 99.75 99.31 99.42 99.29 99.52 99.81 99.81

Page 81: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

51

3.3.3 Determination of analgesics in human plasma

3.3.3.1 Linearity

Three replicates of each concentration in the dynamic range of 0.01–10 µg-1

for DFS and 0.01-20 µg.mL-1 for TMD and FLP were run in plasma matrix. A

linear relationship (Figure 3.3) was obtained between the peak area and

concentration for each drug (Table 3.4). Limit of quantifications (LOQs) of

each compound was first concentration of each drug which was set at three fold

of limits of detections (LODs) at a signal-to-noise ratio of 3. The method

showed reasonable linearity to measure the drugs in biological fluids. Further,

it was also found that linearity of the present method was comparable to the

methods of Mohammed et al., (2001); Lee et al., (2000); where as, the present

method was found better compared to other studies available in literature

(Kuhlmann et al., 1998; Arcelloni et al., 2001 and Panusa et al., 2007).

(a)

y = 0.1401x + 94.961R2 = 0.9963

0

500

1000

1500

0 2000 4000 6000 8000 10000

Plasma Conc. (ng/ml)

Resp

en

se

Page 82: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

52

(b)

Figure 3.3: Standard curve of (a) DFS (b) FLB and (c) TMD in plasma

y = 0.1259x + 119.83R2 = 0.997

0

1000

2000

3000

0 5000 10000 15000 20000

Plasma Conc. (ng/ml)

Resp

on

se

y = 0.1207x + 81.157R2 = 0.9986

0

1000

2000

3000

0 5000 10000 15000 20000

Plasma Conc. (ng/ml)

Resp

en

se

Page 83: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

53

Table 3.4: Standard Curve Parameters of Analgesics in Plasma

Curve Code Slope

Mean (%C.V)

Intercept

Mean (%C.V)

r-square

Mean (%C.V)

DFS 94.96 (0.14) 0.14 (0.84) 0.996 (0.05)

FLP 119.83 (0.62) 0.125 (1.36) 0.998 (0.08)

TMD 70.8 (1.26) 78.6 (0.48) 0.995 (0.44)

3.3.3.2. Extraction efficiency

Efficiency of the extraction method was determined for each drug at lowest

concentration and highest concentration in plasma by comparing the peak

heights of each compound spiked in plasma, to those of standards in the mobile

phase. Several trials were done to find out adequate extraction solvent for

withdrawal of drugs from plasma matrix to form easier method of liquid-liquid

extraction and TCA with acetonitrile was found satisfactory. The recoveries for

analgesics were 99.03–99.80 at lowest, middle and highest concentrations of

standard curve (Table 3.5). These findings were higher than studies available in

literature (Arceloni et al., 2001; Panusa et al., 2007; Gun and Ismail., 2001;

Soledad et al., 1998; Jankowski et al., 2006). The higher values of extraction

procedure indicate efficiency of present method which was justified by higher

sensitivity and accuracy.

Page 84: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

54

Table 3.5: Extraction Efficiency of Analgesics in Plasma

Code

LQC (Mean, %C. V.) HQC (Mean, %C. V.)

Ext. Non-Ext. % Ext. Non-Ext. %

DFS 9.70 (1.73) 9.75 (1.72) 99.52 9.91 (0.15) 9.95 (0.31) 99.60

FLP 9.77 (0.43) 9.80 (0.42) 99.69 19.91 (0.08) 19.95 0.15) 99.80

TMD 9.84 (0.82) 9.93 (0.81) 99.03 19.85 (0.10) 19.92 0.18) 99.63

3.3.3.3. Assay accuracy and precision in plasma

Intra-day accuracy and precision of the assay was performed six batches spikes

plasma samples with DFS at low (0.01 µg.mL-1), middle (1.0 µg.mL-1) and high

(10 µg.mL-1) concentrations, FLP and TMD were spiked at low (0.02 µg.mL-1),

middle (1.0 µg.mL-1) and high (20 µg.mL-1) concentrations. Triplicate samples

at each concentration were analyzed for the intra-day assessment and finding

system error (biasness). For the inter-day precision (measure of repeatability

and reproducibility) analysis of the spiked plasma at the same concentrations of

each drug was performed on 3 different days. The results are given in Table 3.6

and Table 3.7. The values were found in line with studies available in literature

(Sun et al., 2003 and Mayer et al., 2003).

Page 85: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

55

Table 3.6: Between-the-Day Accuracy and Precision in Plasma

Table 3.7: Between-the-Day Accuracy and Precision in Plasma

Curve

Code

DFS FLP TMD

LQC MQC HQC LQC MQC HQC LQC MQC HQC

Units ng.mL-1 (µg.mL-1) ng.mL-1 (µg.mL-1) ng.mL-1 (µg.mL-1)

Batch-01 9.96 19.98 19.94 0.996 2.48 2.47 9.86 19.96 19.95

9.94 19.94 19.92 0.995 2.49 2.48 9.95 19.94 19.93 9.98 19.96 19.95 0.998 2.47 2.49 9.94 19.92 19.94

Batch-02 9.93 19.94 19.90 0.992 2.46 2.48 9.92 19.88 19.92

9.90 19.92 19.88 0.991 2.47 2.46 9.92 19.94 19.90 9.92 19.91 19.84 0.996 2.46 2.48 9.94 19.94 19.92

Batch-03 9.95 19.92 19.89 0.990 2.49 2.47 9.22 19.92 19.90 9.94 19.92 19.82 0.988 2.48 2.48 9.86 19.92 19.94 9.92 19.90 19.80 0.992 2.49 2.47 9.89 19.94 19.88

Mean 9.94 19.93 19.88 0.99 2.487 2.476 9.83 19.93 19.92

S.D. 0.02 0.025 0.053 0.003 0.012 0.008 0.23 0.023 0.023

Nominal 10 20 20 1 2.5 2.5 10 20 20

%CV 0.24 0.13 0.26 0.33 0.49 0.36 2.36 0.11 0.12 %Bias -0.62 -0.34 -0.59 -0.69 -0.93 -0.98 -1.67 -0.4 -0.40 %Acc 99.38 99.66 99.41 99.31 99.07 99.02 98.33 99.64 99.60

Curve

code LQC (ng.mL

-1) MQC (µg.mL

-1) HQC (µg.mL

-1)

DFS FLP TMD DFS FLP TMD DFS FLP TMD

Batch 01

9.82 19.28 19.28 0.995 0.998 0.992 9.98 19.88 19.92

9.88 19.34 19.34 0.992 0.986 0.994 9.94 19.96 19.84

9.86 19.81 19.81 0.998 0.979 0.996 9.881 19.96 19.9

9.85 18.96 18.96 0.969 0.996 0.995 9.96 19.84 19.96

9.90 18.61 18.81 0.996 0.998 0.988 9.61 19.87 19.81

9.82 19.72 19.72 0.994 0.994 0.996 9.92 19.92 19.88

Mean 9.86 19.29 19.32 0.99 0.99 0.99 9.88 19.91 19.89

S.D. 0.03 0.45 0.40 0.01 0.01 0.00 0.14 0.05 0.05

Nominal 10 20 20 1 1 1 10 20 20

%CV 0.31 2.35 2.06 1.09 0.78 0.31 1.39 0.25 0.27

%Bias -1.38 -3.57 -3.40 -0.93 -0.82 -0.65 -1.18 -0.48 -0.58

%Acc 98.62 96.43 96.60 99.07 99.18 99.35 98.82 99.53 99.43

Page 86: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

56

3.3.3.4. Stability

The stability of analgesic drug was studied in acetonitrile on four weeks to

observe change in composition of solution. These studies were conducted at

lowest and highest concentrations (extremes) of each drug. The value of %CV

for DFS was 0.35 for LQC and 0.09 for HQC; similarly it was 0.13 and 0.15

for LQC and 0.22 & 0.25 HQC for FLP and TMD, respectively (Table 3.8).

The freeze-thaw stability of analgesics was also approximated over three

freeze-thaw cycles in plasma. Lowest and highest concentrations of each drug

were spiked in plasma, frozen at -25°C and thawed at room temperature for

three successive events. Table 3.9 furnishes freeze-thaw stabilities of DFS and

found % CV (LQC, -1.80 to -2.01; HQC, -0.34 to -0.54). The values for FLP

and TMD were summarized in Table 3.10 and Table 3.11, respectively. These

findings specify that analgesics were stable in processing for finding

concentration after freezing for holding the samples in ultra low freezer. These

values were also found in good agreement with previous studies and indicate

that analgesics in drug solutions are stable for one month and spiked plasma

after three freeze-thaw cycles.

Table 3.8: Long Term Stability of Analgesics in Standard Drug Solutions

Dilutions Week1 Week2 Week3 Week4 Mean S.D. %CV

DFS LQC 9.94 9.91 9.88 9.86 9.89 0.03 0.35

DFS HQC 19.97 19.95 19.94 19.93 19.95 0.02 0.09 FLP LQC 19.96 19.94 19.92 19.9 19.93 0.03 0.13

FLP HQC 19.98 19.94 19.90 19.88 19.93 0.04 0.22 TMD LQC 19.95 19.92 19.9 19.88 19.91 0.03 0.15 TMD HQC 19.98 19.92 19.94 19.86 19.93 0.05 0.25

Page 87: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

57

Table 3.9: Freeze-Thaw Stability of DFS in Plasma

Curve

Code

Cycle Cycle 0 Cycle 1 Cycle 2 Cycle 3

LQC HQC LQC HQC LQC HQC LQC HQC

DFS 01

9.68 9.56 9.52 9.81 9.54 9.75 9.46 9.97

9.81 9.98 9.91 9.71 9.42 9.68 9.81 9.64

9.89 9.92 9.42 9.84 9.83 9.79 9.59 9.69

Mean 9.79 9.82 9.62 9.79 9.60 9.74 9.62 9.77

S.D. 0.11 0.23 0.26 0.07 0.21 0.06 0.18 0.18

Nominal 10 20 10 20 10 20 10 20

%CV 1.08 2.31 2.69 0.69 2.20 0.57 1.84 1.82

%Diff - - -1.80 -0.34 -2.01 -0.81 -1.77 -0.54

Table 3.10: Freeze-Thaw Stability of FLP in Plasma

Curve

Code

Cycle Cycle 0 Cycle 1 Cycle 2 Cycle 3

LQC HQC LQC HQC LQC HQC LQC HQC

FLP 01

19.96 19.86 19.87 19.78 19.45 19.68 18.85 19.75

19.12 19.79 18.78 19.85 19.52 19.81 19.75 19.62

19.97 19.84 19.1 19.71 19.7 19.10 18.62 19.45

Mean 19.68 19.83 19.25 19.78 19.56 19.53 19.07 19.61

S.D. 0.488 0.036 0.560 0.070 0.129 0.378 0.597 0.150

Nominal 20 20 20 20 20 20 20 20

%CV 2.48 0.18 2.91 0.35 0.66 1.94 3.13 0.77

%Diff - - -2.20 -0.25 -0.64 -1.51 -3.10 -1.13

Page 88: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

58

Table 3.11: Freeze-Thaw Stability of TMD in Plasma

Curve

Code

Cycle Cycle 0 Cycle 1 Cycle 2 Cycle 3

LQC HQC LQC HQC LQC HQC LQC HQC

TMD 01

19.86 19.86 19.78 19.77 19.55 19.72 19.42 19.7

19.92 19.95 18.88 19.87 19.68 19.83 19.60 19.72

19.92 19.94 19.85 19.85 19.77 19.75 19.62 19.65

Mean 19.90 19.92 19.50 19.83 19.67 19.77 19.55 19.69

S.D. 0.03 0.05 0.54 0.05 0.11 0.06 0.11 0.04

Nominal 20 20 20 20 2 20 20 20

%CV 0.17 0.25 2.77 0.27 0.56 0.29 0.56 0.18

%Diff. - - -1.99 -0.44 -1.17 -0.75 -1.78 -1.14

LQC=Lowest quality control concentration in ng.mL-1 HQC= Highest quality control concentration in µg.mL-1

4. Conclusion

A simple and inexpensive simultaneous high performance liquid

chromatographic method for the determination of analgesics employing UV

detection was developed and found successful in analysis of marketed brands,

drug delivery systems and human plasma samples. The validation parameters

were also found in acceptable limits of FDA and ICH. LOQs of each drug in

plasma were as low as 10 ng.mL-1 for DFS and 20 ng.mL-1 for FLP and TMD

which was enough for monitoring the blood levels of each entity. The method

was found accurate, precise and stable that makes it applicable for quality

control operations in pharmaceutical industry, pharmacokinetics studies and

therapeutic drug monitoring of these drugs in clinical settings.

Page 89: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

59

Chapter No. 4

4.1 Introduction

The NSAIDs remain first-line drug therapy for a wide range of rheumatic and

other pain conditions. The combination of drugs for the complete eradication of

disease is requirement of era (Tanveer and Marc, 1997). Diclofenac potassium

(DP) is potassium-[(2, 6-dichlorophenyl) amino]-phenyl acetate (Fig. 1a)

(Henk et al., 1997). It possesses analgesic, anti-inflammatory, and antipyretic

activity. It inhibits prostaglandin synthesis by interfering with the action of

prostaglandin synthetase (cyclooxygenase) (Gupta et al., 2002). Another novel

drug Meloxicam (4-hydroxy- 2-methyl- N- (5-methyl-2-thiazolyl) -2 H-1, 2-

benzothiazine- 3-carboxamide-1, 1-dioxide) of acidic enolcarbox-amide class

(Fig. 1 b) yield peak plasma concentrations in 5-11 hours and peak time is

much longer under pain condition; therefore, a combination is prescribed to fill

this gap and numerous studies have been taken account for using combination

of analgesics (Henk et al., 1997; Gupta et al., 2002; Valat et al., 2001). The late

onset of meloxicam can be advantageously gapped with use of diclofenac

potassium whose blood level appears with in 30 minutes after administration.

Literature search revealed that no HPLC method has yet been available for the

analysis of DP and MX in plasma, advance drug delivery systems as well as in

quality control samples. Limited simultaneous methods exist in literature for

DP and MX separately or/and in different combinations. A study available for

simultaneous determination of diclofenac potassium and tizanidine in tablet

dosage form using spectrophotometery (Sanjay et al., 2006). Simultaneous

Page 90: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

60

narrow bore HPLC using column-switching analysis for aceclofenac and

diclofenac in human plasma (Lee et al., 2000), reverse phase HPLC method for

simultaneous determination of meloxicam and pridinol mesylate in drug

formulations (Silvana et al., 2008), LC-MS method for the simultaneous

determination of piroxicam, meloxicam and tenoxiam in human plasma.

Different assays are available for determination of pharmacokinetics of

individual drugs, either DP or MX, using HPLC technique (Young et al., 2005;

Marzo et al., 2000; Mahmood et al., 2009; Nopamas et al., 2005; Velpandian et

al., 2000; Xu et al., 2001; Dasandi et al., 2002; Busch et al., 1995; Turck et al.,

1995; Shubha et al., 2004; Zwilla et al., 2003; Emirhan and Sedef., 2003). A

method employing CZE (capillary zone electrophoresis) is also available for

the determination of meloxicam in tablets, although benefits using less solvent

and simplicity but the method are not available for simultaneous determination

of meloxicam with any other drug or combinations or in plasma matrix (Limian

and Hian., 2001). A method demonstrated by Silvana et al., (2008) determining

the contents of meloxicam and pridinol mesylate in commercial pharmaceutical

formulations, although the method is simple but is not applicable in plasma

analysis. LC–MS/MS technique for the determination of piroxicam, meloxicam

and tenoxicam in plasma was developed by Young et al., (2005) seems

worthless as the combination of three drugs is not prescribed for a patient

suffering any kind of pain condition. A study reported by Malliou et al., (2005)

describing simultaneous determination of Clobutinol with some anti-

inflammatory drugs in urine samples while the present method quantitated both

Page 91: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

61

analgesics in plasma matrix. Further, difference exists in condition of

experiments and mobile phase compositions as well as results of validation

parameters. Microextraction methods are becoming important in analysis

owing to reduced extraction time, increased efficiency, minimum solvent

expenditure, minimum exposure of toxic solvents and cost effective (Rana et

al., 2008; Wen-Hsein et al., 2009; Blomberg, 2009). No Microextraction

method is available for the determination of diclofenac potassium and

meloxicam. One study is available using microextraction method for analysis

of diclofenac only in dosage forms (Ali et al., 2008). Therefore, the HPLC

method was developed and validated in the present study can advantageously

applied for the determination of DP and MLX in pharmaceuticals, advanced

drug delivery systems, and therapeutic drug monitoring studies for better

patient care.

NH

Cl

Cl

O

O K

NS

O O

OH

CH3

O

NH

S

N

CH3

(a) (b)

Figure 4.1: Structural formula for DP (a) and MLX (b)

Page 92: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

62

4.2 Experimental

4.2.1 Materials and solvents

Diclofenac potassium was received as research material donation from

Novartis Pharmaceutical (Pakistan) Ltd. Meloxicam was donated by Getz

Pharma. (Pakistan) Ltd. Soya Lecithin, chloroform, acetonitrile, methanol and

disodium hydrogen phosphate buffer, ortho phosphoric acid was purchased

from Merck (Germany). HPLC-grade solvents were employed for analyses.

4.2.2 Chromatographic system and optimized conditions of separation

The chromatographic system consisted of series 1200 Agilent Technologies.

Series 1200 pumps, Series 1200 variable wavelength UV and VIS

(Programmable) diode array detector (DAD) which was automatically set at

wavelengths of 280 nm and 340 nm. The drugs were separated ODS Hypersil

(Thermo Electron Corporation) with dimensions of 250 mm Length, 4.6 mm

ID, and 5 µm particle size of internal packing. The mobile phase composition

was 20:20:60 (v/v/v) for acetonitrile, methanol and 20 x 10-3 M sodium

dihydrogen phosphate buffer, respectively. The pH was maintained at 3.7 with

a flow rate of 1.0 ml min−1. An aliquot of 20 µL were injected into HPLC

system and spectra was recorded.

4.2.3 Preparation of stock and working standard solutions

The stock solutions of DP in acetonitrile and MLX in acidic methanol were

prepared in a concentration of 100 µg.mL-1 by dissolving an accurately

weighed (Shimadzu AUX 220) amount (10 mg). The solutions were preserved

at 4°C, in light-resistant containers and were left to attain room temperature

Page 93: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

63

before use. Working dilutions were prepared in concentration ranges of

10ng.mL−1 to 10µg.mL−1 for DP and 15ng.mL−1 to 10µg.mL−1 for MLX in

anticipated range of plasma concentration. All the solutions were stored in

refrigerator throughout the experiments, when not used.

4.2.4 Preparation of liposomes

Modified microencapsulation vesicle (MCV) method of Tomoko Nii and

Fumiyoshi Ishi., (2005) was adopted for preparation of diclofenac potassium

liposomes. Soya lecithin (10 g) was dissolved in 25 mL chloroform and 2.5

gm DP dissolved in 25 mL water separately; both were mixed to form water in

oil emulsion. The formed emulsion was homogenized at 10,000 rpm for half an

hour. This primary emulsion was instantly poured to 100 mL of water in a

round bottom flask at 45°C at agitation speed of 700 rpm to form a water-in-

oil-in-water multiple emulsion. The chloroform was recovered by rotary

vacuum evaporator and DP loaded liposome suspension was freeze dried and

dry powder was filled in capsules of weighing 50 mg for DP oral delivery.

4.2.5 Sample preparation of liposomes, marketed brands and human

plasma

Diclofenac in liposomes equivalent to one dose were weighed and dissolved in

methanol and volume was adjusted to 100 mL. Similarly 20 tablets and

capsules of DP and MLX were ground, LLME method was adopted for

preparation of samples. 150 µL of the sample was transferred to poly propylene

tube and equivalent amount of extraction solvent was added, vortexed and

centrifuged at 3000 rpm for 2 minutes. The constituents were dried under

Page 94: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

64

gentle stream of nitrogen and reconstituted with 60 µL of mobile phase and 20

µL injected into HPLC system. Aliquot of the human plasma (200 µL) spiked

with diclofenac sodium was taken in poly propylene tube and 200 µL of

acetonitrile was added, vortexed for 30 seconds and centrifuged at 3000 rpm

for 2 minutes. The supernatant layer was separated and filtered through

cartridge filter (0.45 µm pore size and 13 mm diameter) and collected in poly

propylene, dried under gentle stream of nitrogen and reconstituted with 60 µL

of mobile phase and 20 µL injected into HPLC system.

4.2.6 Method validation

The linearity, limit of detection, limit of quantification and precision and

accuracy were established. The intra and inter-day precision and accuracy of

the assay were determined by percent coefficient of variation (C.V) and percent

relative error (R.E) values, respectively. Samples of lowest, middle and highest

concentrations were spiked for the determination of precision and accuracy.

Triplicates at each concentration were routed at day 1, 3, 5 and 8 for inter-day

determination of precision and accuracy. Signal-to-noise ratio (s/n) of 3:1 was

selected for the limit of detection (LOD) while three fold of LOD was finalized

for determination of LOQ by comparing test results from spiked plasma

samples with known concentrations of drugs with blank samples.

4.2.7 Recovery

The efficiency of Liquid-Liquid Microextraction (LLME) method was

validated and confirmed by determination of extraction recovery. The peak

response of spiked plasma was determined and compared of with standard QC

Page 95: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

65

samples at Lowest, Mid and Highest concentration. Triplicates of each

concentration level was determined and compared.

4.2.8 Stability studies

Three stability studies were assessed in this study.

(1) Three cycles freeze and thaw stability was determined by thawing at room

temperature for 2 h and then refreezing at −50°C for 24 h.

(2) Bench top stability of spiked plasma stored at room temperature was

evaluated for 12 h.

(3) The long-term stability was assessed by carrying out the experiment for

four weeks at storage temperature -20°C.

Page 96: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

66

4.3 Results and discussion

4.3.1 Screening and optimization

4.3.1.1 Selection of detection wavelength

The HPLC-Diode array Chromatogram (Figure 4.2) for solution containing

both drugs showed maximum wavelength of 280 and 340 for DP and MLX,

respectively. HPLC attached DAD was used for this purpose. This suggested

optimum detection wavelengths were determined in order to favor the

quantification of both drugs in samples.

4.3.1.2 Choice of mobile phase composition

After a series of viewing trials, the phosphate buffers provided improved and

better peak resolution and separation than their acetate and citrate counterparts.

It was also found rational to use methanol-acetonitrile in molar ratio of (20:20)

and phosphate buffer with 60% of aqueous phase produced satisfactory peak

symmetry and selectivity. The phosphate buffer of strength of 20 x 10-3 was

selected and found better to avoid peak tailing and broadening. The effect of

increased pH and buffer concentration reduce the retention time while a smooth

increase of the retention time (Tr) was evidenced with increments in the ionic

strength of aqueous phase. The pH of 3.7 was selected because it produces the

capacity factor (k`), Separation factor (α) and Resolution (R) values of 2.115 ±

0.18, 1.16 ± 0.22 and 1.68 ± 0.32 (mean ± S.D), respectively. In reverse-phase

chromatography, separation is governed by the interactions in mobile phase

because the stationary phase surface is saturated by the molecules of organic

modifier of the mobile phase. A monolayer of methanol is adsorbed on to the

Page 97: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

surface of stationary phase if the concentration is greater than 10% of

methanol. The addition of acetonitrile further facilitated the saturation of

stationary phase layer. The changes in controlled

concentration and flow rate affect elution time, peak separation and resolution.

Therefore, these conditions were optimized to adjust best retention time (tr) of

DP and MLX suitable for analysis of drugs (Fig

Figure 4.2: UV Scan of DP and MLX drug so of HPLC

Figure 4.3: Separation of DP and MLX plasma samples on C

surface of stationary phase if the concentration is greater than 10% of

methanol. The addition of acetonitrile further facilitated the saturation of

stationary phase layer. The changes in controlled variables like pH, buffer

concentration and flow rate affect elution time, peak separation and resolution.

Therefore, these conditions were optimized to adjust best retention time (tr) of

DP and MLX suitable for analysis of drugs (Figure 4.3).

UV Scan of DP and MLX drug solutions in Diode Array Detector of HPLC

Figure 4.3: Separation of DP and MLX plasma samples on C

67

surface of stationary phase if the concentration is greater than 10% of

methanol. The addition of acetonitrile further facilitated the saturation of

variables like pH, buffer

concentration and flow rate affect elution time, peak separation and resolution.

Therefore, these conditions were optimized to adjust best retention time (tr) of

lutions in Diode Array Detector

Figure 4.3: Separation of DP and MLX plasma samples on C 18 Column

Page 98: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

68

4.3.2 Method validation

4.4.2.1 Linearity

Linearity is evaluated by assessing coefficient of correlation (r), slope and

intercept. The present method was linear as the values were with in acceptable

ranges of concerned guidelines (ICH, 2005). The method was linear in

concentrations range of 0.01-10 µg. mL-1 for DP and 0.015-10µg.mL-1 for

MLX spiked in plasma. The chromatograms were gathered and peak area was

recorded. The graph between concentration and peak area were constructed and

presented in Fig. 4.4. The values of standard curve parameters were computed

by Kinetica® software and shown in Table 4.1. Six (n=6) replicates were tested

at each level. The mean value of (r2 was 0.9989 and 0.9979 for DP and MLX,

respectively) showed that method produced good linearity over entire range of

concentrations and found better when compared to the previous studies

available in literature (Lee et al., 2000; Silavana et al., 2008; Young et al.,

2005)

(a)

y = 667.98x + 56.586

R2 = 0.9989

0

900

1800

2700

3600

4500

0 1 2 3 4 5

Plasma Conc. (µg/ml)

Pea

k A

rea

Page 99: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

69

(b) Figure 4.4: Standard Curve of DP (a) and MLX (b) in spiked human plasma

y = 79.688x + 113.67

R2 = 0.9979

0

500

1000

1500

2000

2500

0 5 10 15 20 25 30

Plasma Conc. (µg/ml)

Pea

k A

rea

Page 100: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

70

Table 4.1: Standard curve parameters of DP (a) and MLX (b) in plasma samples

4.3.2.2 Limit of detection and quantification

Limit of detection (LOD) is the concentration of analyte that can be reliably

detected under the stated experimental conditions and give signal-to-noise ratio

of 2 or 3. The signal is the analyte response (peak) and noise is amplitude of

short term noise appeared in the baseline. The limit of detection (LOD) was

established from the relative standard deviation (R.S.D.) of the response and

the slope of calibration curves prepared with known concentrations of DP and

MLX. The values were 3 and 5 ng.mL−1 for DP and MLX, respectively. The

limit of quantification (LOQ) is the amount of analyte that can be quantified

(a)

Curve Code Slope Intercept r2 Conc. Range

DP-01 667.94 56.670 0.9993

0.01-10 (µg.mL-1)

DP-02 668.48 56.542 0.9990

DP-03 667.53 56.546 0.9985

Mean 667.98 6.586 0.9989

S.D. 0.5 0.0728 0.0004

%CV 0.072 0.13 0.040 %error 0.61 0.58 0.52

(b)

Curve Code Slope Intercept r2 Conc. Range

MLX-01 79.539 113.781 0.9982

0.015-10 (µg.mL-1)

MLX-02 80.143 113.658 0.9981

MLX-03 79.382 113.571 0.9975 Mean 79.688 113.670 0.9979 S.D. 0.4 0.11 0.0004

%CV 0.504 0.093 0.038 %error 0.72 0.75 0.67

Page 101: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

71

under the same experimental conditions. This was calculated at 3 multiple of

LOD for both drugs. The values were first values of standard curve as 10 and

15 ng.mL−1 for DP and MLX, respectively (Table 4.1). The LOD and LOQ

values were better than previous studies (Sanjay et al., 2006; Lee at., 2000;

Silavana et al., 2008; Velpandian et al., 2000; Emirhan and Sadaf., 2003;

Limian and Hian., 2001) and comparable with studies employing micro-

extraction methods (Rana et al., 2008; Wen-Hsein et al., 2009; Lars., 2009; Ali

et al., 2008).

4.3.2.3 Precision and Accuracy

Precision is the degree of closeness of results and is reported as % C.V.

Repeatability (intra-day) and reproducibility (inter-day) are divisions of

precision. Accuracy is the measure of systematic error or biasness. An assay is

accurate if the mean value of the result is same as the true value. Nine replicate

injections of the spiked standard solutions in plasma at lowest, mid and highest

standard curve concentration levels of DP and MLX. The concentrations

studied were 10 ng.mL−1, 1µg.mL−1 and 10 µg.mL−1 for DP and 15 ng.mL−1,

1µg.mL−1 and 10 µg.mL−1 for MLX. The values of intra-day precision and

accuracy (Table 4.2) were 99.22 - 99.37% and 99.17 - 99.50% while values of

%bias were -0.63 to -0.78 and -0.50 to -0.83 for DP and MLX, respectively.

The values of Inter-day precision and accuracy were calculated at same

concentrations of DP and MLX as Intra-day studies (Table 4.3). 99.26-99.44%

and 98.67-99.67% precision and accuracy were recorded while values of %bias

were -0.62 to -1.55 and -0.33 to -1.33 for DP and MLX, respectively. The

Page 102: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

72

values were found better than available in literature (Sanjay et al., 2006; Young

et al., 2005; Velpandian et al., 2000).

Table 4.2: Inter-day Precision and Accuracy of DP and MLX in human Plasma DP MLX

Curve Code LQC MQC HQC LQC MQC HQC

Units ng.mL-1 µg.mL-1 µg.mL-1 ng.mL-1 µg.mL-1 µg.mL-1

Nominal 10.00 1.00 10.00 15.00 1.00 10.00

Batch-01

9.92 0.99 9.98 14.990 0.98 9.99

9.9 0.99 9.96 14.950 0.99 9.97

9.94 0.98 9.99 14.970 0.98 9.96

Batch-02

9.96 0.99 9.90 14.940 0.99 9.94

9.94 0.98 9.86 14.960 0.99 9.92

9.96 0.97 9.88 14.940 0.99 9.96

Batch-03

9.92 0.99 9.94 14.900 0.98 9.9

9.96 0.98 9.92 14.980 0.99 9.88

9.94 0.99 9.90 14.920 0.99 9.93

Mean 9.938 0.984 9.926 14.950 0.987 9.939

S.D. 0.0211 0.007 0.045 0.029 0.005 0.035

%CV 0.2121 0.738 0.454 0.192 0.507 0.354

%Bias -0.622 -1.556 -0.744 -0.33 -1.333 -0.611

%Accuracy 99.38 98.44 99.26 99.67 98.67 99.39

Page 103: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

73

Table 4.3: Intra-day Precision and Accuracy of DP and MLX in human Plasma

4.3.3 Recovery

Microextraction procedure was adopted for its added benefits of higher

recovery and sensitivity besides its low solvent consumption and fast. The

method was validated by studying recovery of both drugs from the spiked

plasma samples and it was found that extraction efficiency was better than

conducted in previous studies. The recovery was calculated at three

concentration levels of standard dilutions of 0.01, 1 and 10 µg.mL−1 for DP and

0.015, 1 and 10 µg.mL−1 for MLX (Table 4.4). Peak response of each analyte in

spiked plasma was compared with standard dilution dissolved in the mobile

phase. The extraction efficiency was 99.987% and 99.988% for DP and MLX,

Curve Code LQC (ng.ml-1) MQC (µg.ml-1) HQC (µg.ml-1)

DP MLX DP MLX DP MLX

Nominal 10 15 1.0 1.0 10.0 10.0

Batch-01

9.96 14.94 0.994 0.990 9.97 9.96

9.94 14.92 0.989 0.996 9.92 9.98

9.96 14.88 0.994 0.989 9.94 9.86

9.88 14.95 0.992 0.992 9.95 9.94

9.96 14.92 0.994 0.990 9.88 9.84

9.92 14.94 0.998 0.993 9.87 9.97

9.86 14.98 0.998 0.992 9.95 9.94

9.88 14.96 0.996 0.990 9.88 9.84

9.94 14.94 0.994 0.993 9.87 9.97

Mean 9.94 14.93 0.99 0.99 9.92 9.93

S.D. 0.03 0.03 0.00 0.00 0.04 0.06

%CV 0.32 0.168 0.297 0.260 0.400 0.603

%Bias -0.63 -0.500 -0.650 -0.833 -0.783 -0.750

%Accuracy 99.37 99.50 99.35 99.17 99.22 99.25

Page 104: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

74

respectively. The results were higher than those of 73.1–95.1% in other

published method (Noppamas et al., 2005; Rana et al., 2008; Wen-Hsein et al.,

2009; Lars, 2009; Ali et al., 2008).

Table 4.4: Recovery (Percent Extraction Yield) of DP (a) and MLX (b) in

plasma samples (a)

Curve

Code LQC HQC

EXTRACTED NON-

EXTRACTED EXTRACTED

NON-

EXTRACTED

DP-01

9.88 10.00 9.86 9.92

9.90 10.01 9.88 9.94

9.94 9.99 9.90 9.98

Mean 9.91 10.00 9.88 9.95

S.D. 0.03 0.01 0.02 0.03

%CV 0.308 0.100 0.101 0.153

%PEY 99.07 99.67

(b) Curve

Code LQC HQC

EXTRACTED NON-

EXTRACTED EXTRACTED

NON-EXTRACTED

MLX-01

14.86 14.94 19.84 19.96

14.88 14.95 19.86 19.98

14.90 14.96 19.88 19.95

Mean 14.88 14.95 19.86 19.96

S.D. 0.02 0.01 0.02 0.02

CV 0.134 0.067 0.101 0.077

%PEY 99.53 99.48

Page 105: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

75

4.3.4 Stability

Lowest and highest concentrations (extremes) of each drug were selected for

freezing and thawing. Three cycles freeze-thaw study showed values of

difference were -1.77 (LQC) and -0.543 (HQC) for DP and -4.154 (LQC) and -

2.272 (HQC) for MLX, indicating that samples were stable after freezing and

thawing (Table 4.5). Bench top stability of spiked plasma stored at room

temperature was evaluated for 12 hours (Table 4.6). The values of % C.V. at 12

hour were 0.215 (LQC) and 0.078 (HQC) for DP and 0.815 (LQC) and 0.0213

(HQC) for MLX. The values (mean ± SD) of long-term stability studies were at

LQC 9.893 ± 0.019 for DP and 14.963 ± 0.01 for MLX. The values of MQC

and HQC for both drugs were presented in Table 4.7. It was observed that

plasma samples were stable in three types of studies and comparable to the

previous studies (Sanjay et al., 2006; Lee at., 2000; Silavana et al., 2008;

Young et al., 2005; Limian and Hian, 2001; Rana et al., 2008; Malliou et al.,

2005; Wen-Hsein et al., 2009; Lars, 2009; Ali et al., 2008).

Page 106: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

76

Table 4.5: Freeze-thaw stability DP (a) and MLX (b) in human Plasma (a)

(b)

Cycle

Cycle 0 Cycle 1 Cycle 2 Cycle 3

Curve Code LQC HQC LQC HQC LQC HQC LQC HQC

MLX

14.96 9.86 14.87 9.78 14.45 9.68 13.85 9.75

14.12 9.79 14.78 9.85 14.52 9.81 14.75 9.62

14.97 9.84 14.51 9.71 14.7 9.10 13.62 9.45

Mean 14.683 9.830 14.720 9.780 14.557 9.530 14.073 9.607

S.D. 0.488 0.036 0.187 0.070 0.129 0.378 0.597 0.150

%CV 3.323 0.367 1.273 0.716 0.886 3.967 4.243 1.566

%Difference - - 0.25 -0.51 -0.86 -3.05 -4.15 -2.27

Cycle

Cycle 0 Cycle 1 Cycle 2 Cycle 3

Curve Code LQC HQC LQC HQC LQC HQC LQC HQC

DP

9.68 9.56 9.52 9.81 9.54 9.75 9.46 9.97

9.81 9.98 9.91 9.71 9.42 9.68 9.81 9.64

9.89 9.92 9.42 9.84 9.83 9.79 9.59 9.69

Mean 9.79 9.82 9.62 9.79 9.60 9.74 9.62 9.77

S.D. 0.106 0.227 0.259 0.068 0.211 0.056 0.177 0.178

%CV 1.082 2.313 2.692 0.696 2.197 0.572 1.839 1.821

%Difference - - -1.80 -0.34 -2.01 -0.82 -1.77 -0.54

Page 107: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

77

Table 4.6: Bench top stability DP (a) and MLX (b) in human Plasma (a)

(b)

Curve Code 0 h 3 h 6 h 12 h

LQC HQC LQC HQC LQC HQC LQC HQC

MLX-01

15.013 9.989 14.773 9.982 14.656 9.971 14.613 9.967

15.000 9.973 14.879 9.965 14.864 9.974 14.844 9.964

14.990 9.977 14.982 9.961 14.974 9.984 14.786 9.956

Mean 15.001 9.980 14.878 9.969 14.831 9.976 14.748 9.966

S.D. 0.012 0.0083 0.105 0.0112 0.162 0.007 0.120 0.002

%CV 0.077 0.0834 0.702 0.1118 1.089 1900.0 0.815 0.021

%Difference - - -0.82 -0.10 -1.13 -0.04 -1.69 -0.15

Curve Code 0 h 3 h 6h 12 h

LQC HQC LQC HQC LQC HQC LQC HQC

DP-01

10.013 9.990 9.968 9.989 9.968 9.989 10.013 9.990

10.000 9.980 9.997 9.970 9.997 9.970 10.000 9.980

10.042 9.970 9.992 9.960 9.992 9.960 10.042 9.970

Mean 10.0183 9.980 9.9857 9.973 9.9857 9.973 10.018 9.985

S.D. 0.022 0.010 0.016 0.015 0.016 0.015 0.022 0.007

%CV 0.2146 0.1002 0.1552 0.1477 0.155 0.148 0.215 0.071

%Difference - - -0.33 -0.07 -0.33 -0.07 0.00 0.05

Page 108: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

78

Table 4.7: Long term stability DP and MLX in human Plasma

4.4 Application of present method

The present method was validated successfully and applied for the

simultaneous determination of DP and MLX in pharmaceutical drug products,

advanced drug delivery system and human plasma drug analysis (Table 4.8).

Table 4.8: Application of method in analysis of liposomes, marketed brands and human plasma

SEM= standard error of mean (n=3)

Conc. Units Week 1 Week 2 Week 3 Week 4 Mean S.D. %CV

DP LQC Ng.mL-1 9.92 9.88 9.89 9.88 9.89 0.019 0.191

DP MQC µg.mL-1 0.99 0.97 0.96 0.97 0.97 0.013 1.294

DP LQC µg.mL-1 4.98 4.98 4.96 4.95 4.97 0.015 0.302

MLX LQC Ng.mL-1 14.98 14.97 14.96 14.94 14.96 0.017 0.114

MLX MQC µg.mL-1 0.96 0.95 0.95 0.94 0.950 0.008 0.082

MLX HQC µg.mL-1 9.98 9.96 9.94 9.94 9.955 0.019 0.192

Samples Drug/Brands Stated Found Percentage

± SEM

Liposome DP 50 (mg) 49.94 (mg) 99.92 ± 0.014

Marketed Brands

DP

Caflam ® Novartis 50 49.94 99.88 ± 0.011

Diclorep® Sami 50 49.96 99.92 ± 0.010

Maxit® Hilton 50 99.98 99.96 ± 0.009

MLX Xobix® Hilton 7.5 7.46 99.47 ± 0.011

Melox® Kobec 7.5 7.43 99.10 ± 0.012 Artex® Pharmadic 7.5 7.44 99.20 ± 0.010

Plasma

DP

10 (ng) 9.97 99.70 ± 0.011

1 (µg) 0.998 99.80 ± 0.012

10 (µg) 9.996 99.96 ± 0.089

MLX

15 (ng) 14.89 99.26 ± 0.009

1 (µg) 0.997 99.70 ± 0.010

10 (µg) 9.965 99.65 ± 0.009

Page 109: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

79

4.5 Conclusions

The present research revealed simple and fast RP-HPLC method for the

simultaneous determination of DP and MLX with DAD detection. The

validation parameters were in acceptable limits of guidelines of FDA and ICH.

The Micro extraction procedure was simple and fast, less time consuming and

new composition of mobile phase was enable the method to detect DP and

MLX in shorter period of time. The method will be suitable for routine analysis

of DP and MLX in routine quality control analysis in pharmaceutical industry,

Bioequivalence and pharmacokinetics studies as well as therapeutic drug

monitoring of these drugs in optimizing dosage regimen.

Page 110: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

80

Chapter No. 5

5.1 Introduction

Liposomes are considered as universal drug carriers and used for enhancing

efficacy and pharmacokinetics profile of drugs (Zuzanna et al., 2010).

Phospholipids and cholesterol are main formulating ingredients of liposomes

for entrapping active therapeutic moiety for potential delivery of drugs with

enhanced efficacy. Optimum concentration of these combined materials may

affect the entrapment efficiency of active substances. In order to achieve

maximum entrapment and optimum release profile, the relative amount of

cholesterol and phospholipids has to be adjusted and optimized. It is revealed

in literature that increasing concentration of cholesterol may decrease percent

drug entrapment with a subsequent lowering of phospholipids contents

(Shivhare et al., 2009). Further, the drug entrapment of drugs increases as the

phospholipids contents are increased due to saturation of lipid bilayer with

relation to drug, while the low phospholipid contents provide limited

entrapment capacity (Gregoriadis, 2007). Therefore, objective of this study was

to evaluate the effect of formulating variables (cholesterol and phospholipids)

on the entrapment efficiency of drug using a statistical model, Response

Surface Methodology (R.S.M.) for determining the optimum concentration of

variables.

Since its utilization in applied field after 1966, the response-surface

methodology (R.S.M.) has made its place in many fields. One of its utilization

Page 111: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

81

is the optimization of multifactor in design and development of drug delivery

systems with higher accuracy and efficiency (Mayer et al., 1989).

Diclofenac sodium (DFS), a commonly used non-steroidal anti-inflammatory

drug (NSAID), employed in therapeutics since 1970s. It exhibits potent

analgesic and anti-inflammatory properties by impairing prostaglandin

synthesis by inhibition of the cyclooxygenase isozymes COX-1 and COX-2. It

is widely used in the treatment of various inflammatory diseases such as

arthritis and osteoarthritis (Todd and Sorkin, 1988; Laurence, 2007). DFS was

entrapped with different combinations cholesterol and phospholipids in order to

get a formulation containing optimum concentration of structural components

in the study design which is described in coming section.

5.2 Materials and Methods

5.2.1. Materials

Diclofenac sodium (DFS) was taken as research material donation from

Novartis Pharmaceutical (Pakistan) Limited, Karachi-Pakistan. Cholesterol

(CH) and Purified Soya Lecithin (PSL) was obtained from Sigma-Aldrich

Chemie GmbH, Germany. Chloroform, Ethanol, Methanol (HPLC grade,

purchased from Sigma-Aldrich Chemie GmbH, Germany). Distilled water was

prepared by double distillation process in the Department of Pharmacy, the

Islamia University of Bahawalpur.

Page 112: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

82

5.2.2. Preparation of liposomes

Method 1: Micro-emulsification evaporation method (MEE): The newly

developed method in the present study, described in detail in Chapter 6. The

brief description is given as under:

i. DFS (10 gm) was dissolved in 50 ml mixture of water and

methanol (49.5:0.5 v/v) and the solution was sonicated

(Transsonic ELMA, Singapore) for 2 minutes until clear solution

obtained and was designated as solution A.

ii. The cholesterol and soya lecithin (SL) was taken as 10 gm,

dissolved in 50 ml mixture of chloroform/ethanol (1:1 v/v) and

the solution was sonicated for 2 minutes until clear yellowish

solution obtained and was designated as solution B.

iii. Solution A was then added slowly into solution B under

homogenization (Heidolph Instruments GmBH and Co.,

Germany) at a speed of 8000 rpm for 30 minutes. A milky

suspension with clear texture was resulted.

iv. Organic solvents were removed from dispersion by rotary

evaporation technique (Laborota 4000, Heidolph Germany) under

reduced pressure. The temperature was set at 40°C.

v. The suspension was freeze-dried (Christ GmBH, Germany) and

white to yellowish powdered free flowing liposomes were

obtained.

All the formulations of central composite design of R.S.M. were prepared by

MEE method. The other two methods described below were used to compare

entrapment efficiency of optimized formulation (Formulation No. 9).

Page 113: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

83

Method 2: Thin Layer Hydration method (TLH): This method involves two

steps procedure for the preparation of liposomes. The procedure described by

Bangham et al., (1965) was used to prepared liposomes of DFS.

i. In first step, quantity of the constituting lipid phase, i.e.,

cholesterol and soya lecithin were taken in 10 gm in the ratio of

(25:75) and dissolved in 50mL chloroform. The solution was

transferred to the flask of thermostat controlled rotary evaporator

(Laborota 4000, Heidolph Germany), preset at 40ºC. The dry

lipid film was deposited on the walls of rotary flask.

ii. In second step, DFS was dissolved in water with some amount of

methanol. The drug solution was added to the dry thin film to

form drug loaded liposomes. The resultant liposomes were

processed for the determination of entrapment of DFS.

Method 3: Reverse Phase Evaporation Method (REV): This method involves

five steps procedure for the preparation of liposomes.

i. The DFS liposomes were prepared by reverse-phase evaporation

method as previously described by Szoka and Papahadjopoulos,

(1978) and Pleumchitt et al., (2003) after incorporation of

changes. 50 mg diclofenac sodium was taken and dissolved and

adjusted to 50 mL with water (added with small fraction of

methanol in order to enhance solubility).

ii. The cholesterol and purified soya lecithin (25:75) were taken in

round bottom flask and dissolved in mixture of chloroform and

Page 114: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

84

ethanol (50:50). The mixture was warmed at 40ºC under reduced

pressure in a rotary evaporator (Laborota 4000, Heidolph

Germany) until a dry, uniform, thin film on the inner wall of the

flask was obtained.

iii. The phospholipid contents were withdrawn from flask and placed

in an ultrasonic bath (Transsonic ELMA, Singapore). The drug

solution prepared in step (i) was mixed with lipid contents of

flask. This mixture was sonicated for 10 min. to form w/o

emulsion.

iv. The emulsion was again transferred to a rotary evaporator to

slowly evaporate the solvent under reduced pressure at 25ºC until

a viscous gel was formed, which subsequently became a

homogeneous aqueous suspension. The evaporation process was

further continued for at least 30 min. at 30-35ºC to ensure

complete removal of all traces of organic solvent.

This liposomal suspension was finally dispersed in 15 mL phosphated buffered

saline solution and subjected to determine percent entrapment of DFS.

5.2.3. Entrapment Efficiency (EE)

Entrapment efficiency of diclofenac was determined by centrifugation method.

This was previously described by Shivhare et al., (2009) and El-Samaligy et al.,

(2006). A portion equal to 50 mg of liposomes of diclofenac DFS were taken

(separately) and dissolved in 10 ml of BPS to make a homogenized dispersion.

The dispersion was subjected to centrifugation at 5000 rpm for 30 minutes. The

Page 115: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

85

clear supernatant was carefully removed to bring separation of the un-

entrapped diclofenac. The sediment in centrifuge tube was diluted to 100 ml of

PBS (maintained at pH 7.4). The analysis was performed for liposomal

formulations of DFS using different methods which were developed and

validated as part of this study. The chromatographic system of Agilent

Technologies (series 1200) with Series 1200 pumps, Series 1200 variable

wavelength UV-VIS (Programmable) diode array detector (DAD)

automatically set at wavelengths of 282 nm, using reversed phase mode

employed for the determination of DFS in liposomal formulations (detailed

description has been given in chapter 3). The percent entrapment was

determined by the same procedures as described in analysis of DFS and DFP

and calculated by equation:

100 DiclofnacTotal

Sediment in c DiclofenaFree (%)y EfficiencEntrapment ×=

5.2.4. Particle Size measurement and size distribution

Dynamic light scattering measurements employing Zetasizer (Malvern., UK)

was used for determination of mean particle size, size distribution,

polydispersity index and zeta potential. Distilled de-ionized water filtered

through syringe filter (0.22 micron) for sample preparation by the dilution of

liposomes suspensions. Measurements were made as mean ± standard error of

mean (S.E.M.) in triplicates.

5.3 Study Design for liposomal formulations

Response surface methodology (R.S.M.) is being employed in pharmaceutical

research for optimization variables of formulations. Experimental design,

Page 116: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

86

regression analysis, constraints optimization and validation are important tasks

in applying R.S.M. (Hagalavadi et al., 2007; Gregg Stetsko, 1986). By use of

R.S.M, efficient and economical method can be developed to find the required

information to understand relationships and design a mathematical model

which can be used to characterize and optimize a formulation or process.

Without the use of a mathematical model, it is unlikely that the optimum

formulation or process conditions can be determined (Gregg Stetsko, 1986).

A central composite rotatable design (CCD) with two independent variables i.e

Cholesterol (X1) and phospholipids (X2) at five levels was employed at α equal

to 1.267. The percent entrapment was taken as dependent (Y) variable. The

central composite design for two independent variables is given in Table 5.1

(coded values) and Table 5.2 (decoded of actual values).

Page 117: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

87

Table 5.1: The composition and observed response from randomized runs in central composite design (Coded values)

Run Factor 1 (X1) Factor 2 (X2) Response (Y)

Cholesterol (mg) B:SL (mg) Entrapment (%)

1 0 0 54.27 2 0 0 55.49 3 1.267 0 42.25 4 1 -1 39.47 5 1 1 48.25 6 0 0 55.87 7 0 1.267 78.47 8 0 -1.267 37.14 9 -1 1 82.56

10 -1.267 0 65.45 11 0 0 56.89 12 0 0 55.24 13 -1 -1 45.87

Table 5.2: The composition and observed response from randomized runs

in central composite design (Actual values)

Run

Variable 1 Variable 2 Response

A:cholestrol B:SL Entrapment

(mg) (mg) (%)

1 37.5 50 54.27 2 37.5 50 55.49 3 53.33 50 42.25 4 50 25 39.47 5 50 75 48.25 6 37.5 50 55.87 7 37.5 81.67 78.47 8 37.5 18.32 37.14 9 25 75 82.56 10 21.66 50 65.45 11 37.5 50 56.89 12 37.5 50 55.24 13 25 25 45.87

The coded level used were −1.267, -1, 0, +1 and +1.267, respectively, and the

corresponding actual values for each variable are listed in Table 5.3.

Page 118: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

88

Table 5.3: Coded level values for X1 and X2

1.267 1 0 -1 -1.267

Cholesterol 53.33879 50 37.5 25 18.32241

PSL 81.67759 75 50 25 21.66121

The quadratic model was used for correlation between the independent

variables and response and final equation in terms of coded factors is as

follows.

Y = b0 + b1X1 + b2X1 + b12X1X2 + b11X12 + b22X2

2

where Y is % entrapment, b0 is the average results of the replicated center

point; b1 and b2 are the main half-effects of coded variables X1 and X2,

respectively; b11 and b22 are the squared effects; b12 is the factor interaction

half-effect.

Design and analysis of central composite experiment were carried out with

Design Expert software (stat Ease Inc.) version 8.0.1 version. The central

composite design with two variables, including the factors, their levels, and the

result from each test is shown in Table 5.4.

Page 119: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

89

Table 5.4: Standardized main effects of variable on the responses

Factor Estimate df Error Low High

Intercept 55.69 1 1.23 52.77

A-cholesterol -9.72 1 1.04 -12.17

B-SL 13.57 1 1.04 11.12

AB -6.98 1 1.39 -10.27

A2 -1.7 1 1.23 -4.6

B2 0.76 1 1.23 -2.14

5.4 Results and Discussion Analysis of variance (ANOVA) and estimated regression coefficient were

applied to evaluate response Y. A series of experiments was carried out by

considering a central composite design (Table 5.2). The standard error of

coefficient is a measure of the variation in estimating the coefficient, the

polynomial model was postulated based on Table 5.3. Various statistical data

(standard error of estimate, sum of squares of the errors, F statistics, and p

value) were examined. The effects of squared terms were found as statistically

significant. The coefficients of the nonlinear polynomial model (Equation 1)

and p value were shown in Table 5.5. The response factors at any regime in the

interval of our experiment design can be calculated using the following

Equation.

Y = 55.69-9.7X1 + 13.57X2 -6.98X1X2-1.70X12 + 0.76X2

2

Page 120: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

90

Table 5.5: Summary of results of model analysis and lack of fit for measured responses

Source Sum of Squares df Mean square F value p-value

Model 2221.72 5 444.34 57.28 < 0.0001

A-cholesterol 681.58 1 681.58 87.87 < 0.0001

B-SL 1327.47 1 1327.47 171.13 < 0.0001

AB 194.74 1 194.74 25.1 0.0015

A2 14.93 1 14.93 1.93 0.2079

B2 2.99 1 2.99 0.39 0.5544

Residual 54.3 7 7.76 ------- ------

Lack of Fit 50.66 3 16.89 18.58 0.0082

Pure Error 3.64 4 0.91

Total 2276.02 12

5.4.1 Model validation

Experimental results and the predicted values obtained using nonlinear

polynomial model (Equation 2) are given in (Figure 5.1). It is can be seen, the

predicted values match the experimental values reasonably well with R-Square

of 0.976 and R-Square (adjusted) of 0.959 for response Y.

Page 121: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

91

Figure 5.1: Linear correlation plots between Predicted vs. Actual Response

values for response Y 5.4.2 Response surface (contour) plots

In order to gain insight about the effect of each variable, the three dimensional

(3D) and contour (2D) plots for the predicted responses were also formed,

based on the model polynomial function to analyze the change in response

surface as shown in Figure 5.2 and Figure 5.3. These figures show the 2D and

3D plots relationship between two variables and response Y (% Entrapment) at

center level of other variable. Figure 5.3 shows the effect of cholesterol

concentration (X1) and Phospholipid (X2) on % Entrapment (Y).

Actual

Pre

dic

ted

Predicted vs. Actual

30.00

40.00

50.00

60.00

70.00

80.00

90.00

30.00 40.00 50.00 60.00 70.00 80.00 90.00

Page 122: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

92

Figure 5.2: Contour curve plot showing the influence of Cholesterol and Soya

lecithin on response Y (Percent drug entrapment)

Figure 5.3: Response Surface plot showing the effect of Cholesterol and Soya

lecithin on response Y (Percent drug entrapment)

25.00 30.00 35.00 40.00 45.00 50.00

25.00

30.00

35.00

40.00

45.00

50.00

55.00

60.00

65.00

70.00

75.00entrapment

A: cholestrol

B:

SL

40

50

60

70

80

5

25.00

30.00

35.00

40.00

45.00

50.00

25.00 30.00

35.00 40.00

45.00 50.00

55.00 60.00

65.00 70.00

75.00

30

40

50

60

70

80

90

entr

apm

ent

A: cholestrol B: SL

Page 123: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

93

5.4.3 Response optimization

The result of response surface and contour plots are shown in Figure 5.2 and

5.3. This result demonstrates that all the response surfaces have the maximum

points. Therefore, response surface optimization could be found depending on

cholesterol concentration (X1) and concentration of phospholipids (X2), for

gaining maximum entrapment efficiency in liposomal formulations. Response

surface methodology using CCD for two variables at five levels have

demonstrated that entrapment level was found as maximum at concentration of

X1 25 mg and concentration of X2 75 mg with an entrapped amount equal to

82.56% of active drug.

The results of response surface methodology were found in accordance with

the studies available in literature. A study for incorporation of cholesterol and

phophadidylcholine was conducted by Shivhare et al., (2009) stated influence

of cholesterol as retardant for entrapment of drugs. Although cholesterol

increases stability and rigidity of liposomes but at the same time, the increasing

concentration reduces the entrapment efficiency of liposomal formulations.

Further, it was also studied that increasing concentrations of phospholipids

increased entrapment efficiency by enhancing retaining of drugs in the aqueous

core of liposomes.

5.5 Comparison of different methods of preparations of liposomes

The comparison study was conducted to evaluate percent drug entrapment in

liposomal formulations prepared with three different techniques i.e. micro

emulsification-evaporation (MEE) followed by freeze-dying; reverse phase

Page 124: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

94

evaporation and thin layer hydration method. The parameters evaluated were

particle size, polydispersity index and zeta potential. It was observed that

microemulasification-evaporation technique produced liposomes with highest

entrapment of 82.56% (±0.020 as S.E.M), where as the entrapment values were

56.32 (± 0.027, S.E.M.) and 62.71 (± 0.031 as S.E.M value) for thin layer

hydration (TLH) method and reverse phase evaporation (REV) method (Table

5.6).

Percent entrapment was highest with MEE method than other two counterpart

methods. The reason for this higher entrapment was the selection of optimum

concentration of soya lecithin and cholesterol, appropriate organic solvents as

well as their ratio to make liposomes of desired characteristics. Further, after

emulsification and evaporation process, liposomes were subjected to freeze-

drying which also enhances entrapment capacity of liposomes. Whereas,

reverse phase evaporation (REV) method was also found to be efficient process

than thin layer hydration (TLH) method in selecting appropriate ratios of

aqueous volume to organic phase volume to obtain higher entrapment (Figure

5.4). The size of vesicles prepared by MEE, REV and TLH methods were 1.10

± 0.009, 1.25 ± 0.013 and 1.28 ± 0.015 (mean ± SEM) where as poly dispersity

index was 0.139 ± 0.006, 0.328 ± 0.014 and 0.276 ± 0.017. MEE method

produced liposomes of relatively less size and uniform size distribution

ensuring the uniformity of production of liposomes (Figure 5.5–5.7). The

smaller vesicle size has advantage of higher release profile of liposomes was

found in accordance with the study available in literature describing the effect

Page 125: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

95

of small particle on the release behavior of delivery system. The small vesicle

size provides higher surface area for the release at the site of action and

produce higher bioavailability (Odeberg et al., 2003). Another property of

liposomes formulations is the zeta potential which is the measure of stability of

the formed liposomes. The values of zeta potential for the liposomal

formulations prepared by MEE, REV and TLH methods were 51.58 ± 0.09,

36.29 ± 0.12 and 32.67 ± 0.11 (mean ± SEM). The values of zeta potential

were higher for liposomes of MEE method showing more stability of formed

liposomes as compared to REV and TLH method (Figure 5.8–5.10).

Table 5.6: Comparison of different methods of preparation for evaluation of characteristics of liposomes (n=3)

Sr.

No. Parameter MEE TLH REV

1 Particle size 1.10 ± 0.009 1.25 ± 0.013 1.28 ± 0.015

2 Polydispersity index

0.139 ± 0.006 0.328 ± 0.014 0.276 ± 0.017

3 Zeta potential (ζ) 51.58 ± 0.09 36.29 ± 0.12 32.67 ± 0.11

4 EE (%, w/w) 82.56 ± 0.020 56.32 ± 0.027 62.71 ± 0.031

Page 126: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

96

Figure 5.4: Comparison of entrapment efficiencies obtained with different

method of preparation of Liposomes.

Figure 5.5: Size range of the selected liposomal formulation (09) prepared with

MEE method (Zeta Sizer, Malvern, UK)

82.56

56.3262.71

0

15

30

45

60

75

90

MEE TLH REV

Method of Preparation

En

tra

pm

ent

Eff

icie

ncy

(%

)

Page 127: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

97

Figure 5.6: Size range of the selected liposomal formulation (09) prepared with

TLH method (Zeta Sizer, Malvern, UK)

Figure 5.7: Size range of the selected liposomal formulation (09) prepared with

REV method (Zeta Sizer, Malvern, UK)

Page 128: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

98

Figure 5.8: Size range of the selected liposomal formulation (09) prepared

with REV method (Zeta Sizer, Malvern, UK)

Figure 5.9: Zeta potential of the selected liposomal formulation (09) prepared

with MEE method (Zeta Sizer, Malvern, UK)

Page 129: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

99

Figure 5.10: Zeta potential of the selected liposomal formulation (09) prepared

with REV method (Zeta Sizer, Malvern, UK)

5.6 Conclusion

A central composite rotatable design with two factors at five levels using

different ratio of cholesterol and soya lecithin was employed for optimization

of liposome preparations of DFS. The quantitative effects of the factors at

different levels on the entrapment of DFS could be predicted by using

polynomial equations. The observed responses were in close agreement with

the predicted values for optimized formulations. The quadratic RSM applied

helped in understanding of the interaction between two factors and its effect on

the entrapment of DFS in liposomal formulation. The RSM fruited out an

optimized liposomal formulation consisted of cholesterol (25) and soya lecithin

(75) with maximum entrapment efficiency of 82.56%. Further, the comparison

of method of preparation demonstrated that MEE method was most appropriate

for the preparation of liposomes than TLH and REV, with added benefits of

high entrapment as well as stability and higher bioavailability.

Page 130: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

100

Chapter No. 6

6.1. Introduction

Liposomes consist of hydrophilic and hydrophobic layers, spherical in shape

and can be adopted for incorporation of water soluble as well as water

insoluble drugs for producing sustained actions (Tomoko and Fumiyoshi, 2005;

Gorka et al., 2005; Zucker, 2009). Various articles published on liposomes

demonstrate delivery of drugs to various systems such as pulmonary (Efstathios

et al., 2005), ophthalmic (Omaima et al., 1997; Law and Hung, 1998)

transdermal (Francesca et al., 2006; Ebtessam et al., 2002), buccal (El-

Samaligy et al., 2006), mucoadhesive and oral (Takeuchi et al., 2003; El-

Samaligy et al., 2004; Filipovic et al., 2001; Hongming and Robert, 1998),

vaginal (Zeljka et al., 2006) and parenterals (Gruber et al., 2000) . Articles

available on natural and synthetic phospholipids materials declaring them as

preferable candidate for liposomal drug delivery systems (LDDS)

(Humberstone and Charman, 1997; Theresa and Elaine, 1996; Sriram and

Rhodes, 1995; Boris and Lasic, 1996).

Numerous procedures have been used for the fabrication of liposomes

including thin layer hydration (Tsotas et al., 2007; Bhalerao and Harshal, 2003;

Maria et al., 2005), reverse phase solvent-evaporation (Pleumchitt et al., 2003),

micro-fluidization technology (Kosaraju et al., 2006), sonication (Sivakumar

and Panduranga, 2000), spray freeze-dried (Lyle et al., 2005), ethanol injection

(Minghui et al., 2008) and microencapsulation vesicle methods (Tomoko et al.,

2002, Tomoko and Fumiyoshi, 2005). Although different methods produce

Page 131: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

101

liposomes of desired characteristics but there exist a problem in using large

volume organic solvents which is considered hazardous for health of patients

and formulation experts and the environment during production of liposomes.

Thin layer hydration, Sonication and Extrusion methods are widely used for

preparing liposomes. However, the applications of these techniques were

greatly limited due to stability, lows entrapment efficiencies and less residence

time of formed liposomes. Certain methods have less efficiency in removal of

toxic solvents (ethers, ethyl acetate, methanol and dichloromethane).

In the present study, we have developed new promising method called the

micro-emulsification evaporation (MEE) followed by freeze drying. The

technical characteristic of this method is the use of combination techniques that

are considered best for the preparation of liposomes. The incorporation of

freeze drying provides desired characteristics of uniform particle size, stability,

elegance of produced liposomes and short reconstitution time. Another

important characteristic of the present method is the use of combination

organic solvents, chloroform/ethanol instead of chloroform alone, which is

used in the previous studies (Tomoko and Fumiyoshi, 2005; Tomoko et al.,

2002).

Diclofenac, marketed as sodium, potassium (oral) and diethyl amine

(transdermal) salts of non-steroidal anti-inflammatory drugs (NSAID) belong

to fenamate class exhibit anti-inflammatory, analgesic and antipyretic action by

reversible inhibition of prostaglandin synthesis. These salts employed in

Page 132: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

102

different pain and rheumatic conditions such as rheumatoid arthritis,

osteoarthritis, ankylosing spondylitis, non articulate rheumatism and spot

injuries. Toxicity of the central nervous, gastrointestinal, renal and

cardiovascular system occurs after long-term use in various arthritic conditions.

The bioavailability of sodium and potassium salts of diclofenac by oral

administration is low and about 60 percent of the dose can be accessed in

systemic circulation. Therefore, diclofenac has been classified as BCS-II drug

having less solubility in water (Chuasuwan et al., 2009; Sweatman, 2009;

Sajeev et al., 2002).

To overcome the problems of low solubility and bioavailability, an attempt has

been made to formulate sodium and potassium salts of diclofenac into

liposomes. The low solubility was enhanced by the addition of fractions of

methanol in water, purified soya lecithin and purified egg lecithin were

dissolved and homogenized in combination of organic solvents

(chloroform/ethanol). Chloroform has been used in pharmaceutical

formulations because it is less toxic than other solvents such as

dichloromethane, ethyl acetate and diethyl ether. Ethanol was also preferred,

because it is biocompatible, largely used in pharmaceutical preparations and

forms a monophonic system with water. Therefore, not requiring sonication

step for preparation of liposomes.

The mixture of drug and lecithin was subjected to emulsification and

evaporation process. The method was further modified by subjecting

Page 133: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

103

suspension obtained after vacuum evaporation to freeze-drying process which

leads to the production of solid liposomes. In this work, the effect of poly

ethylene glycol was also observed to extend the release of diclofenac loaded

liposomes.

The formulated liposomes of diclofenac sodium and potassium salts were

characterized for measurement of size, size distribution and zeta potential using

Zetasizer; scanning electron microscopic (SEM) studies for morphological

analysis; differential scanning calorimetry (DSC) and X-ray powder

diffractometry for the physical state of drug in liposomes; Fourier transform

infra-red spectroscopy (FT-IR) for the determination of interaction between the

drug and phospholipids. The reverse phase high-performance liquid

chromatography (RP-HPLC) was used for determining the drug encapsulation

efficiency (EE) and in-vitro release of DFS and DFP liposomes. The

experimentation was conducted for in-vivo evaluation of liposomal

formulations after f-2 test of in-vitro data for formulations and marketed brand.

6.2. Experimental

6.2.1. Materials

Diclofenac sodium (DFS) and Diclofenac potassium (DFP) was supplied by

Novartis Pharmaceutical (Pakistan) Limited, Karachi-Pakistan. Purified Soya

Lecithin (PSL) and Purified Egg Lecithin (PEL) were obtained from Sigma-

Aldrich Chemie GmbH, Germany. Poly Ethylene Glycol (PEG) was purchased

Page 134: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

104

from Merck, USA. Methanol (HPLC grade) was provided from Sigma-Aldrich

Chemie GmbH, Germany. All other chemicals and solvents were of reagent

grade. Distilled water was prepared by double distillation process in the

Department of Pharmacy, the Islamia University of Bahawalpur.

6.2.2. Preparation of liposomes

The liposomes of diclofenac (DFS and DFP) were prepared according to the

procedure described as follows:

1. DFS and DFP (each 10 gm, taken separately in each experiment) was

dissolved in 50 ml mixture of water and methanol (49.5:0.5 v/v) and the

solution was sonicated (Transsonic ELMA, Singapore) for 2 minutes

until clear solution obtained. It was designated as solution A.

2. The purified soya lecithin (PSL) and purified egg lecithin (PEL) were

taken separately in 10 gm, were dissolved in 50 ml of mixture of

chloroform/ethanol (1:1 v/v) and the solution was sonicated for 2

minutes until clear yellowish solution obtained. It was designated as

solution B.

3. Solution A was then added slowly into the solution B under

homogenization (Heidolph Instruments GmBH and Co., Germany) at a

speed of 8000 rpm for 30 minutes. A milky suspension with clear

texture was obtained.

Page 135: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

105

4. Organic solvents were removed from dispersion by rotary evaporation

technique (Laborota 4000, Heidolph Germany) under reduced pressure.

The temperature was set at 40°C.

5. The suspension was freeze-dried (Christ GmBH, Germany) and white to

yellowish powdered free flowing liposomes were obtained.

6. The formulation containing poly ethylene glycols were prepared by

incorporating PEG water dispersion before evaporation step.

Using micro-emulsification evaporation method (MEE), all formulations

(n=24) of DFS and DFP using PSL and PEL using fixed ratio were prepared

following same composition described in Table 6.1.

6.2.3. Morphology of liposomes

The shape, surface morphology and size of the produced liposomes were

investigated by scanning electron microscope (SEM, JSM-5310 LV Scanning

Microscope). The liposomes samples were diluted with PBS buffer in 1 to 10

ratios and Negative staining was employed using 2%, (w/v) phospho-tungstic

acid (PTA) and drying at room temperature before fixing to microscope. Before

investigation, the liposomes samples were flatted on a double side sticky tape

that was sticked on the standard sample stand.

6.2.4. Particle Size measurement and size distribution

The mean particle size, size distribution, polydispersity index and zeta potential

of liposomes were assessed by particle size analyzer named as Zetasizer

Page 136: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

106

(Malvern., UK) Samples were prepared by dilution of liposomes suspensions

with appropriately distilled de-ionized water filtered through syringe filter

(0.22 micron). Each value reported was the mean values ± standard error of

mean (S.E.M) for three replicate samples.

6.2.5. Differential Scanning Calorimetric Analysis

Differential scanning calorimetry analysis (Q200, TA instruments) was used

for both analgesics (DFS and DFP) loaded liposomes for physical state

characterization with thermal transitions. 2 mg of sample was taken in

aluminum pans, sealed and protected with pure dry Argon gas. Properties

measured by TA Instruments DSC (Differential Scanning Calorimeter)

techniques include glass transitions, "cold" crystallization, phase changes,

melting, crystallization, product stability. All samples were scanned at a

temperature charge of 5°C/min and the heat flow from 0 to 300°C. Standard

reference (Indium) was used to calibrate the temperature and energy scale of

instrument.

6.2.6. X-ray diffractometry (XRD)

Freeze-dried samples of all formulations of DFS/DFP-liposomes were

evaluated for drug crystalline state by powder XRD (D8 Discover Bruker,

Germany). In this technique, 40 kV and a current of 50 mA for the generator

were applied with Cu as the tube anode material. The solids were exposed to a

Cu-K radiation, over a range of 2θ angles from 0° to 35°, at an angular speed of

2° (2θ)/min.

Page 137: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

107

6.2.7. FT-IR studies

IR spectra of DFS/DFP, PSL/PEL and formulations of liposomes (PL-01) were

obtained by FT-IR (Perkin Elmer), using KBr pellet disk technique. All

samples of selected formulations were scanned in the IR range from 4000-600

cm-1. The apparatus was set at absorbance scale and spectra of selected

formulations were taken as representative of all formulations.

6.2.8. Entrapment Efficiency (EE)

Entrapment efficiency of diclofenac was determined by centrifugation method.

This was previously described by Shivhare et al., (2009) and El-Samaligy et al.,

(2006). A portion equal to 100 mg of liposomes of diclofenac (both DFS and

DFP) were taken (separately) and dissolved in 5 ml of BPS to make a

homogenized dispersion. The dispersion was subjected to centrifugation at

5000 rpm for 30 minutes. The clear supernatant was carefully removed to bring

separation of un-entrapped diclofenac. The sediment in centrifuge tube was

diluted to 100 ml of PBS (maintained at pH of 7.4). The analysis was

performed for liposomal formulations of diclofenac (DFS and DFP) using

different methods which were developed and validated as part of this study.

The chromatographic system of Agilent Technologies (series 1200) with Series

1200 pumps, Series 1200 variable wavelength UV-VIS (Programmable) diode

array detector (DAD) which was automatically set at wavelengths of 282 nm,

using reversed phase mode was employed for the determination of DFS in

liposomal formulations (detailed description has been given in chapter 3).

Whereas, another method was developed for the determination of DFP

Page 138: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

108

liposomal formulations (detailed description has been given in chapter 4). The

percent entrapment was determined by same procedures as described in

analysis of DFS and DFP and percent entrapment was determined by following

equation.

100 DiclofnacTotal

Sediment in c DiclofenaFree (%)y EfficiencEntrapment ×= (1)

6.2.9 Stability studies of liposomal formulations of diclofenac

Duplicate samples of the selected formulations of DFS and DFP were sealed in

glass containers and then placed in temperature-controlled stability chambers

(Sanyu, Japan) at 30°C, 40°C and temperature/humidity controlled stability

chamber at 40°C/75% RH for the duration of six months. Samples were

withdrawn at 0, 1, 2, 3, and 6 months for the evaluation of physical and

chemical stabilities. The physical stability was evaluated by visual inspection

for physical changes (such as phase separation & drug precipitation), and

percent drug content & T60 (time for 60% of the drug release) for chemical

stability which were determined by the validated procedures described in

chapter 3 and 4. The release profile was determined by procedure described for

in-vitro evaluation of drugs.

6.2.10 In vitro release study

The release of DFS and DFP from all liposome formulations were performed

by dissolution test. 5 mg of DFS-loaded liposomes were suspended in 5 ml

phosphate buffer solution (PBS, pH 7.4) and placed in the vessels of USP

dissolution apparatus II (Pharma Test, Germany), set at continuous stirring

speed at 50 rpm. Dissolution test was conducted in 900 ml of 0.1M buffer

Page 139: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

109

solution at a maintained temperature of 37 ± 0.5°C. At pre-determined time

intervals (0, 0.5, 1, 1.5, 2, 3, 4, 6, 8, 10, 12, 16, 20 and 24 hours) an aliquot

amount (5 ml) of dissolution media was drawn by an automatic sampler unit

(Pharma Test, Germany). All the samples were analyzed by RP-HPLC analysis

was carried out as described for DFS and DFP in chapter 3 and 4, respectively.

In the data analysis, cumulative percentage of DFS release from liposomes was

calculated using a mean of three samples values.

6.2.10.1 In-vitro Kinetics and modeling

All formulations (Table 6.2) were investigated for release of DFS and DFP

from liposomes. Different models were employed such as zero order and first

order to determine order of release by higuchi, Hixon Crowell and pappa’s to

determine the mechanism of release from liposomal formulations of diclofenac

salts (DFS and DFP).

6.2.11. In-vivo Studies

6.2.11.1 Subject selection, clinical design, drug administration and

sampling schedule

The study protocol was approved by The Pharmacy Research Ethics

Committee (PREC) of the Department of Pharmacy, the Islamia University of

Bahawalpur. Eighteen healthy adult, male, human subjects with mean age

group 23.4 ± 0.081 years and average weight 61.67 ± 6.89 kg were included in

the study. Subjects were excluded from the study if one more or criteria were

present at time of medical screening: allergic to diclofenac, history or clinical

data of renal or liver disease, positive test for hepatitis B, HIV, history of

Page 140: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

110

alcohol, drug addiction or donated blood within 72 days prior to study. Test

liposomal formulations of DFS (LP-05 and LP-11) & reference formulation

(Phlogin-SR®) and (LP-17 and LP-23) liposomal formulations of DFP &

reference drug brand (Caflam) were administered with 240 ml of water on

different occasions. The study was conducted as 18×3 single dose, randomized,

open, and complete crossover Latin-Square design. Volunteers were fasted

overnight before and 3h after drug administration. Blood sample (5 mL) were

collected before drug administration and 0.5, 1.0, 1.5, 2.0, 2.5, 3.0, 4.0, 6.0,

8.0, 12.0, 16.0, 20.0 and 24.0 hr post drug administration through an indwelling

cannula into pre-heparinised glass vials. After drug administration standard

breakfast, lunch and dinner were provided at 3, 9 and 16 hr of post dose. The

blood samples were immediately centrifuged; plasma was separated and stored

at -50 ± 5°C until analysis. After a washout period of 7 days, the study was

repeated for twice in the same manner to complete the crossover Latin-square

design. The developed HPLC methods were utilized for analysis of plasma

samples obtained at various time intervals as described in chapters 3 and 4 for

DFS and DFP, respectively.

6.2.11.2 Pharmacokinetic analysis

The plasma concentration profile obtained was put into computer software on

Microsoft Excel® to plot graphs and Kinetica® 4.4version for determination of

pharmacokinetic parameters. The maximum diclofenac concentration Cmax

and the corresponding peak time Tmax were determined by the inspection of

individual drug plasma concentration–time curves. The elimination rate

Page 141: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

111

constant Kel was obtained from the least-square fitted terminal log-linear

portion of plasma concentration–time profile. The elimination half-life T½ was

calculated as 0.693/Kel. The area under the curve to the last measurable

concentration (AUC0–t) was calculated by the linear trapezoidal rule. The area

under the curve extrapolated to infinity (AUC0–∞) was calculated as AUC0–t

+Ct/Kel where Ct is the last measurable concentration. All other parameters

were calculated according to the non-compartmental model approach.

6.2.11.3 Statistical analysis

For the purpose of bioequivalence analysis AUC0–t, AUC0–∞ and Cmax were

considered as primary variables. Bioequivalence of two selected liposomal

formulations of DFS prepared with PSL and PEL were compared to the

sustained release formulation available in market, Phlogin SR®. Similarly, two

liposomal formulations of DFP formulated with two types of lipids (PSL and

PEL) were compared with available drug product in market, Caflam®, was

assessed by means of an analysis of variance (ANOVA) for Latin Square

crossover design and calculating 95% confidence interval of the ratio of

test/reference using log transformed data. The formulations were considered

bioequivalent when the difference between two compared parameters was

found statistically insignificant (p > 0.05) and confidence interval for these

parameters fell within acceptable range of bioequivalence rule -20+25% (80-

125%).

Page 142: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

112

6.3 Results and Discussion

6.3.1 Fabrication, Morphology, Size and Size Distribution of Liposomes

Liposomes are vesicular structures prepared by different phospholipids derived

from natural or synthetic materials. The amount of drug loaded into liposomes

and the size of liposomes play important roles in the pharmacokinetic and

pharmacodynamics parameters of drug. Hence, accurate and rapid

measurement of the size of liposomes is essential for novel and effective drug

delivery systems. The oral liposomal formulations of diclofenac (both salts;

sodium and potassium) were prepared successfully using the MEE method

(Table 6.1). The liposomes were clear, spherical and homogeneous with light-

yellowish color. The size (Table 6.2) of all liposomal formulations (LP-01 to

LP-24) prepared with both analgesic salts employing purified soya lecithin and

purified egg lecithin were found in the range of 1.01 ± 0.011 to 1.94 ± 0.009

µm (mean ± SEM) at three repeated values (n=3). The graphical representation

of the size of selected liposomal formulations (LP-01, LP-07, LP-13 and LP-

19) measured by Zetasizer (Malvern, UK) are presented in Figure 6.1-6.4. The

size of liposomes has been shown to be an important factor in the efficient

delivery of therapeutic agent to the target site (Nagayasu et al., 1999; Legros,

2007). The size of liposomes also have effects on the pharmacokinetics of

liposomes and liposome-encapsulated drugs by (i) the enhancement in

circulation amount and residence time of liposomes in the blood, (ii) the

accumulation of liposomes at the target site and (iii) in vivo drug release from

liposomes. In addition, the size also affects therapeutic activity of a liposomal

Page 143: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

113

preparation. It is also worth mentioning that liposome size is also important in

the design of a more effective liposomal preparation (Nagayasu et al., 1999;

Dan Marino 2002; Grant et al., 2003). The liposome size was 1.01 ± 0.011,

1.12 ± 0.009 and 1.54 ± 0.008 µm (mean ± SEM) for LP-01, LP-02 and LP-03,

respectively. The formulations (LP-01 to LP-03) are of diclofenac sodium and

soya lecithin. The increased particle size was observed by the increased

concentration of phospholipids. The studies of size measurement of LP-04 to

LP-06 demonstrated the values from 1.65 ± 0.009 to 1.72 ± 0.009 µm (Mean ±

SEM). The size measurement values of batches of diclofenac sodium and egg

lecithin showed values in range of 1.15 ± 0.009 to 1.18 ± 0.007 (mean ± EM)

for LP-07 to LP-09 while it was 1.72 ± 0.008 to 1.78 ± 0.009 (mean ± SEM)

for LP-10 to LP-12. The values of size for diclofenac potassium liposomal

formulations were found as 1.24 ± 0.011 to 1.29 ± 0.008 (mean ± SEM) for LP-

13 to LP-15 while the values for batches LP-16 to LP-18 were found in the

rage of 1.73 ± 0.007 to 1.79 ± 0.009 (mean ± SEM). Similarly, the batches of

diclofenac potassium with egg lecithin (LP-19 to LP-21) and (LP-22 to LP-24)

were found in the range of (1. 45 ± 0.008 to 1.51 ± 0.006) to (1.71 ± 0.005 to

1.78 ± 0.0060), respectively. It was observed in the data that vesicular size of

liposomal formulations with egg lecithins were found comparatively larger

than formations of soya lecithins. The polydispersity index is the measurement

of homogeneity of dispersion, ranging from 0.0 (homogeneous) to 1.0

(heterogeneous) for the size distribution of liposomal formulations ranged from

0.59 ± 0.009 to 0.74 ± 0.007 (Taylor et al., 2007; Montanari et al., 2007). The

Page 144: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

114

values of polydispersity index were presented in Table 6.2. The range PDI was

found from 0.182 ± 0.003 to 0.224 ± 0.002 (mean ± SEM, at n=3). The pattern

of size distribution was specified in the intensity of the light scattering and the

polydispersity index was defined as the log normal distribution width of the

particle diameter.

Further, the morphology of selected formulations of DFS liposomes prepared

with PSL and PEL were examined by scanning electron microscopy and SEM

graph of LP-01 ha been shown Figure 6.5. The result indicated that the

liposomes appeared to be round in shape and smooth on the surface. Moreover,

the particle size distribution was well-proportioned in good agreement with the

result measured by DLS.

The particle size analysis by Zetasizer accessed in the present research revealed

following information.

1. The particle size was found in the range of 1-2 µm showed that

liposomes were of intermediate size. The size range depicted that

liposomes were of consisted of multilayer as size of liposomes is

indicator of lamellarity (number of lipid layers); Small sizes are

unilamellar where as lager are multilamellar liposomes. Our results were

found in conformance with MLVs (Amarnath and Uma., 1997).

2. The phospholipids used were neutral in nature with high transition

temperature showing stability of the formulations. The incorporation of

hydrophilic polymer provided additional benefits of stability and low

opsonization thus provide long circulation of drug in body. This was

Page 145: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

115

evident by longer half life of liposomal formulation calculated in this

study. All was based upon the size of liposomes. The results were found

in line with the studies available in literature that intermediate sized

liposomes are retained within the blood compartment and can circulate

for long periods while the larger liposomes are readily taken up by

Kupffer cells and mononuclear phagocyte system (Amarnath and Uma.,

1997; Lopes et al., 2007; Maria et al., 2006).

3. Another important fact is that the release of drug from liposomes also

depends upon size; the release profile of liposomal formulation in the

present study was sustained as the small liposomes release faster than

large liposomes due to no of lipid layers. This was in conformance with

the study of Masahiro et al., (2007) described that small liposomes have

large curvature and the packing between the lipids in the membranes is

looser and the release is faster compared to the larger liposomes. The

additional information was gathered that increase in lamellarity

enhances the entrapment of drugs was also found in accordance with the

present studies.

4. The size of liposomes also affect the desired pharmacokinetic behavior

and performance of liposomal drug carriers in terms of drug targeting

and/or controlled release as well as for achieving meaningful results

during model membrane studies (Roopa and Kusum, 2009; El-Samaligy

et al., 2006).

Page 146: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

116

Table 6.1: Composition of liposomal formulations of DFS and DFP (n=24)

PSL= purified soya lecithin, PEL= purified egg lecithin, PEG= polyethylene glycol, DFS= diclofenac sodium DFP= diclofenac potassium

Batch Formulation

code

Components and their concentration (mg)

Phospholipids Cholesterol Diclofenac PEG

Ratio 75 : 25

Purified Soya Lecithin and Diclofenac sodium

DFS-I

LP-01 PSL, 75 25 DFS, 100

LP-02 PSL, 150 50 DFS, 100

LP-03 PSL, 225 75 DFS, 100

DFS-II

LP-04 PSL, 150 50 DFS, 100 15

LP-05 PSL, 150 50 DFS, 100 30

LP-06 PSL, 150 50 DFS, 100 45

Purified Egg Lecithin and Diclofenac sodium

DFS-III

LP-07 PEL, 75 25 DFS, 100

LP-08 PEL, 150 50 DFS, 100

LP-09 PEL, 225 75 DFS, 100

DFS-IV

LP-10 PEL, 150 50 DFS, 100 15

LP-11 PEL, 150 50 DFS, 100 30

LP-12 PEL, 150 50 DFS, 100 45 Purified Soya Lecithin and Diclofenac potassium

DFP-I

LP-13 PSL, 75 25 DFP, 100

LP-14 PSL, 150 50 DFP, 100

LP-15 PSL, 225 75 DFP, 100

DFP-II

LP-16 PSL, 150 50 DFP, 100 15

LP-17 PSL, 150 50 DFP, 100 30

LP-18 PSL, 150 50 DFP, 100 45 Purified Egg Lecithin and Diclofenac potassium

DFP-III

LP-19 PEL, 75 25 DFP, 100

LP-20 PEL, 150 50 DFP, 100

LP-21 PEL, 225 75 DFP, 100

DFP-IV

LP-22 PEL, 150 50 DFP, 100 15

LP-23 PEL, 150 50 DFP, 100 30

LP-24 PEL, 150 50 DFP, 100 45

Page 147: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

117

Table 6.2: Characterization of (n= 24) Liposomal Formulations of DFS and DFP (Mean ±SEM)

Sr.

No

Formulation

code Particle size

Polydispersity

index

Zeta

potential (ζ) EE (%, w/w)

1 LP-01 1.01 ± 0.011 0.198 ± 0.004 48.11 ± 0.06 82.14 ± 0.018

2 LP-02 1.12 ± 0.009 0.202 ± 0.003 47.24 ± 0.05 86.82 ± 0.017

3 LP-03 1.54 ± 0.008 0.204 ± 0.005 48.30 ± 0.04 88.94 ± 0.012

4 LP-04 1.68 ± 0.009 0.184 ± 0.005 53.05 ± 0.02 86.68 ± 0.013

5 LP-05 1.65 ± 0.010 0.182 ± 0.003 54.15 ± 0.04 86.64 ± 0.015

6 LP-06 1.72 ± 0.009 0.202 ± 0.001 53.18 ± 0.03 86.59 ±0.016

7 LP-07 1.15 ± 0.008 0.224 ± 0.002 50.95 ± 0.04 83.15 ± 0.017

8 LP-08 1.17 ± 0.009 0.201 ± 0.004 48.98 ± 0.02 87.24 ± 0.018

9 LP-09 1.18 ± 0.007 0.198 ± 0.002 49.01 ± 0.03 89.14 ± 0.015

10 LP-10 1.74 ± 0.008 0.185 ± 0.003 54.21 ± 0.04 87.34 ± 0.012

11 LP-11 1.72 ± 0.009 0.201 ± 0.004 55.24 ± 0.03 87.29 ± 0.018

12 LP-12 1.78 ± 0.007 0.204 ± 0.005 53.28 ± 0.04 87.32 ± 0.017

13 LP-13 1.24 ± 0.011 0.203 ± 0.005 46.93 ± 0.02 82.27 ± 0.016

14 LP-14 1.29 ± 0.012 0.206 ± 0.003 47.28 ± 0.03 86.55 ± 0.012

15 LP-15 1.27 ± 0.008 0.200 ± 0.004 48.35 ± 0.03 89.35 ± 0.017

16 LP-16 1.83 ± 0.008 0.212 ± 0.007 54.95 ± 0.05 86.69 ± 0.015

17 LP-17 1.89 ± 0.007 0.186 ± 0.006 53.84 ± 0.04 86.37 ± 0.013

18 LP-18 1.94 ± 0.009 0.184 ± 0.005 54.88 ± 0.03 86.44 ± 0.014

19 LP-19 1.45 ± 0.008 0.201 ± 0.003 47.11 ± 0.04 84.33 ± 0.019

20 LP-20 1.48 ± 0.007 0.202 ± 0.004 48.44 ± 0.02 87.19 ± 0.015

21 LP-21 1.51 ± 0.006 0.204 ± 0.003 49.76 ± 0.03 89.21 ± 0.013

22 LP-22 1.71 ± 0.005 0.202 ± 0.005 54.68 ± 0.04 87.22 ± 0.018

23 LP-23 1.75 ± 0.004 0.204 ± 0.004 55.74 ± 0.02 87.27 ± 0.014

24 LP-24 1.78 ± 0.006 0.201 ± 0.004 55.91 ± 0.03 87.29 ± 0.012

Page 148: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

118

Figure 6.1: Size of the liposomal formulation of DFS with PSL (LP-01) measured by

Zetasizer (n=3)

Figure 6.2: Size of the liposomal formulation of DFS with PEL (LP-07) measured by

Zetasizer (n=3)

Page 149: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

119

Figure 6.3: Size of the liposomal formulation of DFP with PSL (LP-13) measured by

Zetasizer (n=3)

Figure 6.4: Size of the liposomal formulation of DFP with PEL (LP-19) measured by

Zetasizer (n=3)

Page 150: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

(a) Figure 6.5: Scanning electron microscopic images of selected liposomal

formulation of DFS (LP 6.3.2 Measurement of Zeta potential ( The electric charge on surface particles when com

electric potential located at the shear plane is called the zeta potential and

denoted by symbol (ζ). The zeta

and aggregation processes. The plane of shear is an imaginary surface

unscrambling the layer of liquid vaulted to the solid surface in motion. Zeta

potential is a useful indicator of particle surface charge, wh

predict and control the stability of colloidal suspensions or emulsions. The

greater the zeta potential the more likely the suspension is to be stable because

(b)

Scanning electron microscopic images of selected liposomal formulation of DFS (LP-01)

Zeta potential (ζ)

The electric charge on surface particles when come in contact with a liquid,

electric potential located at the shear plane is called the zeta potential and

). The zeta potential is a mean to understand dispersion

and aggregation processes. The plane of shear is an imaginary surface

unscrambling the layer of liquid vaulted to the solid surface in motion. Zeta

potential is a useful indicator of particle surface charge, which can be used to

predict and control the stability of colloidal suspensions or emulsions. The

greater the zeta potential the more likely the suspension is to be stable because

120

Scanning electron microscopic images of selected liposomal

e in contact with a liquid,

electric potential located at the shear plane is called the zeta potential and

potential is a mean to understand dispersion

and aggregation processes. The plane of shear is an imaginary surface

unscrambling the layer of liquid vaulted to the solid surface in motion. Zeta

ich can be used to

predict and control the stability of colloidal suspensions or emulsions. The

greater the zeta potential the more likely the suspension is to be stable because

Page 151: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

121

the charged particles repel one another and thus overcome the natural tendency

to aggregate (Schmidt, 1986).

In the present study, the zeta potential (ζ) is measured using Zetasizer

(Malvern, UK) at 25°C using dynamic light scattering principle. Measurements

were made on the neat liposome samples, where the samples were ten folds

diluted with PBS. The values (means of 3 repeat measurements with standard

error of mean) were summarized in Table 6.2. The values of (ζ) were found in

the range of 47.24-48.30 for LP-01 to LP-03 (purified soya lecithin and

diclofenac sodium) and values increased with LP-04 to LP-06 added with PEG

as 53.05 - 54.15. The values were found in range of 48.98 - 50.95 for LP-07 to

LP-09 (purified egg lecithin and diclofenac sodium) and values increased with

LP-10 to LP-12 added with PEG as 53.28 - 55.24. The values indicate that

liposomes preparations were stable as the values of zeta potential were in the

range depicting stability of liposomes. Similarly, the zeta potential values of

LP-13 to LP-15 were found in range of 46.93 - 48.35 (diclofenac potassium

and purified soya lecithin) and LP-16-LP-18 (diclofenac potassium and

purified soya lecithin with PEG) in the range of 53.84 - 55.95. The values of

zeta potential for LP-19 to LP-21 were found in range of 47.11 - 49.76

(diclofenac potassium and purified egg lecithin) and LP-21 to LP-24

(diclofenac potassium and purified egg lecithin with PEG) in the range of 54.68

- 55.91. The values of the zeta potential were shown graphically in Figure 6.5-

6.8. It is noted in the formulations that (ζ) of purified egg lecithin formulation

was slightly greater than soya lecithin formulations indicating the enhanced

Page 152: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

122

stability of these formulation and subsequently release profile characteristics

(Sriram and Rhodes, 1995). The values of zeta potential calculated in present

study were in accordance with studies of Knecht et al. (2008) and Mehnert et

al. (2001) stated that measurement of the zeta potential allows predictions to be

made about the storage stability of liposomal formulations. It is currently

admitted that zeta potentials in the [30] to [60] mV are required for full

electrostatic stabilization. The potentials between [5] and |15|mV are in the

region of limited flocculation; and between [5] and [3] mV of maximum

flocculation. Thus, particle aggregation is less likely to occur for charged

particles (high zeta potential) due to electric repulsion (Ney, 1973; Heiati et al.

1998; Delgado et al. 2005).

Figure 6.6: Zeta potential of the liposomal formulation of DFS with PSL (LP-01)

measured by Zetasizer (n=3)

Page 153: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

123

Figure 6.7: Zeta potential of the liposomal formulation of DFS with PEL (LP-07)

measured by Zetasizer (n=3)

Figure 6.8: Zeta potential of the liposomal formulation of DFP with PSL (LP-07)

measured by Zetasizer (n=3)

Page 154: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

Figure 6.9: Zeta potential of the liposomal fmeasured by Zetasizer (n=3)

: Zeta potential of the liposomal formulation of DFP with PEL (LPmeasured by Zetasizer (n=3)

124

ormulation of DFP with PEL (LP-19)

Page 155: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

125

6.3.3. Differential scanning calorimetric (DSC) evaluation of liposomes

formulation The calorimetric analysis was carried out to find out the variation in properties

of DFS or DFP, PSL or PEL and liposomal formulations and effect of DFS or

DFP on liposomal formulation. For this purpose, sample is subjected to a

controlled temperature program, usually a temperature scan and the heat flow

to or from the sample is monitored in comparison to a reference material.

Figure 6.10 shows representative DSC thermograms of DFS with PSL and PEL

in selected liposomal formulations LP-01 to LP-03 and LP-07 to LP-09 and

DFP with and PEL in liposomal formulations LP-04 to LP-06 and LP-10 to LP-

12. DFS showed single endothermic peak at about 236°C illustrating purity of

drug (Irene et al., 2007). PSL showed three peaks; one minor and small peak at

90°C, one peak at 180°C and small broader peak at 285°C. This profile by PSL

showed significant melting, hence graph demonstrated the span of melting at

180°C representing that PSL remains stable over extreme of temperature

ranges. PEL showed more intense peak at 110°C and peaks at 173°C and a

broader peak at 294°C. The difference between thermograms of two lipids is

their composition; as PEL contains more contents of phosphatidyl choline. The

formulations (LP-01 to LP-03 and LP-07 to LP-09) also demonstrated peaks at

170-185°C and 230-245°C indicating interaction of DFS with PSL and PEL but

the content showed no change in nature of formulation compared to individual

entities. A previous study conducted by Lucaina et al., (2006) showed similar

interaction of diclofenac with soya phosphatidyl choline in liposomes

preparations. The results are in agreement with the present study. It is also

Page 156: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

126

demonstrated that entrapment of diclofenac in aqueous phase interacted with

lipid phase of PEL causing slight shift of endothermic peaks towards lower

temperatures and enhanced intensity of peaks. The results are also in line with

literature studies of Maria et al., (2005); and Vringer and Ronde, (1995). It is

also stated in reference that when diclofenac interacts with phospholipids

during preparation, interactions results in decreasing decomposition

temperature and intensity of the peaks in comparison to drug decomposition

which is found similar in present investigation that lowering of decomposition

temperatures are observed in all formulations. It is also noted that the decrease

in temperature was less than previous studies establishing our finding that we

have incorporated increasing ratio of phospholipids which has compensated

this lower decrease in decomposition temperatures (Bucci et. al., 2000).

Another reason is the incorporation of diclofenac in aqueous phase of the

liposomes. A study of incorporation of methotrexate demonstrating no change

in position of drug was seen when drug was incorporated in aqueous phase of

liposomes (Michele et al., 2004).

Page 157: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

127

Figure 6.10: DSC of selected formulations of DFS liposomes

Page 158: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

128

6.3.4 X-ray Diffraction (XRD) studies

The XRD studied were performed for DFS, PSL and DFS-PSL liposomal

formulations as representative samples of whole study. The amorphous nature

of PSL was confirmed from x-ray diffraction analysis. The crystalline nature of

drug was clearly demonstrated by characteristic XRD pattern of DFS

demonstrated peaks appearing at 6.08, 7.85, 16.20, 17.51 and 21.84 at 2θ

values attributable to the crystalline drug. These values and pattern was found

in line with previous studies of Pignatello et al., (2002). The pattern of peaks of

PSL with incorporated cholesterol demonstrated amorphous behavior as shown

in Figure 6.11. The peaks of PSL appeared at 20 and 26.5 at 2θ values. The

liposomal formulations confirmed the existence of drug probably in the

crystalline form. LP-01 and LP-07 were investigated for XRD studies. The

peaks were observed at 6, 16.85 and 25.20. The appearance of diffractogran

showed similar behavior of drug with incorporated PSL depicting little

interaction of drug with phospholipid portion of formulations but a small shift

and merging of peaks of drug and PSL indicating weak interaction. The drugs

retained its crystallinity in liposomes demonstrating that drugs remain in

separate phase than PSL; this is in conformance as described in preparation

steps that drug is dispersed in aqueous phase. The XRD showed the stability of

drug in liposomal as it has not interacted with PSL and maintained the integrity

of formed liposomes of DFS. The DSC and XRD studies collectively indicated

that a portion of drug incorporated existed as solid solution in the liposomal

formulation where the drug showed its identity in system. The presence of drug

Page 159: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

129

as a solid in aqueous portion in liposomes could be responsible for the

sustained release from liposomes responsible to prolong drug release from

liposomes.

Figure 6.11: XRD of some selected formulations of DFS liposomes

Page 160: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

130

6.3.5 FT-IR studies

Significant text presented on application of FTIR to characterize the phase

behavior of phospholipids. The present study is undertaken to characterize

formulated liposomes of diclofenac in different concentrations of PSL and

PEL. The FT-IR spectra of diclofenac sodium demonstrated distinguishing

peaks at 3381.57 cm-1 due to NH stretching of secondary amine, 1572.66 cm-1

owing to -C = O stretching of carboxyl ion and at 745.35 cm-1 because of C–Cl

stretching (Pignatello et al., 2002). The FTIR spectra of soya lecithin displayed

characteristic peaks at 3600–3000 cm-1 due to OH stretching vibration, 3000–

2800 cm-1 for –CH2 –C=O cm-1 stretching and OH bending vibration between

1770–1500 cm-1. The groups C-O-C, P-O-C and P=O showed stretching

vibration between 1200-1050 (Whittinghill et al., 2000). Individually, both the

drugs and soya lecithin spectra showed sharp peaks demonstrating strong

vibrations but when liposomal formulation is studied in FTIR, the spectra

showed weak vibrations demonstrating reduced interfacial tension (Figure

6.12). In the IR spectra of solid dispersions the peak due to drug carboxyl

group was shifted to 1577.49 cm-1 whereas the signal resulting from the

possibility of any chemical interaction between drug and Soya lecithin. A weak

electrostatic interaction between carboxyl group of drug and ammonium group

of polymers during co-dissolution and solvent evaporation could not be ruled

out. A weak electrostatic interaction between carboxyl group of NSAIDs and

the ammonium group of the polymer has been reported in the literature.

Page 161: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

131

The characteristic infrared bands of drug were markedly affected in the blend;

some bands completely disappeared. These changes can provide an evidence of

interaction between lipophilic drug and phospholipids. The interaction may be

with the non-polar tails (Immordino et al., 2002). Such interaction may enhance

drug permeability which contributes to higher drug bioavailability (Yanyu et

al., 2006).

Figure 6.12: FT-IR of selected formulations of DFS liposomes

Page 162: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

132

6.3.6 Encapsulation efficiency

Entrapment efficiency abbreviated as EE, also described as percent entrapment

yield (PEY). It is defined as “the ratio of the quantity of active entity remaining

with in the liposomal layers to the initial amount of drug added to the system”.

Centrifugation method was adopted for the determination of concentration of

entrapped diclofenac as described by Shivhare et al., (2009) and El-Samaligy et

al., (2006).

The entrapment of diclofenac in liposomal formulations was investigated in

optimization study (Chapter 5). However, entrapment studies were also

conducted in this study. The value of entrapment for LP-01 was 82.14 ± 0.018

% (mean ± SEM) which rise to 86.94 ± 0.012 % for LP-03 (Table 6.2). LP-01

contains an amount of phospholipid content (75:25; Soya lecithin to

cholesterol) and diclofenac sodium (1:1) ratio. LP-02 and LP-03 contained

increased ratio of lipid to drug (2:1) and (3:1), respectively. The relative

increase in the ratio of lipid content increase entrapment efficiency brought

slight increased entrapment of drug. Entrapment of diclofenac in liposomal

formulations (LP-07 to LP-09) prepared with egg lecithins showed slightly

higher entrapment than liposomal formulation of soya lecithins (LP-01 to LP-

03). The formulation pattern for diclofenac potassium liposomes from LP-13

to LP-24 was same as for LP-01 to LP-12 diclofenac sodium (Table 6.1). It was

observed that entrapment of diclofenac potassium liposomal formulations

showed relatively higher entrapment values than liposomal formulations of

diclofenac sodium. High fatty acid contents of egg-lecithin are responsible for

Page 163: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

133

high entrapment efficiency and controlling the diffusion of drug from vesicles.

Another important justification for higher entrapment of diclofenac salts with

egg lecithins was due to higher hydration effects than soya lecithins. This has

increase the tendency for entrapping diclofenac salts in liposomal formulations

(Maha et al., 2008). The entrapment efficiency obtained in the present study

(82.14-89.35%) was much higher than liposomal formulations of diclofenac

sodium (63%) for ocular administration by Sun et al., (2006) and diclofenac

entrapment was also higher compared to the sustained release formulation of

diclofenac sodium with alginates, a study presented by Manjunatha et al.,

(2007). The entrapment of diclofenac sodium in microsphere formulation was

found as > 80% by Nande et al., (2006) and 70-80% in controlled release

formulation study by Ehad et al., (1997) was less than present study. The

higher values justified with higher efficiency of method of preparation which is

developed in the present study and selection of components liposomes and

solvent system. The results obtained in present study are found in line with the

previous studies of Marija et al., (2008); Liang et al., (2004) and Cui-Tao et al.,

(2010).

6.3.7 Stability studies of liposomal formulations

Four formulations of liposome (LP-02, LP-05, LP-08 and LP-11) of DFS

prepared with PSL and PEL were incorporated in stability testing studies.

Similarly, four formulation of DFP (LP-14, LP-17, LP-20 and LP-23) prepared

with PSL and PEL, respectively.

Page 164: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

134

The storage conditions were 30ºC, 40ºC and 40ºC75%RH for the duration of

six months. The data points for sampling and analysis were selected as 0, 1, 2,

3 and 6 months for determining percentage content and release profiles.

Properly labeled containers wrapped in aluminum foil were stored in stability

chambers (Sanyu, Japan). The required amount of liposomes was taken off

from stability chambers at time of sampling at specific period and analyzed

using specific procedures of analysis for DFS and DFP liposomes (Chapter No.

3. and 4). The release profile of each formulation was determined by the same

procedure as discussed in in-vitro studies. The data of all formulations are

presented in Table 6.13-6.52.

On physical inspection, all liposomal formulations were found to be reasonably

stable in terms of phase separation, drug precipitation, aggregation and vesicle

disruption over the entire studied storage period. Changes in the color,

formation of sediment, and increase in turbidity of supernatant were not seen in

all formulations at all temperature conditions. Microscopic characteristics,

shape and uniformity were maintained to the considerable extent for all

liposomal formulations.

On chemical analysis, it was observed that there was a proportional decrease in

percent drug assay found at 0, 1, 2, 3 and 6 months of the stability study

program. Where as, minor difference in T60 release profile was observed in

similar manner. The percent drug assay for all liposomal formulations was

decreased in proportional manner from 99.82 to 98.11 at 30ºC, 99.88 to 98.22

at 40ºC and 99.82 to 98.15 at 40ºC/75% RH at start to six months of stability

Page 165: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

135

study. Approximately 1 % decrease in drug assay at 30ºC assuring stability of

all liposomal formulation of DFS and DFP for at least two years. While percent

assay decreased approximately to 2 % at 40ºC and 40ºC/75% RH at six months.

Similarly, the drug release profile (T60) was decreased from 6.16 to 5.88 and

6.85 to 6.64 hrs for LP-02 and LP-05, respectively. It indicates that release

profile was increased with the storage to the initial value up to six months at 30

ºC. Similar behavior of release profile was observed with all formulations of

liposomes. At higher temperatures (40ºC and 40ºC/75% RH), liposomal

formulations showed a gradual increased release but it was not significantly

higher from the release profile of formulations stored at 30ºC indicating the

consistent release of DFS and DFP from liposomes. Similar phenomenon was

with release profile by Elorza et al., (1997) who described the consistent and

gradual increase in release profile from liposomes. Another important similar

finding of retaining 96% of the incorporated drug at higher temperature has

been demonstrated by Roopa and Kusum, (2009). Another study presented by

Patel and Misra, (1999) described the stability of Clofazimine liposomes,

determined by four different techniques (Sonicated, Vortexed, Gel filtration

and Ultracentrifugation) were found to have same active drug entrapment

(61.02 ± 0.3, 60.09 ± 0.9, 61.02 ± 0.3 and 61.97 ± 0.5) after storage at different

temperatures indicating stability of entrapment efficiencies.

Page 166: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

136

Table 6.3: Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at initial time of stability studies

Formulation Percent Drug Content Time for 60 % release (T60)

30ºC 40ºC 40ºC

75%RH 30ºC 40ºC

40ºC 75%RH

LP-02 99.95 99.88 99.82 6.16 6.08 5.98

LP-05 99.88 99.76 99.70 6.85 6.74 6.55

LP-08 99.78 99.72 99.68 7.60 7.52 7.38

LP-11 99.82 99.75 99.72 7.98 7.73 7.63

LP-14 99.83 99.78 99.73 6.78 6.52 6.38

LP-17 99.85 99.76 99.72 7.82 7.46 7.33

LP-20 99.78 99.67 99.64 6.80 6.62 6.36

LP-23 99.80 99.72 99.68 8.45 8.19 7.88

Table 6.4: Percent drug content and T60 release profile of liposomal

formulations of DFS and DFP at one month period of stability studies

Formulation Percent Drug Content Time for 60 % release (T60)

30ºC 40ºC 40ºC

75%RH 30ºC 40ºC

40ºC 75%RH

LP-02 99.82 99.65 99.40 6.11 6.01 5.92

LP-05 99.80 99.58 99.38 6.78 6.71 6.48

LP-08 99.72 99.52 99.31 7.56 7.48 7.32

LP-11 99.71 99.58 99.33 7.91 7.67 7.55

LP-14 99.70 99.55 99.36 6.72 6.47 6.32

LP-17 99.72 99.56 99.35 7.76 7.42 7.30

LP-20 99.73 99.53 99.35 6.75 6.56 6.32

LP-23 99.68 99.50 99.34 8.41 8.14 7.83

Page 167: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

137

Table 6.5: Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at two month period of

stability studies

Formulation Percent Drug Content Time for 60 % release (T60)

30ºC 40ºC 40ºC

75%RH 30ºC 40ºC

40ºC 75%RH

LP-02 99.32 99.16 98.95 6.07 5.99 5.88

LP-05 99.35 99.14 98.92 6.73 6.67 6.44

LP-08 99.41 99.12 98.95 7.52 7.43 7.28

LP-11 99.36 99.11 98.96 7.87 7.61 7.51

LP-14 99.30 99.16 98.96 6.67 6.42 6.28

LP-17 99.26 99.12 98.98 7.73 7.37 7.28

LP-20 99.22 99.11 98.95 6.72 6.52 6.29

LP-23 99.25 99.18 98.95 8.37 8.11 7.77

Table 6.6: Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at three month period of stability studies

Formulation Percent Drug Content Time for 60 % release (T60)

30ºC 40ºC 40ºC

75%RH 30ºC 40ºC

40ºC 75%RH

LP-02 99.12 98.70 98.60 5.98 5.94 5.85

LP-05 99.16 99.50 98.64 6.68 6.60 6.41

LP-08 99.12 99.42 98.62 7.50 7.39 7.25

LP-11 99.10 99.40 98.61 7.85 7.58 7.47

LP-14 99.12 99.38 98.68 6.63 6.37 6.25

LP-17 99.10 99.34 98.66 7.66 7.32 7.24

LP-20 99.16 99.44 98.63 6.65 6.44 6.23

LP-23 99.12 99.52 98.66 8.32 8.07 7.72

Page 168: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

138

Table 6.7: Percent drug content and T60 release profile of liposomal formulations of DFS and DFP at six month period of stability studies

Formulation Percent Drug Content Time for 60 % release (T60)

30ºC 40ºC 40ºC

75%RH 30ºC 40ºC

40ºC 75%RH

LP-02 98.96 98.50 98.15 5.88 5.85 5.81

LP-05 98.99 98.55 98.23 6.63 6.53 6.37

LP-08 98.91 98.65 98.68 7.48 7.34 7.20

LP-11 98.95 98.80 97.99 7.82 7.53 7.41

LP-14 98.92 98.78 98.42 6.61 6.29 6.21

LP-17 98.98 98.74 98.74 7.63 7.27 7.20

LP-20 98.93 98.74 98.63 6.62 6.40 6.18

LP-23 98.97 98.22 97.98 8.30 8.02 7.66

Page 169: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

139

6.4 In-vitro Studies

In-vitro release profile of formulations can be better estimated though

optimized dissolution process. Dissolution is a sensitive, reproducible and

relatively straightforward test that can be used to effectively monitor batch-to-

batch variability and ensure bioequivalence, once bioavailability has been

established. Secondly, under some circumstances, in vitro dissolution may be

used as a surrogate indicator of in-vivo dissolution profile and therefore, as a

tool to predict the extent of absorption where dissolution (solubility of the

drugs) is rate limiting (Christopher and Charman, 2001; Horter, 2001).

USP apparatus-II (paddle method) was used for determination of percent drug

release from the liposomal preparations in 900 ml of dissolution media. DFS

and DFP Liposomes equal to 100 mg were placed in dissolution media.

Dispersion method was applied for the determination of diclofenac release.

Dissolution medium consists of phosphate buffered saline (PBS) maintained at

pH 7.4, 37 ºC temperature and stirring speed of 100 rpm were applied. The

phosphate buffered saline was prepared by dissolving 8 g of NaCl, 0.19 g

KH2PO4 and 2.38 g Na2HPO4 in one liter of distilled water. Percent drug

release from liposomal formulations were calculated by taking peak response

(HPLC) of diclofenac dissolved in strength and adjusted at same volume as

incorporated in single dose of drug (Charman et al., 1997; Rajesh et al., 2001).

The analysis procedure was adopted same as described in Chapter 3 and 4 for

DFS and DFP.

Page 170: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

140

6.4.1 Profile of Liposomal Formulations of Diclofenac

6.4.1.1 Diclofenac liposomes prepared with Purified Soya Lecithin

First batch of liposomal formulations (DFS-I) comprised three types of

formulations. These formulations were prepared with lecithin which was

separated from purified soya lecithin, out of which three formulations were

selected as representative to show the drug release profile. Release profile of

loaded drug into vesicles was determined by plotting cumulative percentage of

drug release against time (hr).

The three formulations LP-01, LP-02 and LP-03 contained purified soya

lecithin and cholesterol in molar ratio of 75:25 with increasing proportion 100,

200 and 300 mg where as concentration of drug was kept constant. The

formulations were designed to estimate the effect of lecithin on the release of

incorporated drug. These liposomal formulations LP-01, LP-02, and LP-03

released 98.56 % of drug over 10 hrs, 99.98 % after 12 hrs and 98.68 % after

16 hrs, respectively. Release kinetic models were applied to determine the

mechanism, release pattern from these formulations clearly showed zero order

0.9740, 0.9850 and 0.9449 and followed Higuchi’s model 0.9955, 0.9835 and

0.9940 for LP-01, LP-02, and LP-03, respectively. The results indicate

diffusional mechanism for release of drug from liposomes (Figure 6.13 – 6.19).

Similarly, a batch of diclofenac potassium (DFP-I) was prepared having same

composition to compare the release characteristics with liposomes of

diclofenac sodium. These liposomal formulations of diclofenac potassium LP-

13, LP-14, and LP-15 released 98.63 % of drug after 08 hrs, 99.94 % after 12

Page 171: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

141

hrs and 98.07 % up to 16 hrs, respectively and indicated the release profile of

zero order 0.9549, 0.9217 and 0.9415 and Higuchi’s model 0.9908, 0.9904 and

0.9868, LP-13, LP-14, and LP-15, respectively. Data of all applied kinetic

models for liposomal formulations presented graphically in Figures 6.33-6.37.

6.4.1.2 Liposomes prepared with Soya Lecithin and PEG

Second batch of liposomal formulations (DFS-II) was contained three types of

formulations. These formulations were also prepared with purified lecithin and

total three formulations LP-04, LP-05, and LP-06 were selected as

representative to show the drug release profile. But in this case, all three

formulations LP-04, LP-05, and LP-06 contained lecithin and cholesterol

(150:50) and drug in constant amount (100 mg). Polyethylene glycol (PEG)

was incorporated with increasing proportions 15, 30 and 45 mg in LP-04, LP-

05, and LP-06 formulations. This batch was designed to show the effect of

increasing concentration PEG on the release profile of incorporated drug.

Release profile of loaded drug into vesicles was determined by plotting the

cumulative percentage of drug release against time (hr).

The liposomal formulations LP-04, LP-05, and LP-06 released 99.72 % of drug

after 16 hrs, 99.22 % after 20 hrs and 97.48 % up to 20 hrs, respectively. All in-

vitro kinetics models were applied to determine release pattern from these

formulations which clearly indicated that release profile followed zero order

0.9326, 0.9384 and 0.9330 and Higuchi’s model 0.9917, 0.9793 and 0.9937 for

LP-04, LP-05, and LP-06, respectively. The results mention diffusional

mechanism for release of drug from loaded liposomes (Figures 6.18-6.22).

Page 172: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

142

A batch of diclofenac potassium (DFP-II) of same composition was prepared.

Liposomal formulations of diclofenac potassium LP-16, LP-17, and LP-18

released 99.82 % of drug after 16 hrs, 98.73 % after 20 hrs and 98.89 % up to

20 hrs, respectively and indicated the release profile of zero order 0.9483,

0.9229 and 0.9591 and Higuchi’s model 0.9968, 0.9904 and 0.9934 for LP-16,

LP-17, and LP-18, respectively. The data of kinetic models have been

described graphically in Figure 6.38-6.42.

6.4.1.3 Diclofenac liposomes prepared with Purified Egg Lecithin

Third batch of diclofenac sodium liposomal formulations (DFS-III) was

prepared with lecithin which was separated from egg and representative three

formulations LP-07, LP-08, and LP-09 were subjected to illustrate the release

profile of drug. Composition was comprised of drug and cholesterol with same

amount but egg lecithin was added in varying ratios (100, 200 and 300 mg) in

selected three formulations. These liposomal formulations LP-07, LP-08, and

LP-09 released 98.12 % of drug after 12 hrs, 99.77 % after 16 hrs and 98.69 %

up to 16 hrs, respectively. All in-vitro kinetics models (Figures 6.23-6.27) were

applied to determine release pattern from these formulations which clearly

indicated that release profile followed zero order 0.9204, 0.9747 and 0.9596

and Higuchi’s model 0.9813, 0.9963 and 0.9947 for LP-07, LP-08, and LP-09,

respectively.

A comparative batch of diclofenac potassium liposomes (DFP-III) of same

composition was prepared and these formulations LP-19, LP-20, and LP-21

released 99.97 % of drug after 12 hrs, 99.77 % after 16 hrs and 98.69 % up to

Page 173: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

143

16 hrs, respectively and indicated the release profile of zero order 0.9313,

0.9450 and 0.9609 and Higuchi’s model 0.9854, 0.9951 and 0.9957 for LP-19,

LP-20, and LP-21, respectively. The data of kinetics models have been plotted

graphically in Figure 6.6.43-6.47.

6.4.1.4 Liposomes prepared with Egg Lecithin and PEG

Fourth batch of diclofenac sodium liposomal formulations (DFS-IV) was

prepared with egg lecithin and representative three formulations LP-10, LP-11,

and LP-12 were comprised of lecithin, drug and cholesterol with constant

amount but PEG was added in varying ratios (10, 20 and 30 mg). These

formulations LP-10, LP-11, and LP-12 released 99.87 %, 99.22 % after 20 hrs

and 99.98 % up to 24 hrs, respectively. Release profile (Figures 6.28-6.32)

from these formulations also demonstrated the zero order 0.9046, 0.9807 and

0.9135 and Higuchi’s model 0.9837, 0.9847 and 0.9881 for LP-10, LP-11, and

LP-12, respectively.

Another batch of diclofenac potassium liposomes (DFP-III) was prepared and

these formulations LP-22, LP-23, and LP-24 released 99.78 % of drug after 16

hrs, 98.22 % after 20 hrs and 99.83 % up to 24 hrs, respectively and indicated

the release profile of zero order 0.9424, 0.9412 and 0.9412 and Higuchi’s

model 0.9932, 0.9938 and 0.9952 for LP-22, LP-23, and LP-24, respectively.

The data of kinetics models for these formulations have been shown for visual

inspection Figure 6.48-6.52. The results of each applied release kinetic model

showing individual parameters have been presented in Table 6.8.

Page 174: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

144

Zero Order Model

0

20

40

60

80

100

120

0 3 6 9 12 15 18

Time (hr)C

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-01 LP-02 LP-03

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 3 6 9 12 15 18

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-01 LP-02 LP-03

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-01 LP-02 LP-03

Figure 6.13: Zero order release model of oral liposomal formulations of diclofenac sodium.

Figure 6.14: First order release model of oral liposomal formulations of diclofenac sodium.

Figure 6.15: Higuchi release model of oral liposomal formulations of diclofenac sodium.

Page 175: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

145

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 3 6 9 12 15 18

Time (hr)

Cub

ic roo

t of cu

mul

ativ

e

perc

ent o

f dr

ug rel

ease

d

LP-01 LP-02 LP-03

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-01 LP-02 LP-03

Zero Order Model

0

20

40

60

80

100

120

0 4 8 12 16 20

Time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-04 LP-05 LP-06

Figure 6.16: Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

Figure 6.17: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

Figure 6.18: Zero order release model of oral liposomal formulations of diclofenac sodium.

Page 176: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

146

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 3 6 9 12 15 18

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-04 LP-05 LP-06

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ulat

ive pe

rcen

t of d

rug

rele

ased

LP-04 LP-05 LP-06

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 3 6 9 12 15 18

Time (hr)

Cub

ic r

oot

of

cum

ula

tive

per

cent

of

drug

rel

ease

d

LP-04 LP-05 LP-06

Figure 6.19: First order release model of oral liposomal formulations of diclofenac sodium.

Figure 6.20: Higuchi release model of oral liposomal formulations of diclofenac sodium.

Figure6.21: Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

Page 177: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

147

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-04 LP-05 LP-06

Zero Order Model

0

20

40

60

80

100

120

0 4 8 12 16 20

Time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-07 LP-08 LP-09

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 3 6 9 12 15 18

Time (hr)

Log

of cu

mul

ativ

e pe

rcen

t

of d

rug

rele

ased

LP-07 LP-08 LP-09

Figure 6.22: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

Figure 6.23: Zero order release model of oral liposomal formulations of diclofenac sodium.

Figure 6.24: First order release model of oral liposomal formulations of diclofenac sodium.

Page 178: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

148

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-07 LP-08 LP-09

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 3 6 9 12 15 18

Time (hr)

Cub

ic roo

t of cu

mul

ativ

e pe

rcen

t

of d

rug

rele

ased

LP-07 LP-08 LP-09

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t

of d

rug

rele

ased

LP-07 LP-08 LP-09

Figure 6.25: Higuchi release model of oral liposomal formulations of diclofenac sodium.

Figure 6.26: Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

Figure 6.27: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

Page 179: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

149

Zero Order Model

0

20

40

60

80

100

120

0 4 8 12 16 20

Time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-10 LP-11 LP-12

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 3 6 9 12 15 18 21 24

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t of dr

ug

rele

ased

LP-10 LP-11 LP-12

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-10 LP-11 LP-12

Figure 6.28: Zero order release model of oral liposomal formulations of diclofenac sodium.

Figure 6.29: First order release model of oral liposomal formulations of diclofenac sodium.

Figure 6.30: Higuchi release model of oral liposomal formulations of diclofenac sodium.

Page 180: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

150

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 3 6 9 12 15 18 21 24

Time (hr)

Cub

ic roo

t of cu

mul

ativ

e pe

rcen

t

of d

rug

releas

ed

LP-10 LP-11 LP-12

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0 1.2

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t

of d

rug

rele

ased

LP-10 LP-11 LP-12

Zero Order Model

0

20

40

60

80

100

120

0 3 6 9 12 15 18

Time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-13 LP-14 LP-15

Figure 6.31: Hixon Crowell release model of oral liposomal formulations of diclofenac sodium.

Figure 6.32: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

Figure 6.33: Zero order release model of oral liposomal formulations of diclofenac potassium.

Page 181: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

151

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 3 6 9 12 15 18

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t

of d

rug

rele

ased

LP-13 LP-14 LP-15

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ulat

ive

per

cent o

f dr

ug

rele

ased

LP-13 LP-14 LP-15

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 3 6 9 12 15 18

Time (hr)

Cub

ic ro

ot o

f cum

ulat

ive pe

rcen

t

of d

rug

rele

ased

LP-13 LP-14 LP-15

Figure 6.34: First order release model of oral liposomal formulations of diclofenac potassium.

Figure 6.35: Higuchi release model of oral liposomal formulations of diclofenac potassium.

Figure 6.36: Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

Page 182: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

152

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.7 0.8

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t

of d

rug

rele

ased

LP-13 LP-14 LP-15

Zero Order Model

0

20

40

60

80

100

120

0 5 10 15 20

Time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-16 LP-17 LP-18

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 5 10 15 20

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-16 LP-17 LP-18

Figure 6.37: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac potassium

Figure 6.38: Zero order release model of oral liposomal formulations of diclofenac potassium.

Figure 6.39: First order release model of oral liposomal formulations of diclofenac potassium.

Page 183: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

153

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ula

tive

per

cent

of dr

ug

rele

ased

LP-16 LP-17 LP-18

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 4 8 12 16 20

Time (hr)

Cub

ic roo

t of

cum

ulat

ive

perc

ent

of dr

ug rel

ease

d

LP-16 LP-17 LP-18

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0

Log of time (hr)

Log

of cu

mul

ativ

e pe

rcen

t of dr

ug

rele

ased

LP-16 LP-17 LP-18

Figure 6.40: Higuchi release model of oral liposomal formulations of diclofenac potassium.

Figure 6.41: Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

Figure 6.42: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

Page 184: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

154

Zero Order Model

0

20

40

60

80

100

120

0 3 6 9 12 15 18

Time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-19 LP-20 LP-21

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 3 6 9 12 15 18

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-19 LP-20 LP-21

Higuchi Model

0

20

40

60

80

100

120

0 1 2 3 4 5

Squar root of time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-19 LP-20 LP-21

Figure 6.43: Zero order release model of oral liposomal formulations of diclofenac potassium.

Figure 6.44: First order release model of oral liposomal formulations of diclofenac potassium.

Figure 6.45: Higuchi release model of oral liposomal formulations of diclofenac potassium.

Page 185: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

155

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 3 6 9 12 15 18

Time (hr)

Cub

ic ro

ot o

f cum

ulat

ive pe

rcen

t

of d

rug

rele

ased

LP-19 LP-20 LP-21

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-19 LP-20 LP-21

Zero Order Model

0

20

40

60

80

100

120

0 4 8 12 16 20 24

Time (hr)

Cum

ulat

ive pe

rcen

t of d

rug

rele

ased

LP-22 LP-23 LP-24

Figure 6.46: Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

Figure 6.47: Pappas release model (first 60% drug release) of oral liposomal formulations of diclofenac sodium.

Figure 6.48: Zero order release model of oral liposomal formulations of diclofenac potassium.

Page 186: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

156

First Order Model

0.0

0.5

1.0

1.5

2.0

2.5

0 4 8 12 16 20 24

Time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-22 LP-23 LP-24

Higuchi Model

0

20

40

60

80

100

120

0 1 1 2 2 3 3 4 4

Squar root of time (hr)

Cum

ulat

ive

perc

ent o

f dru

g

rele

ased

LP-22 LP-23 LP-24

Hixon Crowell Model

0.0

1.0

2.0

3.0

4.0

5.0

0 4 8 12 16 20 24

Time (hr)

Cub

ic ro

ot o

f cum

ulat

ive

perc

ent

of d

rug

rele

ased

LP-22 LP-23 LP-24

Figure 6.49: First order release model of oral liposomal formulations of diclofenac potassium.

Figure 6.50: Higuchi release model of oral liposomal formulations of diclofenac potassium.

Figure 6.51: Hixon Crowell release model of oral liposomal formulations of diclofenac potassium.

Page 187: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

157

6.4.2 Discussion of In-vitro Studies

Liposomes are considered to improve solubility, lipidization and sustained

actions of drug substances through increase in effective lumenal drug solubility

by two mechanisms, primarily by stimulating higher secretion of bile salts,

phospholipid and cholesterol, which leads to the formation of enteric diverse

micelles resulting in higher solubilization capacity of the GI tract. The later

phenomenon is the intercalation of administered lipids (liposomal preparations)

into these bile salts structures leading to swelling of micellar structures and

providing a mean for sustained delivery of drugs and also form solubilized

phase from which enhanced absorption may occur (Humberstone and Charman,

1997). Keeping in view of these characteristics of enhancing solubility and

sustaining properties of liposomes, the current study was aimed to prepare

different batches of liposomal formulation of BCS class-II drugs, diclofenac

Peppas Model

0.0

0.4

0.8

1.2

1.6

2.0

0.0 0.2 0.4 0.6 0.8 1.0

Log of time (hr)

Log

of c

umul

ativ

e pe

rcen

t of d

rug

rele

ased

LP-22 LP-23 LP-24

Figure 6.52: Pappas release model (First 60% drug release) of oral liposomal formulations of diclofenac sodium.

Page 188: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

158

sodium and diclofenac potassium having low solubility (Chuasuwan et al.,

2009). The phospholipid components were composed of purified soya lecithin,

purified egg lecithin and cholesterol in the fixed ratio of 75:25 (lecithin to

cholesterol) and prolonged release of drug was seen with an incorporation of

polyethylene glycol (PEG) in liposomal formulations.

Fatty acid contents of phospholipids are principle determinants of their

properties. Differences in fatty acid part can modify the characteristics of

phospholipids molecules because of variations in chain length as well as degree

of unsaturation. Egg lecithin has more fatty acid part than soya lecithin.

Release profile of liposomal formulations prepared from soya lecithin is

different from liposomes containing lecithin extracted from egg (Sriram

Vemuri and Rhodes, 1995). In this study egg-lecithin retarded release of drug

(both diclofenac sodium and diclofenac potassium) more as compared to soy-

lecithin as shown in Figures 6.23, 6.23, 6.33 and 6.43. High fatty acid contents

of egg-lecithin are responsible for high entrapment efficiency and retarding

capacity by controlling the diffusion of drug from vesicles. The release kinetics

of the incorporated drugs was found influenced with the concentration of

phospholipids content in liposomal formulations. It was observed that overall

capacity of increased phospholipids to retard the release rate with increasing

lipid concentration (Luciana et al., 2006). It was also discussed in a literature

study that purified egg lecithins were found to have significant higher

hydration effects than soya lecithins, this effect brought higher release profile

of diclofenac salts from liposomal formulations (Maha et al., 2008).

Page 189: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

159

An optimized amount of cholesterol was added in all formulations because

cholesterol improves the fluidity and stability of bilayer membrane in

liposomes but higher concentration of cholesterol decrease the percentage of

phosphatidylcholine. This decrease in phophadidylcholine can decrease the

entrapment efficiency and release of drug from lipid vesicles (Shivhare et al.,

2009; Sriram and Rhodes, 1995). Moreover, it is also reported by Cocera et

al., (2003) that incremental inclusion of cholesterol in phospholipid vesicles

produce proportional decrease of release rates and also brought changes in the

bilayer packing and ordering. Therefore, an increased amount of optimized

combination of phospholipid component in all formulations to explain the

effect of nature of lecithin and PEG concentrations on release profile of drug in

dissolution media.

The incorporation of PEG retard the release from drug product and have shown

to increases the residence time in blood circulation by decreasing the

recognition of immune system and form protective layer on surface of

liposomes which lead to reduction in clearance of liposomes (Tamer Shehata et

al., 2008; Valerie Centis and Patrick Vermette, 2008; Sriram and Rhodes,

1995).

In the present study, in-vitro release profile revealed that increase in

concentrations of PEG retarded the release of drug from liposomes. The

stabilizing effect of PEG to liposome surface reinforced to retard diffusion of

drug that ultimately has decreased the release which is demonstrated in Figure

6.22, 6.32, 6.42 and 6.52. So liposomal formulations (LP-06, LP-12, LP-18 and

Page 190: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

160

LP-24) which were prepared from egg-lecithin and contained higher amount of

PEG showed more delay/retard in release of drug. Diclofenac potassium

liposomal formulations (LP-13 to LP-24) showed higher cumulative percentage

of drug release as compared to liposomes of diclofenac sodium which was

mainly due to higher solubility characteristics of diclofenac potassium

illustrated in Figure 6.33, 6.38, 6.43 and 6.48. The higher release from the

liposomal formulations of diclofenac potassium in comparison with diclofenac

sodium revealed an other important information that diclofenac potassium has

comparative higher solubility than diclofenac sodium due to presence of two

water molecules (dihydrate form) than diclofenac sodium having four water

molecules (tetra hydrate form) and potassium salt has more water loving

properties than sodium salt of diclofenac (Fini et al., 2001).

In-vitro kinetic release model were applied on all formulations which evidently

showed the zero order (mean r2 value of DFS 0.9329 ± 0.008 and DFP 0.9416

± 0.004) (mean ± SEM) and Higuchi release pattern (mean r2 value of DFS

0.9855 ± 0.002 and DFP 0.9441 ± 0.001). This release profile revealed that

diffusion of drug is a principle process for release of drug from vesicles.

Sustained release and target delivery are good with liposomes as it is the

potential of liposomal formulations to encapsulate a drug of hydrophilic and

lipophilic nature which offers several advantages over conventional drug

delivery systems. However, water soluble drugs tend to leak out during

preparation as well as during storage on the shelf. The leakage problem is one

of the limiting factors in commercial development of liposome products.

Page 191: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

161

Nevertheless, limited success was attained to improve the long-term retention

of encapsulated solutes by freeze-drying of lyophilization (Sriram and Rhodes,

1995). In the present study, all formulations of diclofenac sodium and

diclofenac potassium were freeze dried as final step of their preparation.

Therefore, consistent, stable and sustained release liposomal formulations were

developed.

Page 192: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

162

Table 6.8: In-vitro release parameters of liposomal formulations of diclofenac sodium and diclofenac potassium.

Formulations Release Parameters

Zero Order First Order Higuchi Korsmeyer-

Peppas

Hixson-

Crowell Diclofenac

Sodium r2

k0

(h-1)

r2

k1

(h-1) r

2

kH

(h-1) r

2

n

value r

2

kHC

(h-1/3 )

DFS-I

LP-01 0.9740 8.832 0.8286 0.0834 0.9955 35.235 0.9227 0.9629 0.8925 0.2233

LP-02 0.9850 7.617 0.8422 0.0711 0.9835 32.385 0.8140 0.7424 0.9157 0.1908

LP-03 0.9449 6.599 0.7148 0.0685 0.9940 31.901 0.9424 1.147 0.8164 0.1718

DFS-II

LP-04 0.9326 6.566 0.6968 0.0654 0.9917 31.907 0.9369 1.235 0.9784 0.1662

LP-05 0.9384 6.482 0.6452 0.0535 0.9793 29.136 0.9323 1.193 0.7521 0.1353

LP-06 0.9330 5.082 0.6588 0.0553 0.9937 27.20 0.9194 1.204 0.7811 0.1356

DFS-III

LP-07 0.9204 7.581 0.7084 0.0732 0.9813 21.865 0.9536 1.306 0.7930 0.1931

LP-08 0.9596 6.188 0.7725 0.0616 0.9947 21.918 0.9609 0.9453 0.8554 0.1607

LP-09 0.9747 6.151 0.7686 0.0673 0.9963 23.769 0.9584 1.046 0.8643 0.1689

DFS-IV

LP-10 0.9046 5.094 0.6730 0.0509 0.9837 18.749 0.9502 1.204 0.7721 0.1316

LP-11 0.9135 5.154 0.6722 0.055 0.9847 20.815 0.9397 1.172 0.7775 0.1382

LP-12 0.9807 4.204 0.7553 0.0463 0.9881 20.478 0.9456 0.9293 0.8698 0.1151

Diclofenac

Potassium

Zero Order First Order Higuchi Korsmeyer-

Peppas

Hixson-

Crowell

r2

k0

(h-1)

r2

k1

(h-1) r

2

kH

(h-1) r

2

n

value r

2

kHC

(h-1/3 )

DFP-I

LP-13 0.9549 12.694 0.7222 0.1406 0.9908 46.56 0.8146 1.470 0.7394 0.2168

LP-14 0.9415 7.961 0.6666 0.0879 0.9904 34.743 0.7990 1.249 0.7828 0.2159

LP-15 0.9217 6.351 0.6571 0.0702 0.9868 30.968 0.8341 1.265 0.7758 0.1734

DFP-II

LP-16 0.9483 6.256 0.6898 0.0716 0.9968 30.228 0.8560 1.248 0.7521 0.1373

LP-17 0.9229 5.033 0.5873 0.0614 0.9904 27.038 0.7888 1.299 0.7390 0.1440

LP-18 0.9591 4.80 0.6052 0.0631 0.9934 25.334 0.7948 1.192 0.7679 0.1438

DFP-III

LP-19 0.9313 8.178 0.6648 0.0923 0.9854 35.789 0.8573 1.629 0.7786 0.2258

LP-20 0.9450 6.274 0.6508 0.0739 0.9951 30.340 0.8244 1.348 0.7829 0.1774

LP-21 0.9609 6.351 0.6363 0.0829 0.9957 30.465 0.8060 1.351 0.7912 0.1894

DFP-IV

LP-22 0.9424 6.315 0.6383 0.0781 0.9932 30.550 0.8266 1.460 0.7737 0.1832

LP-23 0.9412 5.081 0.6032 0.065 0.9938 27.078 0.8003 1.354 0.7609 0.1494

LP-24 0.9412 4.379 0.5819 0.0583 0.9952 25.355 0.8120 1.388 0.6619 0.1412

Page 193: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

6.5 Similarity factor determination

6.5.1 Comparison of release profile of Liposomal formulation of

sodium and Phlogin-SR

Brand

The aim of current task was to establish the comparison of in

profile of formulations of liposomes with the available sustained release brand

of diclofenac sodium. This was accomplished with the application of similarity

factor (f-2). The values of

dissolution profiles of liposomal formulations considered as test and Phlogin

SR® (Brooks Pharmaceuticals

this objective, Moore and Flanner, (1996) devised model for calculating the

values of f-2 test. The equation can be utilized to calculate similarities in

dissolution profiles of products. The equation described minimum value of 50

as the ratio of comparison of release profile to

borderline value for similarity o

considered as the conversion factor which takes into account variability

between samples at each time point

Where f-2 represents the similarity factor, n

points (observations), wt is optional weight, R

from reference formulation, and T

Similarity factor determination (f-2)

Comparison of release profile of Liposomal formulation of

SR® Brooks Pharmaceuticals (Pakistan) as Reference

current task was to establish the comparison of in-vi

profile of formulations of liposomes with the available sustained release brand

of diclofenac sodium. This was accomplished with the application of similarity

). The values of f-2 were used to calculate the variability in

profiles of liposomal formulations considered as test and Phlogin

ceuticals, Pakistan) as reference formulation. To achieve

this objective, Moore and Flanner, (1996) devised model for calculating the

test. The equation can be utilized to calculate similarities in

products. The equation described minimum value of 50

as the ratio of comparison of release profile to f-2 similarity factor.

borderline value for similarity or dissimilarity of the formulations and

considered as the conversion factor which takes into account variability

between samples at each time point (Mukesh et al., 2005; Saranadasa

represents the similarity factor, n is the value of number of data

points (observations), wt is optional weight, Rt is percentage drug dissolved

from reference formulation, and Tt is percent drug dissolved from test

163

Comparison of release profile of Liposomal formulation of diclofenac

Brooks Pharmaceuticals (Pakistan) as Reference

vitro release

profile of formulations of liposomes with the available sustained release brand

of diclofenac sodium. This was accomplished with the application of similarity

were used to calculate the variability in

profiles of liposomal formulations considered as test and Phlogin-

, Pakistan) as reference formulation. To achieve

this objective, Moore and Flanner, (1996) devised model for calculating the

test. The equation can be utilized to calculate similarities in

products. The equation described minimum value of 50

similarity factor. 50 is the

r dissimilarity of the formulations and f-2 was

considered as the conversion factor which takes into account variability

Saranadasa, 2005).

is the value of number of data

is percentage drug dissolved

is percent drug dissolved from test

(i)

Page 194: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

164

formulation. Conservatively, a test formulation of drug product is considered

comparable to the reference drug product if the value of f-2 of the two release

profiles lies in between (50 and 100). The lower acceptable value (i.e., 50)

communicates to 10% average absolute difference between a reference product

and a test product at each time point. In reality, a dissolution study will show

different values of difference between R and T at each time point. Among all

the formulation of liposomes, the release profile of all the formulations of

diclofenac sodium were compared with that of reference product (Phlogin-

SR®), a sustained release brand available in market. Table 6.9 summarizes all

the results of f-2 test. The values of LP-05 and LP-13 produced relative

similar profiles of dissolution. So, these formulations were selected for in-vivo

evaluation. Therefore, from the data of similarity factor (f-2 value), it is

revealed that LP-5 and LP-11 prepared liposomal formulations of diclofenac

sodium with purified soya lecithin (PSL) and purified egg lecithin (PEL)

produced the similarity with marketed SR formulation.

Page 195: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

165

Table 6.9: f2-values of liposomal formulations calculated from drug release profile and compared with reference drug product Phlogin-SR®

Comparison of release profile F2-value

Reference Vs LP-01 32.16

Reference Vs LP-02 39.32

Reference Vs LP-03 43.24

Reference Vs LP-04 45.48

Reference Vs LP-05 61.70

Reference Vs LP-06 44.88

Reference Vs LP-07 46.22

Reference Vs LP-08 45.44

Reference Vs LP-09 46.76

Reference Vs LP-10 46.66

Reference Vs LP-11 62.24

Reference Vs LP-12 47.66

Page 196: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

166

6.6 In-vivo Evaluation of Liposomal Formulations of Diclofenac

This study was conducted to evaluate the bioavailability and pharmacokinetics

of oral formulations liposomes of diclofenac salts (DFS/DFP) in eighteen

healthy subjects. The Latin-square crossover and single blind design was

adopted and the selection of subjects was randomized. The volunteers were

fasted for at least 10 hours prior to administration of dose. Based on the in-vitro

dissolution and physico-chemical characterization studies, liposomes of DFS

and DFP formulated with PSL and PEL phospholipids with fixed proportion of

cholesterol (75:25) in the presence of PEG (10%) were chosen for in vivo

studies. The mentioned formulations were characterized as stable, small

particle size and low particle size distribution. These formulations of liposomes

also showed good entrapment efficiency (82.65%). Scanning electron

microscopy of these formulation samples revealed that liposomes were

spherical, homogeneous in size and free from aggregates. Liposomes were

filled in capsules shells just before administration. The selected formulations

of DFS liposomes were compared in-vivo with the available sustained release

marketed brand of Diclofenac sodium (Phlogin-SR®, 100 mg) while DFP

liposomes were compared with the available marketed brand of Diclofenac

potassium (Caflam, 50 mg).

Liposomal formulations of DFS LP-05 and LP-11 formulated with purified

soya lecithin (PSL) and purified egg lecithin (PEL) were administered in an

oral dose of 100 mg where as liposomal formulations of DFP (LP-17 and LP-

23) formulated with purified soya lecithin (PSL) and purified egg lecithin

Page 197: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

167

(PEL) were administered in the oral dose of 50 mg and blood samples were

obtained for 24 hours. Quantitative analysis of diclofenac salts (DFS/DFP) in

plasma was established by a validated reversed-phase high-performance liquid

chromatographic (HPLC) assay. The plasma concentrations were tabulated and

plotted in different figures for visual inspection. Model independent

pharmacokinetic parameters were estimated using area and moment analysis.

The mean maximum concentration (Cmax), time for maximum concentration

(Tmax), area under the plasma concentration–time curve (AUC), area under the

first moment curve (AUMC), the mean residence time (MRT), half life (T1/2),

elimination rate constant (Ke), volume of distribution (VD) and clearance (CLT)

were calculated for the selected liposomal formulations.

6.6.1 Plasma concentration profile of liposomal formulations of DFS

6.6.1.1 Liposomes prepared with Soya Lecithin (LP-05) & Egg Lecithin (LP-11)

The plasma level time profiles of liposomal formulations of diclofenac sodium

(LP-05, LP-11 and Phlogin-SR®) are shown in Figure 6.53 to 6.70. The sum of

plasma concentrations of eighteen subjects are given in Table 6.10 to 6.12 and

graphically shown in Figure 6.71 to 6.73. The mean ± SEM plasma

concentrations of LP-05, LP-11 and Phlogin-SR® are given in Table 6.13 and

compared statistically in Figure 6.74.

Page 198: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

168

Figure 6.53: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogin-SR® administered in the oral dose of 100 mg in Subject No. 1.

Figure 6.54: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 2.

Figure 6.55: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 3.

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DFP-PSL DFP-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

Page 199: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

169

Figure 6.56: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 4.

Figure 6.57: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP- 11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 5.

Figure 6.58: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 6.

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGEN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

Page 200: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

170

Figure 6.59: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 7.

Figure 6.60: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 8.

Figure 6.61: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP- 11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 9.

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-SL DSF-EL PHLOGIN-SR

Page 201: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

171

Figure 6.62: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No.10.

Figure 63: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No.11.

Figure 6.64: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No.12.

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

Page 202: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

172

Figure 6.65: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No.13.

Figure 6.66: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No.14.

Figure 6.67: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No.15.

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

Page 203: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

173

Figure 6.68: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in the oral dose of 100 mg in Subject No.16.

Figure 6.69: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in the oral dose of 100 mg in Subject No. 17.

Figure 6.70: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05 and

LP-11) and Phlogin-SR® administered in oral dose of 100 mg in Subject No. 18.

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

DSF-PSL DSF-PEL PHLOGIN-SR

Page 204: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

174

Table 6.10: Plasma level time profile of oral formulation of Liposomes of DFS (LP-05) administered in the oral dose of 100 mg in healthy subjects (n=18)

Time (Hrs) S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13

0.5 0.318 0.290 0.273 0.309 0.278 0.311 0.253 0.345 0.376 0.315 0.304 0.287 0.281

1 0.670 0.569 0.476 0.599 0.581 0.660 0.514 0.537 0.318 0.585 0.617 0.520 0.585

1.5 1.552 1.468 1.257 1.183 1.216 1.229 1.229 1.238 1.330 1.282 1.524 1.265 1.225

2 2.098 1.959 1.956 1.954 1.886 2.054 1.974 1.954 2.116 1.959 2.043 1.976 1.971

2.5 2.369 2.268 2.198 2.367 2.340 2.363 2.318 2.437 2.517 2.294 2.353 2.348 2.340

3 2.451 2.432 2.468 2.461 2.556 2.517 2.370 2.532 2.588 2.498 2.443 2.447 2.481

4 2.474 2.266 2.213 2.093 2.098 2.017 2.250 2.318 2.264 2.052 2.365 2.142 2.122

6 2.196 2.052 1.964 1.952 1.959 1.947 1.953 1.978 1.944 1.780 2.142 1.948 1.953

8 1.805 1.660 1.478 1.626 1.579 1.498 1.506 1.518 1.398 1.543 1.742 1.498 1.528

12 0.852 0.801 1.051 1.008 0.984 0.976 0.893 1.004 0.978 0.988 0.881 0.955 0.951

16 0.502 0.489 0.409 0.358 0.359 0.322 0.481 0.290 0.463 0.316 0.530 0.360 0.387

20 0.139 0.135 0.109 0.149 0.130 0.155 0.125 0.116 0.129 0.141 0.141 0.125 0.137

24 0.092 0.087 0.075 0.041 0.037 0.053 0.077 0.073 0.093 0.046 0.090 0.067 0.056

Page 205: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

175

Table 6.11: Plasma level time profile of oral formulation of Liposomes of DFS (LP-11) administered in the oral dose of 100 mg in healthy subjects (n=18)

Time (Hrs) S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13

0.5 0.345 0.348 0.336 0.316 0.323 0.327 0.327 0.391 0.368 0.365 0.399 0.365 0.343

1 0.648 0.677 0.652 0.443 0.500 0.634 0.648 0.699 0.648 0.569 0.336 0.665 0.659

1.5 1.600 1.686 1.586 1.493 1.403 1.253 1.288 1.302 1.403 1.311 1.408 1.345 1.624

2 2.402 2.502 2.455 2.226 2.001 2.070 2.090 2.175 2.123 2.070 2.241 2.088 2.384

2.5 2.817 2.723 2.645 2.593 2.446 2.507 2.507 2.502 2.576 2.581 2.665 2.507 2.659

3 2.709 2.579 2.508 2.530 2.613 2.606 2.786 2.665 2.642 2.682 2.741 2.646 2.599

4 2.510 2.236 2.447 2.308 2.219 2.217 2.326 2.136 2.403 2.455 2.398 2.173 2.398

6 2.267 2.031 2.090 2.028 2.004 2.067 2.093 2.062 2.071 2.094 2.059 1.885 2.129

8 1.832 1.290 1.605 1.529 1.669 1.722 1.829 1.587 1.697 1.608 1.481 1.634 1.576

12 0.990 0.992 0.757 0.956 1.046 1.068 0.993 1.034 1.063 1.063 1.036 1.046 0.913

16 0.657 0.367 0.401 0.348 0.531 0.379 0.336 0.341 0.773 0.307 0.490 0.334 0.475

20 0.150 0.118 0.132 0.126 0.227 0.158 0.138 0.164 0.323 0.122 0.137 0.149 0.134

24 0.091 0.059 0.081 0.086 0.099 0.044 0.039 0.056 0.088 0.077 0.099 0.049 0.077

Page 206: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

176

Table 6.12: Plasma level time profile of sustained release marketed brand of DFS (Phlogin-SR®) administered in the oral dose of 100 mg in healthy subjects (n=18)

Time

(Hrs) S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13

0.5 0.278 0.279 0.282 0.276 0.278 0.273 0.267 0.241 0.254 0.277 0.281 0.283 0.280

1 0.508 0.503 0.499 0.454 0.431 0.546 0.533 0.440 0.471 0.477 0.376 0.468 0.503

1.5 1.363 1.388 1.369 1.338 1.209 1.079 1.097 1.121 0.265 1.169 1.264 1.202 1.373

2 1.868 1.881 1.893 1.992 1.724 1.783 1.801 2.008 1.919 1.861 1.874 1.891 1.881

2.5 2.222 2.257 2.242 2.317 2.107 2.159 2.248 2.356 2.248 2.248 2.282 2.265 2.240

3 2.346 2.202 1.980 2.180 2.251 2.245 2.359 2.321 2.359 2.356 2.353 2.341 2.176

4 2.159 1.979 1.837 1.988 1.912 1.910 1.971 2.102 1.929 2.022 2.066 1.880 1.992

6 1.840 1.507 1.749 1.747 1.726 1.781 1.760 1.757 1.773 1.760 1.760 1.557 1.698

8 1.452 1.211 1.329 1.317 1.438 1.483 1.478 1.215 1.374 1.274 1.272 1.339 1.330

12 0.772 0.815 0.625 0.823 0.901 0.920 0.856 0.826 0.951 0.904 1.044 0.901 0.738

16 0.530 0.384 0.267 0.300 0.458 0.326 0.466 0.430 0.618 0.278 0.672 0.368 0.394

20 0.127 0.106 0.103 0.108 0.196 0.136 0.135 0.140 0.265 0.098 0.198 0.111 0.112

24 0.086 0.052 0.062 0.074 0.085 0.038 0.051 0.048 0.064 0.062 0.069 0.048 0.066

Page 207: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

177

Figure 6.71: Plasma level time profile of oral formulation of Liposomes of

DFS (LP-05) administered in the oral dose of 100 mg in healthy subjects (n=18)

Figure 6.72: Plasma level time profile of oral formulation of Liposomes of

Diclofenac sodium (LP-11) administered in oral dose of 100 mg in healthy subjects (n=18)

Figure 6.73: Plasma level time profile of sustained release marketed brand of

DFS (Phlogin-SR®) administered in the oral dose of 100 mg in healthy subjects (n=18)

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

once

ntr

ation (µg/m

l)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

once

ntr

ation (µg/m

l)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

Page 208: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

178

Table 6.13: Comparison of plasma level time profile of oral formulation of liposomes (LP-05, LP-11) and marketed brand of DFS Phlogin -SR® Brooks Pharma) administered in the oral dose of 100 mg to healthy subjects (n=18)

Time (Hrs) DFP-SL

(Mean ± SEM)

DFP-EL

(Mean ± SEM)

Phlogin-SR®

(Mean ± SEM)

0.5 0.304 ± 0.006 0.347 ± 0.006 0.272 ± 0.003

1 0.557 ± 0.018 0.594 ± 0.022 0.478 ± 0.009

1.5 1.324 ± 0.029 1.412 ± 0.031 1.161 ± 0.059

2 2.003 ± 0.014 2.182 ± 0.036 1.874 ± 0.016

2.5 2.360 ± 0.016 2.563 ± 0.026 2.245 ± 0.013

3 2.484 ± 0.012 2.622 ± 0.021 2.269 ± 0.023

4 2.220 ± 0.028 2.300 ± 0.027 1.977 ± 0.018

6 1.992 ± 0.023 2.050 ± 0.020 1.728 ± 0.018

8 1.577 ± 0.024 1.616 ± 0.030 1.351 ± 0.019

12 0.940 ±0.015 0.996 ± 0.018 0.857 ± 0.021

16 0.415 ±0.017 0.440 ± 0.028 0.423 ± 0.026

20 0.134 ± 0.003 0.164 ± 0.011 0.144 ± 0.010

24 0.070 ± 0.004 0.071 ± 0.004 0.062 ± 0.003

Figure 6.74: Comparison of plasma level time profile of oral formulation of

liposomes (LP-05, LP-11) and marketed brand of DFS (Phlogin-SR® Brooks Pharma) administered in the oral dose of 100 mg to healthy subjects (n=18)

0.0

1.0

2.0

3.0

0 4 8 12 16 20 24Time (Hrs)

Pla

sma C

once

ntr

ati

on (

µg/m

l)

DFS-PSL DFS-PEL Phlogen-SR

Page 209: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

179

6.6.2 Plasma Concentration of Liposomal formulations of DFP

6.6.2.1 Liposomes prepared with PSL (LP-17) and Egg Lecithin (LP-23)

The plasma level time profiles of liposomal formulations of diclofenac

potassium (LP-17, LP-23 and Caflam) were shown in Figure 6.75 to 6.92. The

sum of plasma concentrations of eighteen subjects were given in Table 6.14 to

6.16 and graphically shown in Figure 6.93 to 6.95. The mean ± SEM plasma

concentrations of LP-17, LP-23 and Caflam were given in Table 6.17 and

compared statistically in Figure 6.96.

Page 210: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

180

Figure 6.75: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 1.

Figure 6.76: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 2.

Figure 6.77: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 3.

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

Page 211: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

181

Figure 6.78: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 4.

Figure 6.79: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 5.

Figure 6.80: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 6.

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on

(µg/m

l)

DFP-PSL DFP-P#EL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

Page 212: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

182

Figure 6.81: Plasma level time profile of oral formulation of Liposomes of DFP (LP 17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 7.

Figure 6.82: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP 23) and Caflam® administered in oral dose of 50 mg in Subject No. 8.

Figure 6.83: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 9.

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

Page 213: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

183

Figure 6.84: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 10.

Figure 6.85: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 11.

Figure 6.86: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 12.

0.0

0.5

1.0

1.5

2.0

2.5

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma

Co

ncen

tra

tio

n (

µg

/ml)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (µg/m

l)

DFP-PSL DFP-PEL CAFLAM

Page 214: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

184

Figure 6.87: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP- 23) and Caflam® administered in oral dose of 50 mg in Subject No. 13

Figure 6.88: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 14.

Figure 6.89: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 15.

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

Page 215: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

185

Figure 6.90: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 16.

Figure 6.91: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 17.

Figure 6.92: Plasma level time profile of oral formulation of Liposomes of DFP (LP-17 and

LP-23) and Caflam® administered in oral dose of 50 mg in Subject No. 18.

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

oncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma

Co

ncentr

ati

on (

µg/m

l)

DFP-PSL DFP-PEL CAFLAM

Page 216: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

186

Table 6.14: Plasma level time profile of oral formulation of Liposomes of

Diclofenac potassium (LP-17) administered in the oral dose of 50 mg to healthy subjects (n=18)

Time (Hrs)

S-1

S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-14 S-15

0.5 0.314 0.299 0.287 0.282 0.318 0.287 0.320 0.260 0.355 0.388 0.324 0.313 0.295 0.289 0.322

1 0.630 0.586 0.501 0.491 0.617 0.599 0.679 0.529 0.553 0.327 0.603 0.636 0.536 0.602 0.541

1.5 1.231 1.164 1.074 0.997 0.938 0.965 0.975 0.975 0.982 1.055 1.017 1.209 1.003 0.971 1.051

2 1.465 1.385 1.426 1.383 1.382 1.333 1.452 1.395 1.382 1.496 1.385 1.444 1.397 1.394 1.423

2.5 1.712 1.603 1.752 1.554 1.673 1.655 1.670 1.639 1.723 1.779 1.622 1.663 1.660 1.655 1.676

3 1.729 1.720 1.706 1.745 1.740 1.807 1.779 1.675 1.790 1.830 1.766 1.727 1.730 1.754 1.761

4 1.666 1.602 1.498 1.564 1.480 1.483 1.426 1.591 1.639 1.600 1.451 1.672 1.514 1.500 1.548

6 1.540 1.451 1.362 1.388 1.380 1.385 1.377 1.381 1.398 1.375 1.258 1.514 1.377 1.381 1.381

8 1.244 1.174 0.984 1.045 1.149 1.116 1.059 1.065 1.073 0.989 1.091 1.231 1.059 1.080 1.090

12 0.700 0.566 0.571 0.743 0.713 0.696 0.690 0.631 0.709 0.691 0.698 0.623 0.675 0.672 0.672

16 0.423 0.346 0.221 0.289 0.253 0.254 0.228 0.340 0.205 0.327 0.223 0.374 0.254 0.274 0.291

20 0.106 0.096 0.082 0.077 0.105 0.092 0.109 0.089 0.082 0.091 0.100 0.100 0.088 0.097 0.095

24 0.063 0.062 0.057 0.057 0.029 0.026 0.037 0.055 0.051 0.066 0.033 0.063 0.048 0.039 0.050

Page 217: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

187

Table 6.15: Plasma level time profile of oral formulation of Liposomes of Diclofenac potassium sodium (LP-23) administered in the oral dose of 50 mg to healthy subjects (n=18)

Time (Hrs)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-14

0.5 0.330 0.314 0.301 0.296 0.334 0.301 0.336 0.273 0.373 0.407 0.340 0.329 0.310 0.303

1 0.661 0.616 0.526 0.515 0.648 0.629 0.713 0.555 0.581 0.343 0.633 0.667 0.563 0.632

1.5 1.293 1.223 1.128 1.047 0.985 1.013 1.023 1.023 1.031 1.107 1.067 1.269 1.053 1.020

2 1.538 1.454 1.497 1.452 1.451 1.400 1.525 1.465 1.451 1.571 1.454 1.517 1.467 1.463

2.5 1.798 1.683 1.840 1.632 1.757 1.737 1.754 1.721 1.809 1.868 1.703 1.746 1.743 1.737

3 1.815 1.806 1.792 1.832 1.827 1.897 1.868 1.759 1.880 1.921 1.854 1.813 1.817 1.842

4 1.750 1.682 1.573 1.643 1.554 1.558 1.497 1.670 1.721 1.681 1.523 1.756 1.590 1.575

6 1.616 1.523 1.430 1.458 1.449 1.454 1.446 1.450 1.468 1.443 1.321 1.590 1.446 1.450

8 1.307 1.232 1.033 1.097 1.207 1.172 1.112 1.118 1.127 1.038 1.145 1.293 1.112 1.134

12 0.735 0.594 0.599 0.780 0.748 0.730 0.725 0.663 0.745 0.726 0.733 0.654 0.709 0.706

16 0.444 0.363 0.232 0.303 0.266 0.266 0.239 0.357 0.215 0.343 0.234 0.393 0.267 0.288

20 0.111 0.100 0.087 0.081 0.111 0.097 0.115 0.093 0.086 0.096 0.105 0.105 0.093 0.102

24 0.066 0.065 0.060 0.060 0.031 0.028 0.039 0.057 0.054 0.069 0.034 0.067 0.050 0.041

Page 218: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

188

Table 6.16: Plasma level time profile of oral marketed brand of Diclofenac potassium (CAFLAM® Novartis Pharma) administered in the oral dose of 50 mg to healthy subjects (n=18)

Time (Hrs)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13

0.5 0.254 0.312 0.326 0.304 0.299 0.311 0.315 0.321 0.343 0.398 0.392 0.388 0.297

1 0.521 0.668 0.542 0.588 0.511 0.615 0.599 0.615 0.601 0.611 0.583 0.597 0.577

1.5 1.128 1.123 1.201 1.176 1.316 1.279 1.236 1.389 1.109 1.425 1.180 1.106 1.151

2 1.365 1.356 1.346 1.487 1.367 1.346 1.525 1.505 1.423 1.444 1.408 1.329 1.356

2.5 1.164 1.219 1.234 1.345 1.115 1.218 1.222 1.267 1.458 1.252 1.236 1.220 1.206

3 1.051 1.016 1.065 1.093 1.051 1.029 1.093 1.063 1.080 0.899 1.015 0.916 1.044

4 0.738 0.769 0.659 0.631 0.673 0.591 0.691 0.576 0.648 0.580 0.586 0.573 0.722

6 0.356 0.356 0.342 0.329 0.338 0.398 0.457 0.439 0.428 0.446 0.270 0.356 0.352

8 0.141 0.123 0.116 0.119 0.103 0.125 0.123 0.133 0.110 0.112 0.138 0.131 0.127

12 0.088 0.052 0.036 0.026 0.029 0.034 0.020 0.026 0.021 0.035 0.024 0.030 0.059

Page 219: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

189

Figure 6.93: Plasma level time profile of oral formulation of Liposomes of DFP (LP-11) administered in the oral dose of 50 mg to healthy subjects (Sum, n=18)

Figure 6.94: Plasma level time profile of oral formulation of Liposomes of DFP (LP-23) administered in the oral dose of 50 mg to healthy subjects (Sum, n=18)

Figure 6.95: Plasma level time profile of oral marketed brand of DFP (CAFLAM® Novartis Pharma) (LP-05) administered in the oral dose of 50 mg to healthy subjects (Sum, n=18)

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

0.0

0.5

1.0

1.5

2.0

2.5

0 4 8 12 16 20 24

Time (Hrs)

Pla

sma C

once

ntr

ation (µg/m

l)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

0.0

0.5

1.0

1.5

2.0

0 3 6 9 12

Time (Hrs)

Pla

sma C

on

cen

trati

on

g/m

l)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

Page 220: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

191

Table 6.17: Comparison of plasma level time profile of oral formulation of liposomes (LP-17, LP-23) and marketed brand of Diclofenac potassium(Caflam®- Novartis Pharma) administered in the oral dose of 50 mg to healthy subjects (n=8)

Figure 6.96: Comparison of plasma level time profile of oral formulation of

liposomes (LP-17, LP-23) and marketed brand of DFP (Caflam® Novartis Pharma) administered in the oral dose of 50 mg to healthy

0.0

0.5

1.0

1.5

2.0

0 4 8 12 16 20 24Time (Hrs)

Pla

sma C

oncentr

ati

on (µg/m

l)

PSL PEL Caflam

Time (Hrs) DFP-SL

(Mean ± SEM) DFP-EL

(Mean ± SEM) Caflam®

(Mean ± SEM)

0.5 0.310 ± 0.007 0.326 ± 0.007 0.321 ± 0.038

1 0.567 ± 0.018 0.595 ± 0.019 0.582 ± 0.038

1.5 1.054 ± 0.023 1.106 ± 0.024 1.214 ± 0.092

2 1.414 ± 0.010 1.485 ±0.010 1.399 ± 0.064

2.5 1.673 ± 0.012 1.757 ± 0.013 1.236 ± 0.077

3 1.750 ± 0.009 1.838 ± 0.009 1.035 ± 0.053

4 1.560 ± 0.018 1.638 ± 0.019 0.647 ± 0.055

6 1.405 ± 0.016 1.476 ± 0.017 0.372 ± 0.051

8 1.106 ± 0.018 1.161 ± 0.018 0.121 ± 0.010

12 0.664 ± 0.012 0.697 ± 0.012 0.036 ± 0.017

16 0.293 ± 0.014 0.307 ± 0.012 0.000 ± 0.000

20 0.094 ± 0.002 0.099 ± 0.015 0.000 ± 0.000

24 0.050 ± 0.003 0.053 ± 0.003 0.000 ± 0.000

Page 221: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

192

6.6.3 Pharmacokinetics Parameters of Liposomal Formulations

6.6.3.1 Liposomes of DFS prepared with PSL (LP-05) and PEL (LP-11)

The pharmacokinetics parameters of liposomal formulations of diclofenac

sodium (LP-05, LP-11 and Phlogen-SR®) were shown in Table 6.18, 6.19 and

6.20. The comparison of (mean ± SEM) of Bioavailability and

Pharmacokinetics of oral formulation of liposomes (LP-05, LP-11) and

marketed brand of Diclofenac sodium (Phlogin-SR® Brooks Pharma)

administered in the oral dose of 100 mg in Table 6.21.

6.6.3.2 Liposomes of DFP prepared with PSL (LP-17) and PEL (LP-23)

The pharmacokinetics parameters of liposomal formulations of diclofenac

potassium (LP-17, LP-23 and Caflam®) were shown in Table 6.22, 6.23 and

6.24. The comparison of (mean ± SEM) of Bioavailability and

Pharmacokinetics of oral formulation of liposomes (LP-17, LP-23) and

marketed brand of Diclofenac potassium (Caflam®Novartis Pharma)

administered in the oral dose of 50 mg in Table 6.25.

Page 222: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

191

Table 6.18: Bioavailability and pharmacokinetic parameters of oral formulation of

Liposomes of DFS (LP-05) administered in the oral dose of 100 mg to healthy human subjects (n=18)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

2.474 2.432 2.468 2.461 2.556 2.517 2.37 2.532 2.588 2.498 2.443 2.447 2.481 2.491 2.436 2.499 2.485 2.55

4.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000

0.107 0.105 0.108 0.121 0.124 0.118 0.104 0.116 0.102 0.121 0.104 0.112 0.114 0.110 0.106 0.112 0.106 0.112

6.494 6.615 6.407 5.728 5.568 5.890 6.635 5.962 6.823 5.721 6.647 6.187 6.066 6.298 6.536 6.181 6.542 6.177

24.94 23.39 22.97 23.04 22.77 22.45 22.81 22.84 23.12 22.15 24.62 22.46 22.68 23.01 23.80 22.91 24.29 23.38

25.80 24.22 23.66 23.38 23.06 22.90 23.54 23.47 24.03 22.53 25.49 23.06 23.17 23.64 24.62 23.47 25.13 23.95

185.2 174.5 172.7 171.9 168.2 165.8 171.8 165.5 174.1 163.6 185.5 165.9 168.6 171.6 177.3 170.6 181.0 174.2

214.0 202.3 195.8 182.8 177.7 180.4 196.6 186.0 205.1 175.8 214.5 185.6 184.7 192.6 204.7 189.1 209.0 192.9

8.294 8.355 8.273 7.820 7.705 7.878 8.349 7.926 8.533 7.804 8.415 8.051 7.972 8.147 8.314 8.058 8.318 8.056

0.414 0.433 0.457 0.518 0.540 0.514 0.444 0.495 0.423 0.538 0.409 0.486 0.493 0.466 0.431 0.478 0.422 0.469

3.876 4.130 4.226 4.278 4.336 4.367 4.247 4.261 4.161 4.438 3.924 4.337 4.317 4.230 4.062 4.261 3.979 4.176

Page 223: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

192

Table 6.19: Bioavailability and pharmacokinetic parameters of oral formulation of Liposomes of DFS (LP-11) administered in the oral dose of 100 mg to healthy human subjects (n=18

PARA-

METR S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-

Cmax 2.817 2.723 2.645 2.593 2.613 2.606 2.786 2.665 2.642 2.682 2.741 2.646 2.659 2.583

Tmax 2.500 2.500 2.500 2.500 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 2.500 3.000

Kel 0.106 0.126 0.116 0.110 0.091 0.121 0.129 0.119 0.089 0.116 0.101 0.123 0.114 0.106

Half

life 6.561 5.515 5.970 6.304 7.591 5.743 5.392 5.804 7.810 5.951 6.834 5.644 6.055 6.561

AUC

0-t 27.02 23.13 23.62 23.35 24.97 24.40 24.55 23.79 27.10 24.18 24.49 23.52 24.52 24.24

AUC 0-

inf 27.88 23.60 24.32 24.13 26.06 24.77 24.85 24.26 28.09 24.85 25.46 23.92 25.20 24.96

AUMC

0-t 205.8 165.5 167.7 169.8 196.8 182.1 177.9 175.6 223.3 175.2 184.5 173.3 179.5 182.8

AUMC

0-inf 234.6 180.5 190.4 195.7 234.7 193.8 187.5 190.8 258.2 196.8 217.5 186.1 201.6 206.9

MRT 8.415 7.649 7.831 8.108 9.007 7.827 7.546 7.863 9.193 7.919 8.541 7.780 8.000 8.290

VD 0.379 0.533 0.477 0.456 0.350 0.487 0.517 0.492 0.316 0.469 0.398 0.513 0.454 0.423

CLT 3.587 4.238 4.112 4.144 3.838 4.038 4.024 4.122 3.560 4.025 3.927 4.180 3.969 4.007

Page 224: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

193

Table 6.20: Bioavailability and pharmacokinetic parameters of Phlogin-SR® Brooks

Pharmaceuticals, Pakistan) administered in the oral dose of 100 mg to healthy human subjects (n=18)

PARA-

METR S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S

Cmax 2.346 2.257 2.242 2.317 2.251 2.245 2.359 2.356 2.359 2.356 2.353 2.341 2.24 2.225

Tmax 3.000 2.500 3.000 2.500 3.000 2.500 3.000 2.500 3.000 3.000 2.500 3.000 2.500 3.000

Kel 0.102 0.118 0.120 0.113 0.091 0.120 0.111 0.112 0.078 0.118 0.092 0.118 0.113 0.106

Half

life 6.790 5.849 5.782 6.152 7.589 5.760 6.250 6.208 8.918 5.888 7.548 5.879 6.150 6.531

AUC

0-t 21.97 19.68 18.90 20.25 21.52 21.02 21.52 20.76 22.31 20.33 22.96 20.31 20.18 20.93

AUC 0-

inf 22.81 20.11 19.42 20.91 22.45 21.33 21.98 21.19 23.13 20.86 23.71 20.71 20.77 21.56

AUMC

0-t 166.4 144.7 132.7 146.5 169.6 156.8 163.6 154.8 185.8 146.9 187.5 151.2 147.9 157.7

AUMC

0-inf 194.9 158.9 149.4 168.0 202.1 167.0 178.8 169.0 216.2 164.1 213.7 164.5 167.2 178.5

MRT 8.541 7.901 7.694 8.037 8.040 7.829 8.136 7.975 8.344 7.865 8.013 7.940 8.049 8.279

VD 0.447 0.589 0.617 0.539 0.407 0.564 0.505 0.527 0.336 0.564 0.387 0.569 0.543 0.492

CLT 4.383 4.972 5.150 4.782 4.455 4.688 4.550 4.720 4.323 4.794 4.217 4.828 4.815 4.639

Page 225: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

194

Table 6.21: Comparison of (mean ± SEM) of Bioavailability and Pharmacokinetics of oral formulation of liposomes (LP-05, LP-11) and marketed brand of Diclofenac sodium (Phlogin-SR®

Brooks Pharma) administered in the oral dose of 100 mg to healthy subjects (n=18)

Units of Cmax =µg.ml-1, Tmax= hrs, Kel = hr-1, T1=0.5= hrs, AUC (total) = µg.h.ml-1, AUMC (total)= µg.h2.ml-1, MRT = hrs, Ka = hr-1, VD=L.kg-1, CLT=

mL.hr-1

PARA-

METR

DSF-PSL

(Mean ± SEM)

DSF-PEL

(Mean ± SEM)

Phlogin-SR®

(Mean ± SEM)

Cmax 2.485 ± 0.012 2.651 ± 0.021 2.299 ± 0.012

Tmax 3.056 ± 0.053 2.861 ± 0.054 2.833 ± 0.057

Kel 0.111 ± 0.003 0.111 ± 0.003 0.107 ± 0.003

Half life 6.249 ± 0.087 6.282 ± 0.152 6.532 ± 0.189

AUC

0-t 23.20 ± 0.171 24.36 ± 0.279 20.94 ± 0.227

AUC

0-inf 23.84 ± 0.213 25.02 ± 0.307 21.53 ± 0.250

AUMC

0-t 172.7 ± 1.499 182.3 ± 3.429 158.6 ± 3.240

AUM

0-inf 193.9 ± 2.866 204.3 ± 4.794 178.4 ± 4.299

MRT 8.126 ± 0.057 8.148 ± 0.102 8.102 ± 0.053

VD 0.468 ± 0.010 0.447 ± 0.014 0.502 ± 0.017

CLT 4.200 ± 0.037 4.006 ± 0.047 4.656 ± 0.054

Page 226: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

195

Table 6.22: Bioavailability and pharmacokinetic parameters of oral formulation of

Liposomes of Diclofenac potassium (LP-17) administered in the oral dose of 50 mg to healthy human subjects (n=18)

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18

1.729 1.72 1.752 1.745 1.74 1.807 1.779 1.675 1.79 1.83 1.766 1.727 1.73 1.754 1.761 1.722 1.767 1.757

3.000 3.000 2.500 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000

0.110 0.113 0.120 0.115 0.129 0.133 0.126 0.112 0.125 0.110 0.129 0.113 0.120 0.123 0.118 0.114 0.121 0.114

6.302 6.156 5.780 6.041 5.368 5.223 5.496 6.178 5.562 6.321 5.378 6.158 5.780 5.652 5.874 6.087 5.751 6.084

18.25 16.72 15.26 16.40 16.46 16.27 16.05 16.28 16.31 16.48 15.83 17.59 16.04 16.20 16.43 17.02 16.36 17.36

18.82 17.27 15.74 16.90 16.68 16.46 16.35 16.77 16.72 17.08 16.09 18.15 16.44 16.52 16.86 17.56 16.72 17.91

139.3 123.6 108.3 122.5 121.7 119.1 117.4 121.7 117.1 123.2 115.8 131.3 117.4 119.4 121.5 125.6 120.8 128.2

158.2 141.7 123.7 138.7 128.8 125.3 126.7 137.9 130.2 143.2 124.0 149.7 130.4 129.6 135.2 143.5 132.6 146.3

8.408 8.208 7.862 8.210 7.720 7.610 7.752 8.219 7.790 8.379 7.704 8.246 7.933 7.847 8.023 8.171 7.926 8.167

0.584 0.652 0.762 0.679 0.774 0.806 0.771 0.669 0.745 0.642 0.801 0.620 0.730 0.742 0.700 0.648 0.721 0.636

5.313 5.791 6.355 5.919 5.994 6.075 6.117 5.962 5.981 5.853 6.215 5.509 6.084 6.054 5.932 5.694 5.980 5.583

PARA-

METR S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-14 S-15

Cmax 1.815 1.806 1.840 1.832 1.827 1.897 1.868 1.759 1.880 1.921 1.854 1.813 1.817 1.842 1.849

Tmax 3.000 3.000 2.500 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000 3.000

Kel 0.110 0.113 0.120 0.115 0.129 0.132 0.126 0.113 0.124 0.110 0.129 0.112 0.120 0.123 0.118

Half

life 6.297 6.145 5.791 6.045 5.389 5.247 5.503 6.159 5.569 6.321 5.361 6.174 5.777 5.656 5.865

AUC

0-t 19.16 17.54 16.03 17.22 17.29 17.08 16.86 17.09 17.13 17.31 16.62 18.48 16.84 17.01 17.25

Page 227: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

196

Table 6.23: Bioavailability and pharmacokinetic parameters of oral formulation of

Liposomes of Diclofenac potassium (LP-23) administered in the oral dose of 50 mg to healthy human subjects (n=18)

AUC 0-

inf 19.76 18.12 16.53 17.74 17.53 17.29 17.17 17.60 17.56 17.94 16.88 19.07 17.26 17.35 17.69

AUMC

0-t 146.2 129.7 113.8 128.6 127.8 125.0 123.3 127.8 123.0 129.4 121.6 137.9 123.4 125.4 127.5

AUMC

0-inf 166.1 148.6 130.0 145.7 135.5 131.7 133.2 144.4 136.9 150.2 129.9 157.6 136.9 136.2 141.8

MRT 8.404 8.202 7.868 8.212 7.731 7.620 7.756 8.205 7.796 8.376 7.695 8.260 7.930 7.849 8.015

VD 0.557 0.622 0.724 0.646 0.734 0.764 0.734 0.639 0.709 0.611 0.766 0.589 0.695 0.706 0.668

CLT 5.061 5.519 6.051 5.636 5.705 5.784 5.824 5.682 5.694 5.575 5.923 5.243 5.794 5.764 5.652

Page 228: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

197

Table 6.24: Bioavailability and pharmacokinetic parameters of marketed brand of Diclofenac potassium (Caflam® Novertis Pharmaceuticals, Pakistan) administered in the oral dose of 50 mg to healthy human subjects (n=18

S-1 S-2 S-3 S-4 S-5 S-6 S-7 S-8 S-9 S-10 S-11 S-12 S-13 S-14 S-15 S-16 S-17 S-18 MEAN

1.365 1.356 1.346 1.487 1.367 1.346 1.525 1.505 1.458 1.444 1.408 1.329 1.356 1.400 1.357 1.354 1.334 1.484

2.000 2.000 2.000 2.000 2.000 2.000 2.000 2.000 2.500 2.000 2.000 2.000 2.000 2.000 2.000 2.000 2.000 2.000

0.197 0.250 0.275 0.299 0.290 0.276 0.312 0.296 0.315 0.286 0.307 0.286 0.236 0.287 0.256 0.276 0.290 0.308

3.511 2.774 2.522 2.318 2.389 2.508 2.222 2.341 2.202 2.423 2.255 2.419 2.939 2.413 2.711 2.516 2.393 2.249

5.359 5.362 5.134 5.207 5.070 5.204 5.549 5.459 5.391 5.308 4.944 4.935 5.288 5.162 5.123 5.096 4.937 5.465

5.805 5.570 5.265 5.294 5.170 5.327 5.613 5.546 5.458 5.430 5.022 5.039 5.538 5.266 5.299 5.223 5.030 5.536

20.52 19.47 18.20 18.06 17.67 18.63 19.84 19.41 19.02 18.82 17.11 17.67 19.42 18.13 18.41 18.06 17.44 19.41

28.13 22.80 20.25 19.39 19.21 20.55 20.81 20.76 20.03 20.71 18.30 19.29 23.48 19.75 21.21 20.04 18.88 20.50

4.846 4.093 3.846 3.662 3.716 3.857 3.708 3.743 3.670 3.814 3.644 3.827 4.240 3.750 4.002 3.838 3.752 3.703

3.401 4.486 5.220 5.648 5.612 5.188 5.557 5.338 5.766 5.269 6.122 5.685 4.259 5.455 4.826 5.275 5.758 5.567

17.23 17.95 18.99 18.89 19.34 18.77 17.82 18.03 18.32 18.42 19.91 19.84 18.06 18.99 18.87 19.15 19.88 18.06

Page 229: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

198

Table 6.25: Comparison of (mean ± SEM) of Bioavailability and

Pharmacokinetics of oral formulation of liposomes (LP-05, LP-11) and marketed brand of Diclofenac potassium (Caflam®

Novartis Pharmaceuticals) administered in the oral dose of 50 mg to healthy subjects (n=18)

Units of Cmax =µg.ml-1, Tmax= hr, Kel = hr-1, T1/2= hrs, AUC (total) = µg.h.ml-1

AUMC (total)= µg.h2.ml-1, MRT = hrs, Ka = hr-1, VD=L.kg-1, CLT= mL.hr-1

PARA-

METR

DSF-PSL (Mean ± SEM)

DSF-PEL (Mean ± SEM)

Caflam (Mean ± SEM)

Cmax 1.753 ± 0.008 1.840 ± 0.009 1.401 ±0.015

Tmax 2.972 ± 0.028 2.972 ± 0.028 2.028 ± 0.027

Kel 0.119 ± 0.002 0.119 ± 0.002 0.280 ± 0.007

Half life 5.845 ± 0.081 5.846 ± 0.079 2.506 ± 0.075

AUC

0-t 16.52 ± 0.161 17.34 ± 0.169 5.221 ± 0.044

AUC

0-inf 16.95 ± 0.179 17.79 ± 0.188 5.357 ± 0.052

AUMC

0-t 121.9 ± 1.563 128.0 ± 1.637 18.63 ± 0.218

AUMC

0-inf 153.9 ± 2.290 142.7 ± 2.393 20.78 ± 0.526

MRT 8.010 ± 0.058 8.010 ± 0.057 3.873 ± 0.068

VD 0.705 ± 0.016 0.671 ± 0.015 5.246 ± 0.153

CLT 5.912 ± 0.060 5.630 ± 0.058 18.70 ± 0.181

Page 230: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

199

6.7 Plasma Level Time Profile of Liposomal Formulation of DFS and DFP

6.7.1 Plasma level time profile of diclofenac sodium (DFS) with purified soya

lecithins (PSL) and purified egg lecithins (PEL)

Bioavailability and Pharmacokinetic parameters of liposomal formulations of

DFS prepared with purified soya lecithin (PSL, LP-05) and purified egg

lecithin (PEL, LP-11) were determined after administration of an oral dose of

100 mg to eighteen healthy human subjects, after processing plasma sample for

the determination of plasma concentration and using pharmacokinetic software

Kinetica® version 4.4.

6.7.2 Comparison of plasma concentration of Liposomal formulations of

diclofenac sodium with Phlogin-SR®

Both of the formulations showed higher plasma concentration than reference

brand. The difference was significant (p<0.05) at all levels of data points with

for DFS-PEL formulations (LP-17) while the significant difference (p<0.05)

was observed with the formulations of (LP-05) at data points of 1.5, 2, 2.5, 3, 4,

6 and 8 hours. The higher values of plasma concentration with liposomes are

well understood and documented in literature. In a study conducted by

Charman et al., (1997), incorporation of lipids into drug formulations enhances

membrane activities of poor soluble drugs. Therefore, results of present study

of enhancing plasma concentration of diclofenac sodium having less solubility

in aqueous medium by formulating with phospholipid vesicles. The relative

higher concentration of diclofenac in plasma as compared to non-lipid based

formulation (Phlogin-SR®) is due to induction of dissolved state

(solubilization) of the poorly soluble drugs by incorporation of lipids resulting

Page 231: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

200

in comparatively better absorption (Humberstone and Charman, 1997). Another

mechanism for enhancing the plasma level has been described by Aungst,

(2000) that liposomes enhances the permeation of drugs in intestine due to

presence of phospholipids and also described that lipid formulations increase

the time available for absorption of drugs by increasing gastric retention time.

Another study conducted by Dintaman et al., (1999) demonstrated that

phospholipids incorporated drugs may get entry into circulation by altering

physical and biochemical barrier function of the GI tract as it is observed that

lipid formulation may enhance the extent of lymphatic transport and improve

bioavailability by reducing the first-pass metabolism. All these mechanisms are

believed to reduce the loss of drugs in biological system and increase its

absorption profiles; therefore, higher concentration appeared in plasma matrix.

6.7.3 Plasma level time profile of diclofenac potassium (DFP) with

purified soya lecithins (PSL) and purified egg lecithins (PEL)

The plasma level-time profile of liposomal formulations of DFP prepared with

purified soya lecithin (PSL, LP-17) and purified egg lecithin (PEL, LP-23)

were administered in the oral dose of 50 mg to eighteen healthy human

subjects. Diclofenac potassium sugar coated preparation available in market as

Caflam®-Novartis Pharmaceuticals Pakistan, (Ltd.) was selected as the

reference product. The test formulations and reference brand were administered

at different occasions according to the Latin Square Design. The plasma drug

concentrations were determined by sensitive and validated HPLC method. The

plasma concentrations at different time intervals were used to compute

bioavailability and pharmacokinetic parameters. Plasma levels from both

Page 232: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

201

formulations (LP-17 and LP-23) were higher than reference brand of diclofenac

potassium.

6.7.4 Comparison of Plasma Concentration of Liposomal Formulations

of Diclofenac Potassium with Caflam®-Novartis Pharmaceuticals

Liposomal formulation (LP-17) and (LP-23) formulated with purified soya

lecithin and purified egg lecithin with incorporation of PEG (10%). Both of the

formulations (LP-17 and LP-23) have shown higher plasma concentration than

reference brand. The difference was significant (p<0.05) at all levels of data

points with for DFP-PSL (LP-17) DFP-PEL (LP-23) formulations.

An important finding of the present study is higher values of plasma

concentration of diclofenac potassium formulations in comparison with

diclofenac sodium. This difference might be due to difference in nature of salts

as diclofenac potassium is considered to have higher water solubility and

permeability than diclofenac sodium. Another study by Chuasuwan et al.,

(2009) demonstrated that diclofenac potassium dissolved faster and hence,

absorbed faster. This is further studied that diclofenac has short onset of action

and therefore having higher plasma concentration after oral administration.

Another important finding of the present study of in-vivo liposomes evaluation

is that the plasma concentration with egg lecithin is comparatively larger than

soya lecithins. This is best explained in a study conducted by Sriram and

Rhodes, (1995) demonstrating that contents of phosphatidyl choline were

comparatively higher in egg lecithins than that of soya lecithins, which in turn

induced more solubilization than soya lecithin formulation. The liposomes can

also enhance solubilization of encapsulated drugs as a function of concentration

Page 233: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

202

of phospholipid and unionized drug by mobilizing the phospholipid contents in

the bilayer.

6.8 Pharmacokinetics Profile of Liposomal Formulation of Diclofenac Sodium

and Diclofenac potassium

The results of plasma concentrations were computed by non-compartmental

pharmacokinetics analysis of all liposomal formulations of diclofenac sodium

and diclofenac potassium. The dose of diclofenac in these formulations was

100 and 50 mg, for sodium and potassium salts, respectively. The

documentation was done for taking informed consent on describing all the

information regarding this study. The Latin-square crossover design was

selected because each subject act as his own control and inter-subject

variability is avoided (Leon Shargel et al., 2005)

6.8.1 Pharmacokinetics Profile of Liposomal Formulations of Diclofenac

Sodium

Chemically, Diclofenac sodium (DFS) is sodium 2- (2, 6- dichloro anilino)

phenyl acetic acid. Since its induction as therapeutic agent in 1974, it is

considered as potent non-steroidal anti-inflammatory drug having analgesic and

antipyretic properties. It is most often prescribed in the treatment of several

diseases including rheumatoid arthritis and osteoarthritis. As the case similar to

other NSAIDs, DFS also having side effects of disturbing gastrointestinal

function and also have toxicity related to kidney and liver when administered

via oral route and it is also a report of cutaneous lesion with the intra-muscular

administration.

Page 234: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

203

Maximum plasma concentration, Cmax is the peak concentration of the drug that

reaches the systemic circulation after administration. The values (mean ± SEM)

of Cmax were found as 2.485 ± 0.012 µg.mL-1 for LP-05. The valves of Cmax for

LP-11 and Phlogin-SR® were calculated as 2.651 ± 0.021 µg.mL-1 and 2.299 ±

0.012 µg.mL-1. After ANOVA application, significant difference in the Cmax

values (P>0.05) was observed between liposomal formulations (LP-05 and LP-

11) compared statistically to the market brand. The increased value of Cmax

with liposomal formulations is evident from the fact that liposomes improve

the bioavailability of drugs. The liposomal formulation (LP-05) composed of

an optimized amount of purified soya lecithin and cholesterol same as (LP-11)

consist of egg lecithin and cholesterol (75:25) while the amount of drug was

same in both formulations. The addition of polyethylene glycol (10%) further

stabilized the formulations. In liposomes, nature of phospholipids is of prime

importance for sustained and improved bioavailability of drugs. The lipids used

in the present study are classified as natural and digestible. The increased

bioavailability of drugs from lipids has been studied by various workers. In a

study conducted by Myers and Stella, (1992) demonstrated that phenomenon

involved in higher absorption of drugs is the incorporation of liposomes into

micellar phase, which in turn enhance the solubilization and hence the peak

plasma concentration of drugs is higher than non-liposomal dosage forms. The

value of Cmax in a pharmacokitics study conducted by Andreas et al., (2000)

stating that nystatin concentration was comparable to intravenous

administration. Similar findings were observed by Zuzanna and Agasta, (2010)

Page 235: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

204

and Sharon et al., (2006). The results of both studies demonstrated higher

absorption and bioavailability of drugs incorporated in liposomes.

The values (mean ± SEM) for Tmax were 3.056 ± 0.053 hrs, 2.86 ± 0.054 hrs

and 2.833 ± 0.057 hrs for LP-05, LP-11 and Phlogin-SR®, respectively. There

was significant difference (p<0.05) for LP-05 and reference brand while it was

non-significant (p>0.05) when comparison was made between LP-11 and

reference brand. Tmax represents rate of drug absorption. So it was observed

that liposomal formulation LP-11 showed higher rate of drug absorption than

LP-05 and reference brand. This is evident by the fact that LP-11 contains egg

lecithins which possess higher content of phospholipids compared to soya

lecithins which in turn possess high solubilization and hence absorption at

faster rate (Shivhare et al., 2009; Sriram and Rhodes, 1995).

Zero moment curve, a component of statistical moment theory also known as

the area under the plasma concentration time curve (AUC) is an important

pharmacokinetic parameter in non-compartmental pharmacokinetics analysis. It

is useful for calculating the relative effectiveness of different drug products.

Trapezoidal rule was applied for its determination. The value of AUC0-∞ of

liposomal formulations (LP-05, LP-11 and reference brand) was found in this

study as 23.84 ± 0.213 µg.hr.mL-1, 25.02 ± 0.301 µg.hr.mL-1 and 21.53 ± 0.250

µg.hr.mL-1 (mean ± SEM), respectively. The ANOVA results indicated that the

value of AUC of LP-05 and reference brand were non-significantly different

from each other. LP-05 value of AUC was higher than reference brand. A

statistical difference (p<0.05) was observed between LP-11 and reference

Page 236: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

205

brand. These present study values were found in line with the studies available

in literature. Roopa and Kusum, (2009) developed liposomal formulation of

rifampicin and isoniazid using different lipids. The pharmacokinetics

evaluation of liposomes has demonstrated significant increased values of AUC

compared to the plain rifampicin (from 374.9 ± 5.41 to 2399.0 ± 3.12

µg.hr.mL-1). In another study presented by Andreas et al., (2000), the values

of AUC0–24 were increased from 11.65 to 67.44 µg.h.mL-1 showing highly

significant increase in value of AUC with liposomal formulation at (P < 0.01).

We have compared liposomal formulations in the present study with available

sustained release brand of diclofenac sodium (Phlogin-SR®); therefore, non-

significant difference was found with LP-05 and significant with LP-11. The

difference between two formulations is due to the nature of phospholipids. Egg

lecithin produced higher values of AUC compared to the soya lecithin

(Shivhare et al., 2009). The value of AUC of diclofenac calculated in a

previous study of Lill et al., (2000) demonstrated values of 7008 ± 2006

ng.hr.mL-1 in healthy male subject after oral administration of 150 mg of

diclofenac sodium. The lesser values of AUC are due to the plain formulation

compared to liposomal formulations of the present study.

Another important parameter of non-compartmental pharmacokinetic analysis

is the first moment curve, i.e. area under first moment curve (AUMC). This

parameter is essential for calculating mean residence time in the body. The

AUMC is calculated by taking product of time and concentration versus time.

Trapezoidal rule is used for calculating this parameter (Shargel et al., 2005).

Page 237: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

206

The value of AUMC0-∞ of liposomal formulations (LP-05, LP-11 and reference

brand) was found in this study as 193.9 ± 2.866 µg.hr2.mL-1, 204.3 ± 4.792

µg.hr2.mL-1 and 178.4 ± 4.299 µg.hr2.mL-1 (mean ± SEM), respectively. The

ANOVA results indicated that the value of AUMC0-∞ of LP-05 and reference

brand were non-significantly (p< 0.05) different. LP-05 value of AUMC0-∞ was

higher than reference brand. A statistical difference (p<0.05) was observed

between LP-11 and reference brand. This dissimilarity in values is due to the

difference in nature of formulations. Reference brand is sustained release

formulation of diclofenac sodium using certain sustained polymers (e.g. ethyl

cellulose) of Brooks Pharmaceuticals Pakistan, where as we have formulated

liposomes using phospholipids which predominantly enhance plasma levels,

hence affecting all pharmacokinetics parameters (Bedu-Addo et al., 1996).

Mean residence time (MRT) also known as sojourn time or staying time is

defined as an average time of a drug molecules staying in the body. Liposomes

are believed to enhance the staying times of the drugs in the body (Shargel et

al., 2005). The incorporation of amphipathic hydrophilic polymers poly

ethylene glycol (PEG) into liposomal formulation also meant for extending

residence time of drugs in body (Sakae et al., 1995; Maruyama et al., 1991).

The value of MRT of liposomal formulations (LP-05, LP-11 and reference

brand) was calculated as 8.126 ± 0.057 hr, 8.148 ± 0.102 hr and 8.102 ± 0.053

hr (mean ± SEM), respectively. The ANOVA results indicated that the value of

MRT of LP-05, LP-11 and reference brand were non-significantly (p<0.05)

Page 238: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

207

different from each other. The values of MRT of liposomal formulation (LP-05

and LP-11) were higher than reference brand.

The values of MRT reported in a previous study as 3.355 ± 0.329 hr in normal

human after oral administration of diclofenac sodium Farid et al., (2005). The

study also conducted in the presence of food in which the value of mean

residence time also extended to 3.655 ± 0.484 hr. The value of MRT in our

study is more than twice than reported by (Farid et al., 2005). The values of

MRT in liposomal formulation of rifampicin and isoniazid were also increased

2.2 ± 0.69 hr to 4.6 and 1.8 ± 0.21 hr to 5.4 ± 0.35 hr (Roopa and Kusum,

2009). This increase in residence time supported the sustained and prolonging

effect of liposomal formulations.

Half-life expresses the time period required for the amount of a drug to

decrease by one-half (Shargel et al., 2005). The half-life is the parameter that

reflects the rate of drug elimination, similar to the elimination rate constant

(Mohsin, 2007; Vinkateswarlu, 2006). The half-life calculated in the present

study for LP-05, LP-11 and Phlogin-SR® as 6.249 ± 0.087 hr, 6.282 ± 0.152 hr

and 6.532 ± 0.189 hr, respectively. The ANOVA results indicated that the

value of T1/2 of LP-05, LP-11 and reference brand were non-significantly

(p<0.05) different from each other. Similarly, the values of elimination rate

constant (Ke) were found as 0.111 ± 0.003 hr-1, 0.111 ± 0.003 hr-1and 0.107 ±

0.003 hr-1 for LP-05, LP-11 and reference drug product (Phlogin-SR®). The

value of half life of diclofenac sodium in the previous study was reported as

1.67 ± 0.74 hr-1 (mean ± SD) and value of Ke was 0.50 ± 0.23 hr-1 by Pejcic at

Page 239: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

208

al., (2006) after intramuscular administration to pegs. Similarly, Abdulla et al.,

(2006) reported the values of diclofenac in the range of 2-3 hr-1 in sheep. Farid

et al., (2005) reported the values of elimination half life as 2.627 ± 0.189 hr-1

in fasting and 2.72 ± 0.29 hr-1 in fed state in normal healthy subjects. The

values of half-life of liposomal preparation of the present study were three fold

increased compared to the conventional dosage forms. This shows that

liposomal preparation will remain in the body for longer period of time, hence

will exert sustained action with less elimination from body. The results of half

lives of liposomal formulations of the present study were also found in line

with the study of (Roopa and Kusum, 2009), where the values of half life were

increased from 2.004 ± 0.49 hr s to 3.37 hrs for rifampicin and 1.7 ± 0.13 hrs to

4.53 ± 0.16 hrs for isoniazid (mean ± SD).

Volume of distribution, abbreviated as VD. It represents the volume that must

be considered in estimating the amount of drug in the body from the

concentration of drug found in the sampling compartment. VD does not have a

true physiological meaning in terms of space, hence the term apparent volume

of distribution is used (Shargel et al., 2005). It is mathematically derived

feature concerning the quantity of drug in the body and concentration of drug

in the measured compartment, usually plasma (Mohsin, 2007). The values of

VD in the present study are 0.468 ± 0.010 L.Kg-1, 0.447 ± 0.014 L.Kg-1 and

0.502 ± 0.017 L.Kg-1 (mean ± SEM) for LP-05, LP-11 and reference drug

product (Phlogin-SR®), respectively. A study conducted by Quintavalle et al.,

(1997) demonstrated the value of volume of distribution as 0.98 for the normal

Page 240: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

209

volunteers. Another summary report of the European Agency for the

Evaluation of Medicinal Products (2003) affirmed that volume of distribution

of diclofenac sodium is small in man and found in the range 0.15 to 0.21 L.Kg-1

(dose was 25 mg of the enteric coated tablet). The value of small volume of

distribution is justified by the extensive binding of the drug to the plasma

proteins. The difference in values of volume of distribution between present

study and literature values is less and may be due to the size of the dose as 100

mg of diclofenac was administered in the present study. It is revealed in

literature that diclofenac has little volume of distribution as this drug is

extensively bound to plasma proteins (99.7%).

Total body clearance is an important pharmacokinetic parameter and is

abbreviated as (CLT). This is a measure of drug elimination with out

identifying the mechanism or process. CLT is defined as volume of plasma fluid

that is cleared of the drug per unit time (Shargel et al., 2005). CLT explains

how rapidly drugs are eliminated, metabolized or distributed all over the body.

It can be considered as the proportionality constant linking the rate of these

processes and drug concentration (Mohsin, 2007). In the present study, the

(Mean ± SEM) values for the total body clearance (CLT) were 4.20 ± 0.037,

4.006 ± 0.047 and 4.656 ± 0.054 mL.h-1.Kg-1 for LP-05, LP-11 and Phlogin-

SR®, respectively. The values of clearance were reported in a previous study as

5.03 ± 0.045 mL.h-1.Kg-1; calculated from dose/AUC0-∞, as 10 mg/kg dose was

administered to rats (Maria et al., (2008). The values of present studies were

comparatively less than study of Maria et al., (2008) demonstrating that less

Page 241: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

210

elimination of drug resulting in longer and sustained effect in body (Roopa and

Kusum, 2009). This finding of less clearance is evident by decreased

elimination rate of drug in the present study. The decreased elimination rate

and longer half lives of dugs indicate decreased clearance of drugs from the

body. In a study reported by Abdulla et al., (2006) the values of clearance after

intravenous and intramuscular administration were 87.86 ± 24.10 and 85.69 ±

40.76 mL.hr-1.Kg-1 (mean ± SD). The values of Abdulla et al., (2006) were

higher than values of clearance in the present study. Another study by Pejcic et

al., (2006) demonstrated the value of CLT as 0.292 ± 0.101 L.hr-1.Kg-1. The

value reported by Davies and Anderson, (1997) was 0.2 L.hr-1.Kg-1 to 0.6 L.hr-

1.Kg-1. In conclusion, values of CLT of literature studies were higher than

described in present study, showing rapid elimination of diclofenac from body

of all the reference studies compared to less CLT values of liposomal

formulations.

Page 242: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

211

6.8.2 Pharmacokinetics Profile of Liposomal Formulations of Diclofenac

potassium

Diclofenac potassium (DFP) is potassium 2- (2, 6- dichloro anilino) phenyl

acetic acid, a faintly yellowish white to light beige, odorless, hygroscopic

crystalline powder. Sparingly soluble in water, that is why placed in BCS class

II drugs. The pharmacokinetics parameters were determined by computing

plasma concentration time profile in Kinetica software®-4.4 version.

The liposomal formulations (LP-17) formulated with purified soya lecithin

(PSL) & cholesterol (75:25) ratio with the addition of poly ethylene glycol

(PEG) in percentage of 10 and (LP-23) formulated with optimized

concentration of purified egg lecithin (PEL) & cholesterol (75:25) ratio with

the addition of poly ethylene glycol (PEG) in concentration of 10 %. The

concentration of diclofenac potassium was kept constant. Comparison of LP-17

and LP-23 was made with commercially available brand of diclofenac

potassium, Caflam®-Novartis Pharmaceuticals Pakistan (Ltd.). As DFP is not

available in sustained release formulations and often is available as coated

(Sugar or film) drug formulation. Therefore, it was decided to select Caflam®-

Novartis Pharmaceuticals as reference standard for evaluating bioavailability

and pharmacokinetics.

The values (mean ± SEM) of Cmax were found as 1.753 ± 0.008 µg.mL-1 for

LP-17. The valves of Cmax for LP-23 and Caflam® were calculated as 1.840 ±

0.009 µg.mL-1 and 1.401 ± 0.015 µg.mL-1. After applying ANOVA, significant

difference in the Cmax values (P>0.05) was observed between liposomal

formulations (LP-17 and LP-23) compared statistically to the market brand.

Page 243: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

212

Similarly, the values (mean ± SEM) for Tmax were 2.972 ± 0.028 hrs, 2.972 ±

0.028 hrs and 2.028 ± 0.027 hrs for LP-17, LP-23 and Caflam®, respectively.

There was significant difference (p<0.05) for (LP-17) and (LP-23) with the

reference brand. The value of AUC0-∞ of liposomal formulations (LP-17, LP-

23 and reference brand) was found in this study as 16.95 ± 0.179 µg.hr.mL-1,

17.79 ± 0.188 µg.hr.mL-1 and 5.357 ± 0.044 µg.hr.mL-1 (mean ± SEM),

respectively. The value of AUC0-∞ was also statistically different from

reference brand (p<0.05). Cmax, Tmax and AUC0-∞ are considered as basic

pharmacokinetic parameters accessed from blood and this is a direct method of

estimation of bioavailability of any dosage form. The parameter furnishes the

information regarding rate and extent of drug absorption and provide mean for

accessing pharmacodynamics of drug products (Shargel et al., 2005). The

values of Cmax, Tmax reported in previous study of Lissy et al., (2010) as 1.125

µg.mL-1, 0.45 hr while values of Cmax, Tmax and AUC0-∞ by Ralph and Keith,

(2009) as 1.156 µg.mL-1, 0.49-0.51 hr and 1521 ± 377 ng.hr.mL-1. The values

in the reported studies were found less than values of liposomal formulations of

the present study. Literature search revealed a study by (Roopa and Kusum,

2009) on pharmacokinetics evaluation of developed liposomal formulation of

rifampicin and isoniazid using lipids demonstrated significant increased values

of Cmax, Tmax and AUC0-∞ compared to plain rifampicin (from 374.9 ± 5.41

µg.hr.mL-1 to 2399.0 ± 3.12 µg.hr.mL-1). Andreas et al., (2000) described

AUC0–24 increased from 11.65 µg.h.mL-1 to 67.44 µg.h.mL-1. It is deliberate in

the current study that liposomal formulations produced higher values as

Page 244: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

213

compared with available sugar coated brand of diclofenac potassium

(Caflam®); therefore, non-significant difference was found with LP-17 and LP-

23. Purified Egg lecithin (PEL) formulations produced higher values of three

above mentioned parameters compared to the soya lecithin (Shivhare et al.,

2009).

The values of AUMC0-∞ of liposomal formulations (LP-17, LP-23) and

reference brand were found in this study as 153.9 ± 2.290 µg.hr2.mL-1, 142.7 ±

2.393 µg.hr2.mL-1 and 20.78 ± 0.526 µg.hr2.mL-1 (mean ± SEM), respectively.

The ANOVA results indicated that the value of AUMC0-∞ of (LP-17 and LP-

23) and reference brand were significantly different. Liposomal formulations

were found significantly higher than reference brand.

The value of MRT of liposomal formulations (LP-17, LP-23 and reference

brand) was calculated as 8.010 ± 0.058, 8.010 ± 0.102 and 8.102 ± 0.053 hr

(mean ± SEM), respectively. The ANOVA results indicated that the value of

MRT of LP-17, LP-23 and reference brand were non-significantly different

from each other. The values of MRT of liposomal formulation (LP-17 and LP-

23) were significantly higher (p<0.05) than reference brand. Liposomes are

believed to enhance the staying times of the drugs in the body. The

incorporation of amphipathic hydrophilic polymers poly ethylene glycol (PEG)

into liposomal formulation also meant for extending residence time of drugs in

body (Sakae et al., 1995; Maruyama et al., 1991). The values of MRT in

liposomal formulation of rifampicin and isoniazid were also increased 2.2 ±

0.69 to 4.6 and 1.8 ± 0.21 to 5.4 ± 0.35 hrs (Roopa and Kusum, 2009). This

Page 245: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

214

increased in residence time supported the sustained and prolonging effect of

liposomal formulations.

The half-life calculated in the present study for LP-17, LP-23 and Caflam® as

5.845 ± 0.081, 5.845 hr ± 0.079 and 2.506 ± 0.075 hr, respectively. The

ANOVA results indicated that the value of T1/2 of (LP-17 & LP-23) and

reference brand were significantly different (p<0.05). Similarly, the values of

elimination rate constant (Ke) were found as 0.119 ± 0.002 hr-1, 0.111 ± 0.002

hr-1 and 0.280 ± 0.007 hr-1for LP-17, LP-23 and reference drug product

(Caflam). The value of half life of diclofenac potassium reported in the range

of 0.4-1.6 hr and mean value 0.9 ± 0.3 hr (mean ± SD) by Burkhard at al.,

(2006) after intravenous administration to human at dose of 22.5 mg. Similarly,

Sweetman, (2009) reported values of half life of diclofenac potassium in the

range of 1-2 hrs in human. The liposomal formulations of DFP presented

higher values of half-life up to three fold increased compared to the

conventional dosage forms showing longer stay of drug in the body for longer

period of time and will exert sustained action. This is confirmed by a literature

study of Roopa and Kusum, (2009) where the values of half lives were

increased from 2.004 ± 0.49 to 3.370 ± 0.26 hrs for rifampicin and 1.7 ± 0.13 to

4.53 ± 0.16 for isoniazid (mean ± SD).

The values of VD in the present study are 0.0.357 ± 0.009 L.Kg-1, 0.334 ± 0.007

L.Kg-1 and 2.680 ± 0.074 L.Kg-1 (mean ± SEM) for LP-17, LP-23 and reference

drug product (Caflam®), respectively. The significant difference (p<0.05) in the

formulations of liposomes and Caflam® was evident that liposomal formulation

Page 246: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

215

remain in plasma and not perfused to the tissues confirming the long retaining

of drugs in body. In the present study, the (Mean ± SEM) values for the total

body clearance (CLT) were 3.003 ± 0.009 mL.h-1.Kg-1, 2.81 ± 0.007 mL.h-1.Kg-

1 and 9.57 ± 0.054 mL.h-1.Kg-1 for LP-17, LP-23 and Caflam, respectively.

Similar (mean ± SEM) values were reported as 5.03 ± 0.045 mL.h-1.Kg-1 in rats

(Maria et al., (2008). The values of our studies were comparatively less than

study of Maria et al., (2008) demonstrating that less elimination of drug from

the body resulting in longer and sustained effect in the body. In a study

reported by Abdulla et al., (2006) the values of clearance after intravenous and

intramuscular administration were 87.86 ± 24.10 mL.hr-1.Kg-1 and 85.69 ±

40.76 mL.hr-1.Kg-1 (mean ± SD). This finding of less values of total body

clearance is evident by decreased elimination rate of drug in the present study

(Roopa and Kusum, 2009). The less values of elimination rate and longer half

lives of dugs indicated comparatively less clearance of drugs from the body.

6.9 Conclusions

In conclusion, DFS and DFP loaded liposomes were satisfactorily produced by

micro emulsification-evaporation (MEE) technique. The following conclusions

have been drawn.

1. The technique has presented some advantages such as avoiding the use

of higher energy apparatus and chlorinated solvent, reproducibility, and

the higher entrapment. The combination of techniques of preparation of

Page 247: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

216

liposomes produced desired characteristics of uniform particle size,

improved elegance and short reconstitution time.

2. The formulation was optimized by application of response surface

methodology. The selection of an optimum concentration of lecithin and

cholesterol (75:25) for maximum entrapment was an important task

completed before comprehensive study.

3. Morphological studies by scanning electron microscopy exhibited

spherical shape with smooth surfaces without aggregation or adhesion

and dynamic light scattering analysis by zetasizing showed that particle

size was in range of 1.01 ± 0.011 to 1.94 ± 0.009 µm with low

polydispersity index upto 0.224 ± 0.002 indicated narrow size

distribution.

4. DSC measurements and X-ray diffraction studies suggested that the drug

in liposomes was found in the semi crystalline or crystalline form. No

change in liposomal formulations was observed compared to individual

entities.

5. FT-IR showed interaction between the drug and phospholipids. These

FT-IR bands reflected weak electrostatics interactions between carboxyl

group of DFS and ammonia group of phospholipids, such interaction

may enhance drug permeability which contributed to higher

bioavailability

Page 248: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

217

6. Two HPLC methods were developed, validated and successfully applied

for analysis of DFS and DFP for entrapment, In-vitro and In-vivo

studies.

7. Entrapment efficiency of liposomal formulations (n=24) were

determined and found in range from 82.14 to 89.35 % (w/w). It was

observed that purified egg lecithin liposomal formulations resulted in

higher entrapment compared to purified soya lecithin liposomes.

8. The In-vitro release profile of DFS/DFP from liposomes exhibited

sustained and controlled release from liposomal formulations. It was

observed that all formulations followed zero order (approximate) and

higuchi release pattern revealed that diffusion of drug was principle

process for release of DFS/DFP from liposomes.

9. Similarity factor (f-2) was determined for liposomal formulations (n=12)

of DFS by comparing the release profiles of liposomal formulations

(LP-01 to LP-12) with reference standard (Phlogin-SR®). It was

determined that LP-05 (DFS prepared with PSL & 10 % PEG) and LP-

11 (DFS prepared with PEL & 10 % PEG) were having similar release

profile compared to the reference standard. Therefore, these

formulations were subjected to In-vivo evaluation.

10. The plasma level time profiles revealed following informations.

I. Plasma level time profile of DFS liposomal formulation prepared

with PEL was higher than PSL liposomes.

Page 249: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

218

II. Bioavailability of DFS liposomal formulation prepared with PEL

was higher than PSL liposomes.

III. DFP liposomal formulations prepared with PEL produced plasma

level higher than PSL liposomes.

IV. Bioavailability of DFP liposomal formulation prepared with PEL

was higher than PSL liposomes.

11. All other pharmacokinetic parameters were found in good agreement

with previous studies and suggesting sustained action of DFS and DFP.

Page 250: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

219

7. References

1. Abdelwahab O., Brian J. A. and Girishchandra B. P. (2003); Short-

term repeated-dose toxicity profile of archaeosomes administered to mice via intravenous and oral routes; International Journal of Toxicology, 22 (1): 9-23.

2. Abdulla Y. A., Khalid M. A., Badraddin M. A. and Rao M. A. K.

(2006); Pharmacokinetics of diclofenac in sheep following intravenous and intramuscular administration; Veterinary Anaesthesia and Analgesia, 33 (4): 241-245

3. Adrian J. E., Poelstra K. and Kamps J. A. (2007); Addressing liver

fibrosis with liposomes targeted to hepatic stellate cells; Journal of Liposome Research, 17 (3-4): 205-218.

4. Agatonovic K. S., Zivanovic L., Zecevic M. and Radulovic D.

(1997); Spectrophotometeric study of diclofenac-Fe (III) complex; Journal of Pharmaceutical and Biomedical Analysis, 16: 147-153.

5. Alessia P., iuseppina M., Giampaolo I. and Luigi G. (2007); High-

performance liquid chromatography analysis of anti-inflammatory pharmaceuticals with ultraviolet and electrospray-mass spectrometry detection in suspected counterfeit homeopathic medicinal products; Journal of Pharmaceutical and Biomedical Analysis, 43 (4): 1221-1227

6. Ali S. Y., Farideh M. and Zarrin E. (2008); Directly suspended

droplet three liquid phase microextraction of diclofenac prior to LC; Chromatographia, 67 (1-2): 49-53.

7. Amarnath S. and Uma S. S. (1997) Liposomes in Drug Delivery:

Progress and Limitations; 154: 123-140. 8. Andreas H. G., Diana M., Kathy W., Ruta P., Vidmantas P., Myrna

C., Aida F. R., Jeremy C., Stephen C. P. and Thomas J. W. (2000); Compartmental Pharmacokinetics and Tissue Distribution of Multilamellar Liposomal Nystatin in Rabbits; Antimicrobial Agents Chemotherapy, 44 (4): 950-957.

9. Arcelloni C., Lanzi R., Pedercini S., Moltani G., Fermo I., Pontiroli

A. and Paroni R. (2001); High performance liquid chromatographic determination of diclofenac in human plasma after solid phase extraction; Journal of Chromatography B, 763: 195-200.

Page 251: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

220

10. Asai Y., and Watanabe S., (1999); Interaction of sesame oil with soybean phosphatidylcholine and their formation of small dispersed particles; Journal of Microencapsulation, 16 (6): 705-713.

11. Auler Jr. J. O., Espada1 E. B., Crivelli1 E., Quintavalle T. B. G.,

Kurata A., Stolf1 N.A.G., Issy A.M., Paschoa O. E. D., Danhof M., Breimer D.D., Chamone D. A. F. and Santos S. R. C. J. (1997); Diclofenac plasma protein binding: PK-PD modelling in cardiac patients submitted to cardiopulmonary bypass; Brazellian Journal Medical & Biological Research, 30 (3) 369-374.

12. Aungst B. J. (2000); Intestinal permeation enhancers; Journal of

Pharmaceutical Sciences, 89: 429-442.

13. Bajdik J. and Pintye H. K. (2009); Effects of lecithin on the mechanical properties of hydroxyl propyl methyl cellulose free films; Pharmazie, 64 (5): 348-349.

14. Bangham A. D., Standish M. M. and Watkins J. C. (1965); Diffusion

of univalent ions across the lamellae of swollen phospholipids; Journal of Molecular Biology, 13: 238-252.

15. Barrett E. R. (2004); Nanosuspensions in drug delivery; Nature

Reviews Drug Discovery, 3: 785-796.

16. Bedu-Addo F. K., Tang P., Xu Y. and Huang L. (1996); Interaction of poly ethylene glycol phospholipid conjugates with cholesterol phosp-hatidylcholine mixtures: sterically stabilized liposome formulations; Pharmaceutical Research, 13: 718-724.

17. Bhaleraoa S. S. and Harshala A. R. (2003); Preparation,

optimization, characterization, and stability studies of salicylic acid liposomes; Drug Development and Industrial Pharmacy, 29 (4): 451-467.

18. Boris C. E. H. and Lasic D. D., (1996); A rigorous theory of remote

loading of drugs into liposomes; Langmuir 1996, 11, 3356-3368.

19. Bucci R., Magri A. D. and Magri A. L. (2000); DSC in the chemical analysis of drugs Determination of diclofenac in pharmaceutical formulations; Journal of Thermal Analysis and Calorimetry, 61: 369-376.

20. Burkhard H., Julia C., Bertold R., Henrike W., Thomas R., Andreas

S., Istvan S., Kay B. and Ulrike W. (2005); Bioavailability of

Page 252: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

221

diclofenac potassium at low doses; British Journal of Clinical Pharmacology, 59:80-84.

21. Busch U., Heinzel G., Narjes H., Nehmiz G., Türck D. and Krimmer

J. (1996); Pharmacokinetics of meloxicam in patients with hepatic cirrhosis in comparison with healthy volunteers; Clinical Drug Investigation, 11: 97-107.

22. Cao J., Sun J., Wang X., Li X. and Deng Y. (2009); N-trimethyl

chitosan-coated multivesicular liposomes for oxymatrine oral delivery; Drug Development and Industrial Pharmacy, 35 (11): 1339-1347.

23. Carla M., Jose L. C., Lima S. Reis A. L. and Margarida B. (2004);

Interaction of antiinflammatory drugs with EPC liposomes:calorimetric study in a broad concentration range; Biophysical Journal, 86: 946-954.

24. Charman C. J., Porter S., Mithani J. B. and Dressman J. B. (1997);

Physiochemical and physiological mechanisms for the effects of food on drug absorption: the role of lipids and pH; Journal of Pharmaceutical Sciences, 86: 269-282.

25. Cho H. A., Park I. S., Kim T. W., Oh Y. K., Yang K. S. and Kim J.

S. (2009); Suppression of hepatitis B virus-derived human hepatocellular carcinoma by NF-kappaB-inducing kinase-specific siRNA using liver-targeting liposomes; Archives of Pharmacal Research, 32 (7): 1077-1086.

26. Christopher J. H. P. and William N. C. (2001); In vitro assessment of

oral lipid based formulations; Advanced Drug Delivery Reviews, 50: S127-S147

27. Chuah A. M, Kuroiwa T, Ichikawa S, Kobayashi I, and Nakajima M

(2009) Formation of biocompatible nanoparticles via the self-assembly of chitosan and modified lecithin; Journal of Food Science, 74 (1): N1-N8

28. Chuasuwan B., Binjesoh V., Polli J. E., Zhang H., Amidon G. L.,

Junginger H. E., Midha K. K., Shah V. P., Stavchansky S., Dressman J. B. and Barends D. M. (2009); Biowaiver monographs for immediate release solid oral dosage forms: Diclofenac sodium and diclofenac potassium; Journal of Pharmaceutical Sciences, 98 (4): 1206-1219

Page 253: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

222

29. Clayton R., Ohagen A., Nicol F., Del Vecchio A. M., Jonckers T. H. and Goethals O. (2009); Sustained and specific in vitro inhibition of HIV-1 replication by a protease inhibitor encapsulated in gp120-targeted liposomes; Antiviral Research, 84 (2): 142-149.

30. Cocera M., Lopez O., Coderch L., Parra J.L. and de la Maza A.

(2003); Permeability investigations of phospholipid liposomes by adding cholesterol; Colloids and Surfaces A, 221: 9-17

31. Cosco D., Paolino D., Muzzalupo R., Celia C., Citraro R., Caponio

D., Picci N. and Fresta M. (2009); Novel PEG-coated niosomes based on bola-surfactant as drug carriers for 5-fluorouracil; Biomedical Microdevices, 11 (5): 1115-1125.

32. Cristina L., Maribel T, Srgio S. and Rogrio G. (1996); Steric

stabilization of nanoparticles: size and surface properties; International Journal of Pharmaceutics, 138: 1-12.

33. Cui T. L., Ying Z. Z., Hui S. G., Lai J.T., Zhi C. Z., Gang T. Z., Qin

Q. T., Zhuo J., Yan-Yan X., Pin T. H., Jing H., Liang W. and Xiao K. L (2010); Comparing encapsulation efficiency and ultrasound-triggered release for protein between phospholipid-based microbubbles and liposomes; Journal of Microencapsulation, 27 (2): 115-121.

34. Damiani P. C., Bearzoti M. and Cabezon M. A. (2008);

Spectrofluorometric determination of diclofenac sodium in tablets and ointments; Journal of Pharmaceutical and Biomedical Analysis, 20: 587-590.

35. Dan Marino (2003); Stealth liposomal technology: Current therapies

& future directions; Drug delivery, 3 (5): Article 1.

36. Danbo Y., Jiabi Z., Ying Z. and Liang G. (2006); Preparation, Characterization, and Pharmacokinetics of Sterically Stabilized Nimodipine-Containing Liposomes; Drug Development and Industrial Pharmacy, 32 (2): 219-227.

37. Dass C. R. (2008); Drug delivery in cancer using liposomes;

Methods in Molecular Biology, 437: 177-182.

38. Dasandi B., Shivaprakash., Saroj H. and Bhatt K. M. (2002); LC determination and pharmacokinetics of meloxicam; Journal of Pharmaceutical and Biomedical analysis, 28: 999-1004.

Page 254: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

223

39. Davies N. M. and Anderson K. E. (1997); Clinical pharmacokinetics of diclofenac: Therapeutic insights and pitfalls; Clinical Pharmacokinetics, 33 (3): 184-213.

40. de Leeuw J., de Vijlder H.C., Bjerring P. and Neumann H. A.

(2009); Liposomes in dermatology today; Journal of European Academy and Dermatology Venereology, 23 (5): 505-516.

41. Delgado V., Gonzalez C. F., Hunter R. J., Koopal L. K. and Lyklema

J. (2005); Measurement and Interpretation of electrokinetic phenomena: (IUPAC technical report); Journal of Pure and Applied Chemistry, 77 (10): 1753-1850.

42. Desormeaux A. and Bergeron M. G. (2005); Lymphoid tissue

targeting of anti-HIV drugs using liposomes; Methods in Enzymology, 391: 330-351.

43. Dey N. S., Majumdar S. and Rao M. (2008); Multiparticulate drug

delivery systems for controlled release; Tropical Journal of Pharmaceutical Research, 7 (3): 1067-1075

44. Dintaman J.M. and Silverman J.A. (1999); Inhibition of P-

glycoprotein by D-a-tocopheryl polyethylene glycol 1000 succinate (TPGS); Pharmaceutical Research, 16: 1550-1556.

45. Dubey V., Mishra D., Nahar M., Jain V. and Jain N. K. (2010);

Enhanced transdermal delivery of an anti-HIV agent via ethanolic liposomes; Nanomedicine, 6 (4): 590-596.

46. Ebtessam A. E., Michael C. B. and Brian W. B. (2002);

Iontophoretic estradiol skin delivery and tritium exchange in ultradeformable liposomes; International Journal of Pharmaceutics, 240 (1-2): 55-66.

47. Edward F. L. and Efraim R. (1989); Role of cholesterol in the

stability of pH-sensitive, large unilamellar liposomes prepared by the detergent-dialysis method; Biochim Biophys Acta, 981 (2): 254-260

48. Efstathios K., Ananta L. A., Rohan B., Ravi V. B. and Ananth V. A.

(2005); Preparation of in vivo cleavable agglomerated liposomes suitable for modulated pulmonary drug delivery; Journal of Controlled Release, 103:159-175.

49. Ehab R. B. and Mina I. T. (2007); Enhanced Transdermal Delivery

of Salbutamol Sulfate via Ethosomes; AAPS PharmSciTech, 8 (4): Article 107. DOI: 10.1208/pt0804107

Page 255: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

224

50. Elorza M. A., Elorza B. and Chantres J. R. (1997); Stability of

liposomal formulations: action of amphiphilic molecules; International Journal of Pharmaceutics, 158: 173-183.

51. Els Etten W. M., Marian T. K., Susan V. S., Irma A. J. M. and

Bakker W. (1998); Administration of liposomal agents and blood clearance capacity of the mononuclear phagocyte system; Antimicrobial agents and Chemotherapy, 42 (7): 1677-1681.

52. Emirhan N. and Sedef K. (2003); Method development and

validation for the analysis of meloxicam in tablets by CZE; Journal of Pharmaceutical and Biomedical Analysis, 31 (2): 393-400.

53. El-Samaligy M. S., Afifi N. N. and Mahmoud E. A. (2006);

Increasing bioavailability of silymarin using a buccal liposomal deliverysystem: Preparation and experimental design investigation; International Journal of Pharmaceutics, 308: 140-148.

54. European Agency for Evaluation of Medicinal Products (EMEA)

(2003); Committee for medicinal drug products. Diclofenac. Summary Report. EMEA-000885/03.

55. Farid S. M. H., Tasnim A., Nasira T. and Fozia H. (2005);

Pharmacokinetics of diclofenac sodium in normal man; Pakistan Journal of Pharmaceutical sciences, 18 (1): 18-24.

56. Federman N. and Denny C. T. (2010); Targetting liposomes towards

novel pediatric anti-cancer therapeutics; Pediatric Research, 67 (5): 514-519.

57. Filipovic G. J., Skalko B. N. and Jalsenjak I. (2001); Mucoadhesive

chitosan-coated liposomes: characteristics and stability; Journal of Microencapsulation, 18 (1): 3-12.

58. Fischer S., Foerg C., Ellenberger S., Merkle H.P. and Gander B.

(2006); One-step preparation of polyelectrolyte-coated PLGA microparticles and their functionalization with model ligands; Journal of Control Release, 111:135-144.

59. Franca P., Paola P., Ida G., Claudio M., Anna R., Ubaldo P., Laura

R. and Rosanna N. (2000); Boron-loaded liposomes in the treatment of hepatic metastases: Preliminary investigation by autoradiography analysis; Drug Delivery, 7 (2): 97-103.

Page 256: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

225

60. Francesca M., Maria L. G., Antonio M. R. and Paola M. (2006); Effect of preparation technique on the properties of liposomes encapsulating ketoprofen–cyclodextrin complexes aimed for transdermal delivery; International Journal of Pharmaceutics, 312: 53-60.

61. Garg M., Asthana A., Agashe H. B., Agrawal G. P. and Jain N K.

(2006); Stavudine-loaded mannosylated liposomes: in-vitro anti-HIV-I activity: Tissue distribution and pharmacokinetics; Journal of Pharmacy and Pharmacology, 58 (5): 605-616.

62. Gohel M.C. and Amin A. (1997); Development and evaluation of

lipospheres of diclofenac sodium; Indian Journal of Pharmaceutical Sciences, 59 (2): 85-88.

63. Gorka O., Rosa M., Alicia R. G. and Jose L. P. (2005); Micro and

nano drug delivery systems in cancer therapy; Cancer Therapy, l 3: 131-138.

64. Grace R. F., Edwards R. S., Mather L. E., Lin Y. and Power I.

(2000); Central and peripheral tissue distribution of diclofenac after subcutaneous injection in the rat; Inflammopharmacology, 8 (1): 43-54.

65. Grant G. J., Barenholz Y., Bolotin E., Bansinath M., Turndorf H.,

Piskoun B. and Davidson E. (2004); A novel liposomal bupivacaine formulation to produce ultra long-acting analgesia; Anesthesiology, 101: 133-137.

66. Gregg S. (1986); Statistical experimental design and its application

to pharmaceutical development problems; Drug Development and Industrial Pharmacy, 12 (8 & 9): 109-1123.

67. Gregoriadis G. (2007); Liposome technology: Entrapment of drugs

and other materials into liposomes: Volume II; Culinary and Hospitality Industry Publications Services, 424.

68. Gruber S.A., Venkataram S., Canafax D.M., Cipolle R.J., Bowers L.

and Elsberry D. (1989) Liposomal formulation eliminates acute toxicity and pump incompatibility of parenteral cyclosporine; Pharmaceutical Research, 6: 601-607.

69. Gun S.H. and Ismail R. (2001); Validation of a high performance

liquid chromatographic method for Tramadol and 0-desmethyl Tramadol in human plasma using solid phase extraction; Journal of Chromatography B, 759: 325-335.

Page 257: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

226

70. Gupta S. K., Bansal P., Bhardwaj R. K., Jaiswal J. and Velpandian T.

(2002); Comparison of analgesic and anti-Inflammatory activity of Meloxicam Gel with Diclofenac and Piroxicam Gels in Animal Models: pharmacokinetic parameters after topical application; Skin Pharmacolology and Applied Skin Physiology, 15: 105-111.

71. Gupta V., Gupta R., Grover R., Khanna R., Jangra V. and Mittal A.

(2008); Delivery of molecules to cancer cells using liposomes from bacterial cultures; Journal of Nanoscience and Nanotechnology, 8 (5): 2328-2333.

72. Gayatri D. S., Venkatesh P. and Udupa N. (2000); Niosomal

sumatriptan succinate for nasal administration; International Journal of Pharmaceutical Sciences, 62 (6): 479-481.

73. Hagalavadi N. S., Pragnesh B. P., Bapusaheb G. D., Purnima A. and

Sinnathambi A. (2007); Design and statistical optimization of glipizide loaded lipospheres using response surface methodology; Acta Pharmaceutica, 57 (3): 269-285.

74. Heiati H., Tawashi R. and Phillips N. C. (1998); Drug retention and

stability of solid lipid nanoparticles containing azidothymidine palmitate after autoclaving, storage and lyophilization; Journal of Microencapsulation, 15: 173-84.

75. Henk S. G., Birger L., Manuel R. D. and Erich B. (1997); A double-

blind, randomized trial to compare meloxicam 15 mg with Diclofenac 100 mg in the treatment of osteoarthritis of the knee; Osteoarthritis and Cartilage, 5: 283-288.

76. Hirai T., Matsomoto S. and Kishi I. (1997); Simultaneous analysis of

several non-steroidal anti inflammatory drugs in human urine by high performance liquid chromatography with normal solid phase extraction; Journal of Chromatography B, 692: 375-388.

77. Hongming C. and Robert L. (1998); Oral particulate delivery: status

and future trends; Advanced Drug Delivery Reviews, 34: 339-350.

78. Humberstone A.J. and Charman W.N. (1997); Lipid based vehicles for the oral delivery of poorly water soluble drugs; Advanced Drug Delivery Reviews, 25: 103-128.

79. ICH. (2005); Validation of Analytical Procedures (Text and

Methodology, Q2/R1): International conference on harmonization of

Page 258: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

227

technical requirements for registration of pharmaceuticals for human use: IFPMA, Geneva.

80. Immordino M.L., Brusa P., Rocco F., Arpicco S., Ceruti M., Cattel

L., Kokkona M., Kallinter, P., Fatouros D. and Antimisiarias S.G. (2002); Stability of SUV liposomes in the presence of cholate salts and pancreatic lipases: effect of lipid composition; European Journal of Pharmaceutical Sciences, 9: 245-252.

81. Ingebrigtsen L. and Brandl M. (2002); Determination of the Size

Distribution of Liposomes by SEC Fractionation, and PCS Analysis and Enzymatic Assay of Lipid Content; AAPS PharmSciTech, 2002; 3 (2): article 7.

82. Irene P., Bettini R. and Giordano F. (2007); Thermal behavior of diclofenac, diclofenac sodium and sodium bicarbonate compositions; Journal of Thermal Analysis and Calorimetry, 90 (3): 903-907

83. Jankowski W., Stefanik E. and Dec N. L. (2006); The simultaneous

determination of carbamazepine and diclofenac in biological samples; Annals of Medical Academy of Silesia, 60 (2). 130-33.

84. Jiraporn C. (2007); Nanosuspension technology for drug delivery;

Walailak Journal of Science and Technology, 4 (2): 139-153.

85. Jukui S., Yingjie D., Siling W., Jinna C., Xiaofei G. and Xiaodong D. (2010); Liposomes incorporating sodium deoxycholate for hexamethylmelamine (HMM) oral delivery: Development, characterization, and in vivo evaluation; Drug Delivery, 17 (3): 164-170

86. Kisoon N., Ariatti M. and Moodley T. (2002); A novel cationic

cholesterol derivative, its formulation into liposomes, and the efficient transfection of the transformed human cell lines HepG2 and HeLa; Drug Delivery, 9 (3): 161-167.

87. Kosarajua S. L., Cindy T. and Andrew L. (2006); Liposomal delivery

systems for encapsulation of ferrous sulfate: Preparation and characterization; Journal of Liposome Research, 16 (4): 347-358.

88. Knecht V., Risselada H. J., Mark A. E. and Marrink S. J. (2008).

Electrophoretic mobility does not always reflect the charge on an oil droplet; Journal of Colloid and Interface Science, 318: 477.

Page 259: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

228

89. Krieger M. L., Eckstein N., Schneider V., Koch M., Royer HD. and Jaehde U. (2010); Overcoming cisplatin resistance of ovarian cancer cells by targeted liposomes in vitro; International Journal of Pharmaceutics, 389 (1-2): 10-17.

90. Kshirsagar N. A. and Pandya S. K. (2005); Editorial Note. Journal of

post graduate medicine, 51: 3. 91. Kuhlmann O. and Krauss G. J. (1997); Crocheted ETFE-reactor for

on-line post-column photoderivatization of diclofenac in high-performance liquid chromatography; Journal of Pharmaceutical and Biomedical Analysis, 16: 553–559.

92. Kuhlmann O., Stoldt G., Gert H. and Krauss G. J. (1998);

Simultaneous determination of diclofenac and oxybuprocaine in human aqueous humor with HPLC and electrochemical detection; Journal of Pharmaceutical and Biomedical Analysis, 17: 1351-1356.

93. Ladbrooke B. D., Williams R. M. and Chapman D. (1968); Studies

of lecithin cholesterol water interactions by DSC and x-ray diffraction; Biochimica and Biophysica Acta; 150: 333-340.

94. Lala L.G., Mello P.M. and Naik S.R. (2002); HP-TLC determination of diclofenac sodium from serum; Journal of Pharmaceutical and Biomedical Analysis, 29: 539-544.

95. Lars G. B. (2009); Two new techniques for sample preparation in

bioanalysis: Microextraction in packed sorbent (MEPS) and use of a bonded monolith as sorbent for sample preparation in polypropylene tips for 96-well plates; Analytical and Bioanalytical Chemistry, 393(3): 797-807.

96. Lasic D. D., Vallner J. J. and Working P. K. (1999); Sterically

stabilized liposomes in cancer therapy and gene delivery; Current Opinion in Molecular Therapeutics, 1 (2): 177-85.

97. Laurence L. B. (2007); Goodman & Gilman's; the Pharmacological

Basis of Therapeutics, 11: 451.

98. Law S.L. and Hung H.Y. (1998); Properties of acyclovir-containing liposomes for potential ocular Delivery: Note; International Journal of Pharmaceutics, 161: 253-259.

99. Lee H. S., Jeong C. K., Choi S. J., Kim S. B., Lee M. H., Ko G. I.

and Sohn D. H. (2000); Simultaneous determination of aceclofenac and diclofenac in human plasma by narrowbore HPLC using

Page 260: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

229

column-switching; Journal of Pharmaceutical and Biomedical Analysis, 23 (5): 775-781.

100. Li F., Sun J. Y., Wang J. Y., Du S. L., Lu W.Y. and Liu M. (2008);

Effect of hepatocyte growth factor encapsulated in targeted liposomes on liver cirrhosis; Journal of Controlled Release, 131 (1): 77-82.

101. Lill J.S., Sullivan T. O., Bauer LA., Horn JR ., Carithers J. R.,

Strandness DE., Lau H., Chan K. and Thakker K. (2000); Pharmacokinetics of diclofenac sodium in chronic active hepatitis and alcoholic cirrhosis; Journal of Clinical Pharmacology, 40: 250-257.

102. Limian Z. and Hian K. L. (2001); Application of static liquid-phase

micro extraction to the analysis of organochlorine pesticides in water; Journal of Chromatography A, 919 (2): 381-388

103. Lissy M., Scallion R., Stiff D. and Mopre K. (2010);

Pharmacokinetics of an oral diclofenac potassium liquid filled soft gelatin capsules with a diclofenac potassium tablets; Expert Opinion on Pharmacotherapy, 11 (5): 701-708.

104. Liu Y., Lu W. L., Guo J., Du J., Li T. and Wu J. W. (2008); A

potential target associated with both cancer and cancer stem cells: a combination therapy for eradication of breast cancer using vinorelbine stealthy liposomes plus parthenolide stealthy liposomes; Journal of Controlled Release, 129 (1): 18-25.

105. Lopes, D.H., Meister, A., Gohlke, A., Hauser, A., Blume, A., and

Winter, R. (2007); Mechanism of islet amyloid polypeptide fibrillation at lipid interfaces studied by infrared reflection absorption spectroscopy. Biophysical Journal, 93: 3132–3141.

106. Luciana B. L., Maria V. S., Newton L. P., Luana C. O. and Anselmo

G. O. (2006); Interaction of sodium diclofenac with freeze-dried soya phosphatidylcholine and unilamellar liposomes; Revista Brasileira de Ciencias Farmaceuticas, 42 (4): 497-504.

107. Lyle G. S., Zhaolin W., Raimar L., Jonathan P. W., Carlos F. L. and

Warren H. F. (2005); Spray-freeze-dried liposomal ciprofloxacin powder for inhaled aerosol drug delivery:Note; International Journal of Pharmaceutics, 305: 180-185.

108. Maha N., Samar M., Nahed D. Mortada. and Elshamy A. A. (2008);

Vesicular aceclofenac systems: A comparative study between

Page 261: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

230

liposomes and niosomes; Journal of Microencapsulation, 25 (7): 499-512.

109. Mehrdad H. and Hosnieh T. (2003); Carrier erythrocytes: An

overview; Drug Delivery; 10 (1): 9-20. 110. Mahmood A., Muhammad I. and Ghulam M. (2009); Comparison of

bioavailability and pharmacokinetics of diclofenac sodium and diclofenac potassium in normal and dehydrated rabbits; Acta Pharmaceutica Sinica, 44 (1-3): 80-84.

111. Malam Y., Loizidou M. and Seifalian A. M. (2009); Liposomes and

nanoparticles: nanosized vehicles for drug delivery in cancer; Trends in Pharmacological Sciences, 30 (11): 592-9.

112. Malliou E. T.

113. Manconi M., Isola R., Falchi A. M., Sinico C. and Fadda A.M.

(2007); Intracellular distribution of fluorescent probes delivered by vesicles of different lipidic composition; Colloids Surface B: Biointerfaces, 57: 143

114. Maria L. M., Marco Z., Guido E., Donatella V., Chiara S., Giuseppe

L. and Anna M. F. (2005); Diclofenac-β-cyclodextrin binary systems: physicochemical characterization and in-vitro dissolution and diffusion studies; AAPS Pharm Sci Tech, 6 (3): E464-E472.

115. Maria R. L. R., Gilberto C. H., and Mario I. O. (2008);

Pharmacokinetics and pharmacodynamics of diclofenac in the presence and absence of glibenclamide in the rat; Journal of Pharmacy and Pharmaceutical Sciences 11 (3): 68-76.

116. Maria L. I., Franco D. and Luigi C. (2006); Stealth liposomes:

review of the basic science, rationale, and clinical applications, existing and potential; International Journal of Nanomedicine, 1 (3): 297-315.

117. Martin C. W. (1995); Sterically stabilized liposome therapeutics;

Advanced Drug Delivery Reviews, 16: 249-265.

118. Martin W., Kohei H. and Hirofumi (2009); Development and in-vitro characterization of liposomes coated with thiolated poly (acrylic acid) for oral drug delivery; Drug Development and Industrial Pharmacy, 35: 209-215.

Page 262: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

231

119. Marty C., Meylan C., Schott H., Ballmer-Hofer K. and Schwendener R. A. (2004); Enhanced heparan sulfate proteoglycan-mediated uptake of cell-penetrating peptide-modified liposomes; Cellular and Molecular Life Sciences, 61: 1785–1794.

120. Maruyama K., Torchilin B. and Huang L. (1991); Activity of

amphipathic poly(ethylene glycol) 5000 to prolong the circulation time of liposomes depends on the liposome size and is unfavorable for immunoliposome binding to target; Biochimica Biophysica Acta, 1062:142-148.

121. Marzo A., Dal B. L., Verga F., Ceppi M. N., Abbondati G., Aleotti

T. R., Crivelli F., Uhr M. R. and Ismaili S. (2000); Pharmacokinetics of diclofenac after oral administration of its potassium salt in sachet and tablet formulations; Arzneimittel-Forschung, 50 (1): 43-47

122. Masahiro Y., Kyoko T., Masayuki A., Yoichi U., Masashi N. and

Noboru A. (2007); Release of drugs from liposomes varies with particle size; Biological and Pharmaceutical Bulletin, 30 (5): 963-966.

123. Mayer L. D., Tai L. C. L., Ko D. S. C., Masin D., Ginsberg R. S.,

Cullis P. R. and Bally M. B. (1989); Influence of vesicle size, lipid composition, and drug-to-lipid ratio on the biological activity of liposomal doxorubicin in mice; Cancer Research, 49: 5922-5930.

124. Mayer X., Namiranian K., dehghanyar P., Stroh R., Mascher H. and

Muller M. (2003); Comparison of UV and tandem mass spectrometric detection for the high-performance liquid chromatographic determination of diclofenac in microdialysis samples; Journal of Pharmaceutical and Biomedical Analysis, 33: 745-754.

125. Mazurek S. and Szostak R. (2006); Quantitative determination of

diclofenac sodium and aminophylline ininjection solutions by FT-Raman spectroscopy; Journal of Pharmaceutical and Biomedical Analysis, 40: 1235-1242.

126. Mehnert W. and Mader K. (2001): Solid lipid nanoparticles:

production, characterization and applications; Advance Drug Delivery Reviews, 47:165-96.

127. Michele T., Elena P., Maria E. C. and Marina G. (2004); Deformable

liposomes for dermal administration of methotrexate; International Journal of Pharmaceutics, 270 (1-2): 119-125

Page 263: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

232

128. Mihaela T., Anca R., Jeremy H. B. and James M. B. (2007); Designing Lipid Nanostructures for Local Delivery of Biologically Active Macromolecules; Journal of Liposome Research, 17 (3-4): 237-248.

129. Millership J. S., Hare L. G., Farry M., Collier P. S., McElnay J. C.,

Shields M. D. and Carson D. J. (2001); The use of hydrophilic lipophilic balanced (HLB) copolymer SPE cartridges for the extraction of diclofenac from small volume paediatric plasma samples; Journal of Pharmaceutical and Biomedical Analysis, 25: 871–879.

130. Minghui F., Shiying X., Shuqin X. and Xiaoming Z.

(2008);Preparation of salidroside nano-liposomes by ethanol injection method and in vitro release study; European Food Research and Technology, 227:167-174.

131. Mishra P. K., Gulbake A., Jain A., Vyas S. P. and Jain S. K. (2009);

Targeted delivery of an anti-cancer agent via steroid coupled liposomes; Drug Delivery, 16 (8): 437-447.

132. Moein M. S., Christy A. H. and Clifford J. M. (2001); Long-

circulating and target-specific nanoparticles: Theory to practice; Pharmacological Reviews, 53: 283-318.

133. Mohammed E., Hamid A., Novotny L. and Hamza H. (2001);

Determination of diclofenac sodium, flufenamic acid,indomethacin and ketoprofen by LC-APCI-MS; Journal of Pharmaceutical and Biomedical Analysis, 24: 587-594.

134. Mohsin A. H. (2007); Basic pharmacokinetics; Taylor and Francos

Group, NY, 978-1-4200-4671-4677.

135. Montanari J., Perez A. P., Di Salvo F., Diz V., Barnadas R., Dicelio L., Doctorovich F., Morilla M. J. and Romero E. L (2007); Photodynamic ultradeformable liposomes: Design and characterization; International Journal of Pharmaceutics, 330: 183-194.

136. Moore J.W. and Flanner H. H., (1996); Mathematical comparison of

curves with an emphasis on in-vitro dissolution profiles. Pharm Tech, 20 (6): 64-74.

137. Moreira J. N., Almeida L. M. and Geraldes C. F. (1997); Carboplatin

liposomes coated with O-palmitoylpullulan: in vitro characterization; International Journal of Pharmaceutics, 147: 153-164.

Page 264: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

233

138. Mukesh C. G., Krishnakant G., Sarvaiya N., Mehta R., Chirag D.,

Soni V., Vyas U. and Rikita K. D. (2005); Assessment of similarity factor using different weighting approaches; Dissolution Technologies, (9): 23-27.

139. Myers R.A. and Stella V.J. (1992); Systemic bioavailability of

penclomedine from oil-in-water emulsions administered intraduodenally to rats; International Journal of Pharmaceutics, 78: 217-226.

140. Nagayasu A., Uchiyama K. and Kiwada H. (1999); The size of

liposomes: a factor which affects their targeting efficiency to tumors and therapeutic activity of liposomal antitumor drugs; Advanced Drug Delivery Reviews, 40 (1-2): 75-87.

141. Nande V. S. , Umesh U. B., Dinesh M. M., Arun T. P. and

Siddheshwar B. J. (2007); Sustained Release Microspheres of Diclofenac Sodium Using PEGylated Rosin Derivatives; Drug Development and Industrial Pharmacy, 33 (10): 1090-1100.

142. Naoto O., Yukihiro N., Atsushi T. and Shoji O. (1993); Tumor

imaging with technetium-99m-DTPA encapsulated in RES-avoiding liposomes; Nuclear Medicine and Biology, 20 (4): 407-412.

143. Naveed A., Mahmood A., Haji M. S. K., Jawad A., Gulfishan.,

Ahmad M. and Muhammad U. (2010); Formulation and characterization of a multiple emulsion containing 1% L-ascorbic acid; Bulletin of Chemical Society of Ethiopia. 24 (1): 1-10.

144. Ney P. (1973); Zeta potentiale und flotierbarkeit von mineralien.

New York: Springer.

145. Noppamas R., Panawan T. and Boonyium K. (2005); Comparative bioavailability study of two meloxicam preparations; Chaiang Mai Medical Bulletin, 44 (3): 91-100.

146. Omaima N., El-Gazayerly and Ahmed H. H. (1997); Preparation and

evaluation of acetazolamide liposomes as an ocular delivery system; International Journal of Pharmaceutics, 158 (2): 121-127.

147. Owais M., Varshney G. C., Choudhury A., Chandra S. and Gupta C.

M. (1995); Chloroquine encapsulated in malaria-infected erythrocyte-specific antibody-bearing liposomes effectively controls chloroquine-resistant Plasmodium berghei infections in mice; Antimicrobial Agents and Chemotherapy, 39 (1):180-184.

Page 265: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

234

148. Panusa A., Multari G. and Incarnato L. G. (2007); High performance

liquid chromatography analysis of anti inflammatory pharmaceuticals with UV detection and electron spray-mass spectrometry detection in suspected counterfeit homeopathic medicinal products; Journal of Pharmaceutical and Biomedical Analysis, 43: 1221-1227.

149. Paola C., Maurizio C., Paola B., Silvia A., Franco D., Luigi C.

(2000); Preparation, characterization and properties of sterically stabilized paclitaxel-containing liposomes; Journal of Controlled Release, 63:19-30.

150. Parampal D. and Khuller G. K. (1997); Lung specific stealth

liposomes: stability, biodistribution and toxicity of liposomal antitubercular drugs in mice; Biochimica et Biophysica Acta, 1334: 161-172

151. Patel V. B. and Misra A. N. (1999); Encapsulation and stability o f

Clofazimine liposomes; Journal of Microencapsulation, 16 (3): 357-367.

152. Paz P. D., Esteso M. C., Alvarez M., Mata M., Chamorro C. A. and

Anel L. (2010); Development of extender based on soybean lecithin for its application in liquid ram semen; Theriogenology, 74 (4): 663-671

153. Pejcic Z., Pokrajac M., and Jezdimirovi M. (2006); Pharmacokinetics

of diclofenac in pigs after intramuscular administration of a single dose; Acta Veterinaria, 56 (4): 323-331.

154. Perrot S., Krause D., Crozes P. and Naim N. (2006); Efficacy and

tolerability of paracetamol/tramadol (325 mg/ 37.5 mg) combination treatment compared with tramadol (50 mg) monotherapy in patients with subacute low back pain; Clinical Therapeutics, 28 (10): 1592-1606.

155. Pignatello R., Ferro M. and Puglisi G. (2002); Preparation of solid

dispersions of nonsteroidal anti-inflammatory drugs with acrylic polymers and studies on mechanisms of drug-polymer interactions; AAPS Pharm Sci Tech, 3 (2): 1-11(Article 10).

156. Pirson P., Steiger R.F., Trouet A., Gillet J. and Herman F. (1980);

Primaquine liposomes in the chemotherapy of experimental murine malaria; Annals of Tropical Medicine Parasitology, 74 (4): 383-391.

Page 266: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

235

157. Pleumchitt R., Narong S., Korakot C. and Krisana K. (2003); Physicochemical properties of amphotericin B liposomes prepared by reverse-phase evaporation method; Drug Development and Industrial Pharmacy, 29 (1): 31-37.

158. Porter, S and Dressman J.B. (2001); Influence of physicochemical

properties on dissolution of drugs in the gastrointestinal tract; Advance Drug Delivery Reviews, 46: 75-87.

159. Raffa R. B. (2001); Pharmacology of oral combination analgesics:

rational therapy for pain; Journal of Clinical Pharmacy and Therapeutics, 26 (4): 257-264.

160. Ralph S. and Keith A. M. (2009); Effect of food intake on the

pharmacokinetics of diclofenac potassium soft gelatin capsules: A single dose, randomized, two way cross-over study; Clinical Therapeutics, 31 (10): 2233-2241.

161. Rana S., Zuzan H., Moustapha H. and Mohamed A. R. (2008); Rapid

and sensitive method for determination of cyclophosphamide in patients plasma samples utilizing microextraction by packed sorbent online with liquid chromatography-tandem mass spectrometry (MEPS-LC-MS/MS); Journal of Liquid Chromatography & Related Technologies, 31 (5): 683-694.

162. Raymond C. R., Paul J. S. and Sian C. O. (2006); Handbook of

pharmaceutical excipients; Volume 5, Published by the Pharmaceutical Press.

163. Report. (2001); Food Allergy and Anaphylaxis Network. Adapted

from Food Allergy News; Vol. 2, No. 1.

164. Roopa P. S. and Kusum D. V. (2009); Lamivudine liposomes for transdermal delivery: Formulation, characterization, stability and in-vitro evaluation; International Journal of Pharmaceutical Sciences and Nanotechnology, 1 (4): 317-326.

165. Rousset X., Vaisman B., Auerbach B., Krause B. R., Homan R.,

Stonik J., Csako G., Shamburek R. and Remaley A. T. (2010); Effect of recombinant human lecithin cholesterol:acyltransferase infusion on lipoprotein metabolism in mice; Journal of Pharmacology and Experimental Therapeutics, 110.169540: 1-26.

166. Sajeev C., Vinay G., Archna R. and Saha R. N. (2002); Oral

controlled release formulation of diclofenac sodium by

Page 267: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

236

microencapsulation with ethyl cellulose; Journal of Microencapsulation 19 (6): 753-760.

167. Sakae U., Kazuo M., Osamu I., Akinori S., Jun-ichi H. and Motoharu

I. (1995); Enhanced tumor targeting and improved antitumor activity doxorubicin by long-circulating liposomes containing amphipathic poly (ethylene glycol); International Journal of Pharmaceutics, 126: 41-48

168. Sanjay R. K., Karthikeyan C., Moorthy N. S. and Trivedi P. (2006);

New spectrophotometric methods applied to the simultaneous determination of diclofenac potassium and tizanidine; Indian journal of pharmaceutical sciences, 68 (3): 317-322.

169. Saranadasa H. (2005); A multivariate test for similarity of two

dissolution profiles; Journal of Biopharmaceutical Statistics, 15: 265-278.

170. Sato A., Takagi M., Shimamoto A., Kawakami S. and Hashida M.

(2007); Small interfering RNA delivery to the liver by intravenous administration of galactosylated cationic liposomes in mice; Biomaterials, 28 (7): 1434-1442.

171. Sato Y., Murase K., Kato J., Kobune M., Sato T. and Kawano Y.

(2008); Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone; Nature Biotechnology, 26 (4): 431-442.

172. Satoshi I., Kei N., Shinji S. and Mitsutoshi N. (2002); Preparation of

gelatin microbeads with a narrow size distribution using microchannel emulsification; AAPS PharmSciTech, 3 (3): Article 25.

173. Schmid M. H. and Korting H.C. (1994); Liposomes: a drug carrier

system for topical treatment in dermatology; Critical Reviews Therapeutics and Drug Carrier System, 11 (2-3): 97-118.

174. Schmidt K. H. (1986); Liposomes as drug carriers; Verlag, Stuttgart

and New York.

175. Semalty A., Semalty M. Rawat B. S. Singh D. and Rawat M. S. M. (2009); Pharmacosomes: The lipid-based new drug delivery system; Expert Opinion on Drug Delivery, 6 (6): 599-612

176. Shamsipur M., Jalali F. and Ershad S. (2005); Preparation of a

diclofenac potentiometric sensor and its application to

Page 268: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

237

pharmaceutical analysis and to drug recovery from biological fluids; Journal of Pharmaceutical and Biomedical Analysis, 37: 943-47.

177. Shargel L., Susanna W. P. and Yu A. B. C. (2005); Applied

biopharmaceutics and pharmacokinetics; 5th edition, Appleton-Century-Crofts. East Norwalk, CT ISBN 0-8385-0129-X.

178. Sharon N. L., Enrico M., Nurzalina., Karim A. K. and Yuen K. H.

(2006); Enhanced oral bioavailability and intestinal lymphatic transport of a hydrophilic drug using liposomes; Drug Development and Industrial Pharmacy, 32: 335-345.

179. Shivhare U. D., Ambulkar D. U., Mathur V. B., Bhusari K. P. and

Godbole M. D. (2009); Formulation and evaluation of pentoxifylline liposome formulation; Digest journal of Nanomaterials and Biostructures, 4 (4): 857-862.

180. Shubha R., Swati G., Rajeshwari R., Bapu C., Manish N. and Harish

P. (2004); Determination of oral meloxicam pharmacokinetic parameters in Asian Indians: Comparison with a German population; Saudi Pharmaceutical Journal, 12 (4): 144-149

181. Siewert M., Dressman J., Brown C. K. and Shah V. P. (2003);

FIP/AAPS Guidelines to Dissolution/In-Vitro Release Testing of Novel/Special Dosage Forms; AAPS Pharm Sci Tech, 4 (1): 43-52.

182. Sihorkar V. and Vyas S. P. (2001); Potential of polysaccharide

anchored liposomes in drug delivery, targeting and immunization; Journal of Pharmacy and Pharmaceutical Sciences, 4 (2): 138-158.

183. Silvana E. V., Patricia M. C. and Teodoro S. K. (2008); Method

development and validation for the simultaneous determination of meloxicam and pridinol mesylate using RP-HPLC and its application in drug formulations; Journal of Pharmaceutical and Biomedical Analysis, 46 (2): 219-225

184. Singla N., Pong A. and Newman K. (2005); Combination oxycodone

5 mg/ibuprofen 400 mg for the treatment of pain after abdominal or pelvic surgery in women; Clinical Therapeutics, 27 (1): 45-57.

185. Sipos., Maria S., Andras S., Istvan E. and Piroska S. (2008); An

assessment of the interactions between diclofenac sodium and ammonio methacrylate copolymer using thermal analysis and raman spectroscopy; Journal of Pharmaceutical and Biomedical Analysis, 46: 288-294.

Page 269: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

238

186. Sivakumar P.A. and Panduranga R. K. (2003); The use of cholesteryl pullulan for the preparation of stable vincristine liposomes; Carbohydrate Polymers, 51: 327-332.

187. Soledad M. G., Isabel M. A., Concepcio N., Pedren S. and Molina J.

(1998); Flow-injection spectrophotometric determination of diclofenac sodium in pharmaceuticals and urine samples. Journal of Pharmaceutical and Biomedical Analysis, 17: 267-273.

188. Srinivasa N. R. and Jennifer A. H. (2005); Combination therapy for

neuropathic pain; New England Journal of Medicine, 352 (11): 1373-1375.

189. Sriram V. and Rhodes C. T. (1995) Preparation and characterization

of liposomes as therapeutic delivery systems: A review. Pharmaceutica Acta Helvetiae, 70: 95-111.

190. Sun J., Yingjie D., Siling W., Jinna C., Xiaofei G. and Xiaodong D.

(2010); Liposomes incorporating sodium deoxycholate forhexamethyl melamine (HMM) oral delivery: Development, characterization, and in vivo evaluation; Drug Delivery, 17 (3): 164-170.

191. Sun Y., Takaba K., Kido H., Nakashima M. N. and NakaShima K.

(2003); Simultaneous determination of arylpropionic acid non-steroidal anti-inflammatory drugs in pharmaceutical formulations and human plasma by HPLC with UV detection; Journal of Pharmaceutical and Biomedical Analysis, 30: 1611-1619.

192. Sunita D. and Lalit T. (2008); Preparation and evaluation of

oxytetracycline hydrochloride microbeads for delayed release; Pakistan Journal of Pharmaceutical Sciences, 21 (2): 103-108

193. Svetlana G., Kevin K., Michael D. I. and Leonid H. (2005); The

potential advantages of nanoparticles drug delivery systems in chemotherapy of tuberculosis; American Journal of Respiratory and Critical Care Medicine, 172: 1487-1490.

194. Sweetman S. C. (2009); Martindale, the Complete Drug Reference,

36th Edition., Pharmaceutical Press, London, U.K; 44-47.

195. Szoka F. C. and Papahadjopoulos D. (1978); Procedure for preparation of liposomes with large internal aqueous space and high capture by reverse-phase evaporation; Proceeding of National Academy of Science USA, 75 (9): 4195-4198.

Page 270: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

239

196. Taburet S. A. M., Singlas E. T., Glass R. C., Thomas F. M. and Leutenegger E. M. (1995); Pharmacokinetic comparison of oral and local action transcutaneous Flurbiprofen in healthy volunteers; Journal of Clinical Pharmacology and Therapeutics, 20 (2): 101-107.

197. Takeuchi H., Matsui Y., Yamamoto H. and Kawashima Y. (2003);

Mucoadhesive properties of carbopol or chitosan-coated liposomes and their effectiveness in the oral administration of calcitonin to rats; Journal of Controlled Release, 86: 235-242.

198. Tamer S., Kenichi O., Kazutaka H. and Toshikiro K. (2008);

Prolongation of residence time of liposome by surface-modification with mixture of hydrophilic polymers; International Journal of Pharmaceutics, 359: 272-279.

199. Tanveer E. T. and Marc C. H. (1997); A Systematic Review of

randomized controlled trials of pharmacological therapy in osteoarthritis of the knee, with an emphasis on trial methodology; Seminars in Arthritis and Rheumatism, 26 (5): 755-770.

200. Taylor T. M., Sylvia G., Michael D. P., Barry D. B. and Jochen W.

(2007); Characterization of Antimicrobial-bearing Liposomes by ζ-Potential, Vesicle Size, and Encapsulation Efficiency; Food Biophysics, 2 (1): 1-9.

201. Tomoko N. and Fumiyoshi I. (2005); Encapsulation efficiency of

water-soluble and insoluble drugs in liposomes prepared by the microencapsulation vesicle method; International Journal of Pharmaceutics, 298: 198-205.

202. Tomoko N., Akira T., Kiminori M. and Fumiyoshi I. (2002); Factors

affecting physicochemical properties of liposomes prepared with hydrogenated purified egg yolk lecithins by the microencapsulation vesicle method; Colloids and Surfaces B: Biointerfaces, 27: 323-332.

203. Theresa M. A. and Elaine H. M. (1996); Therapeutic opportunities

for targeted liposomal drug delivery; Advanced Drug Delivery Reviews, 21 (2): 117-133.

204. Tiwari S., Goyal A. K., Mishra N., Khatri K., Vaidya B. and Mehta

A. (2009); Development and characterization of novel carrier gel core liposomes based transmission blocking malaria vaccine; Journal of Controlled Release, 140 (2): 157-165

205. Torchilin V. (2008); Antibody-modified liposomes for cancer

chemotherapy; Expert Opinion Drug Delivery, 5 (9):1003-2105.

Page 271: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

240

206. Todd P. A. and Sorkin E. M. (1988); Diclofenac sodium: A

reappraisal of its pharmacodynamics, pharmacokinetic properties and therapeutic efficacy; Drugs, 35, 244-285.

207. Tsotas V. A., Mourtas, S. and Antimisiaris S. (2007);

Dexamethasone Incorporating Liposomes: Effect of Lipid Composition on Drug Trapping Efficiency and Vesicle Stability; Drug Delivery, 14 (7): 441-445.

208. Türck D., Busch U., Heinzel G., Narjes H. and Nehmiz G. (1995);

Effect of food on the pharmacokinetics of meloxicam after oral administration; Clinical Drug Investigation, 9: 270-276.

209. Ueoka R., Komizu Y., Matsumoto Y., Zhong Y., Tanaka R. and

Yamamoto N. (2008); Selective inhibitory effects of hybrid liposomes on the growth of HIV type 1-infected cells in vitro; Bioorganic and Medicinal Chemistry Letters, 18 (16): 4578-4580.

210. Ulrich S. and James L. W. (1997); Erythrocytes as drug carriers in

medicine; Springer, Plenum Press NY, ISBN: 0306-45599-4. 211. Ulrich A.S. (2002); Biophysical Aspects of Using Liposomes as

Delivery Vehicles; Bioscience Reports, 22 (2): 129-150.

212. USP30-NF25 (2009); The official compendium of standards; the united states pharmacopoeial convention.

213. Valerie C. and Patrick V. (2008); Physico-chemical properties and

cytotoxicity assessment of PEG-modified liposomes containing human hemoglobin. Colloids and Surfaces B: Biointerfaces, 65 239-246.

214. Valat J. P., Accardo S., Reginster J-Y., Wouters M., Hettich M. and

Lieu P-L. (2001); A comparison of the efficacy and tolerability of meloxicam and diclofenac in the treatment of patients with osteoarthritis of the lumbar spine; Inflammatory Research, 50 (1): S30–S34.

215. Velpandian T., Jaiswal J., Bhardwaj R. K. and Gupta S. K. (2000);

Development and validation of a new high performance liquid chromatographic estimation method of Meloxicam in biological samples; Journal of Chromatography B, 738: 431-6.

216. Venkateswarlu V. (2006); Biopharmaceutics and pharmacokinetics;

Pharma Book Syndicate Hyderabad, ISBN 081-88449-05-9.

Page 272: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

241

217. Verma J. N., Wassef N. M., Wirtz R. A., Atkinson C.T., Aikawa M.

and Loomis L. D. (1991); Phagocytosis of liposomes by macrophages: intracellular fate of liposomal malaria antigen; Biochim Biophys Acta, 1066 (2): 229-238.

218. Vringer de T. and Ronde de H.A.G. (1995); Preparation and

structure of a water-in-oil cream containing lipid nanoparticles; Journal of Pharmaceutical Sciences, 84: 466.

219. Wagner E. J., Krugner H. L. and Heath T. D. (2009); Liposome

dependent delivery of S-adenosyl methionine to cells by liposomes: a potential treatment for liver disease; Journal of Pharmaceutical Sciences, 98 (2): 573-582.

220. Wang Q., Wenhai H. and Deping W. (2007); Preparation of hollow

porous HAP microspheres as drug delivery vehicles; Journal of Wuhan University of Technology, 22 (1): 174-177.

221. Wang S., Xu H., Xu J., Zhang Y., Liu Y. and Deng Y. H. (2010); Sustained liver targeting and improved antiproliferative effect of doxorubicin liposomes modified with galactosylated lipid and PEG-lipid; AAPS Pharm Sci Tech, 11 (2): 870-877.

222. Wen-Hsien T., Hung-Yi C., Ho-Hsien C., Joh-Jong H., Hwi-Chang

C., Shou-Hsun C. and Tzou-Chi H. (2009); Application of dispersive liquid–liquid microextraction and dispersive micro-solid-phase extraction for the determination of quinolones in swine muscle by high-performance liquid chromatography with diode-array detection; Analytica Chimica Acta, 656 (1-2): 56-62.

223. Wenzel J., Zeisig R. and Fichtner I. (2010); Inhibition of metastasis

in a murine 4T1 breast cancer model by liposomes preventing tumor cell-platelet interactions; Clinical and Experimental Metastasis, 27 (1): 25-34.

224. Wenzel J., Zeisig R., Haider W., Habedank S. and Fichtner I. (2009);

Inhibition of pulmonary metastasis in a human MT3 breast cancer xenograft model by dual liposomes preventing intravasal fibrin clot formation; Breast Cancer Research Treatment, 121(1):13-22.

225. Werle M. and Takeuchi H. (2009); Chitosan-aprotinin coated

liposomes for oral peptide delivery: Development, characterisation and in vivo evaluation; International Journal of Pharmaceutics, 370 (1-2): 26-32.

Page 273: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

242

226. Westerhof W. (1885); Possibilities of liposomes as dynamic dosage

form in dermatology. Medical Hypotheses, 16 (3): 283-8.

227. Whittinghill J. M., Norton J. and Prctor A. (2000); Stability Determination of Soya Lecithin Based Emulsions by Fourier Transform Infrared Spectroscopy; Journal of American Oil Chemical Society, 77 (1): 37-42.

228. William B. L., David R. K., Brandon V. S. and Nicholas A. P.

(2010); Polymers for Drug Delivery Systems; Annual Review of Chemical and Biomolecular Engineering, 1: 149-173.

229. Wim H. D. J. and Paul J. A. (2008); Drug delivery and nanoparticles:

Applications and hazards; International Journal of Nanomedicine, 3 (2): 133-149.

230. Xu H. Y., Zhong D., Zhao L., Zhang Y. and Zhang B. (2001);

Pharmacokinetics of meloxicam in healthy Chinese volunteers. Acta Pharmaceutica Sinica 36 (1): 71-3.

231. Yang D., Jiabi Z., Ying Z. and Liang G. (2006); Preparation,

Characterization, and Pharmacokinetics of Sterically Stabilized Nimodipine-Containing Liposomes; Drug Development and Industrial Pharmacy, 32: 219-227.

232. Yanyu X., Yunmei S., Zhipeng C., Qineng P. (2006); The

preparation of silybin phospholipid complex and the study on its pharmacokinetics in rats; International Journal of Pharmaceutics, 307: 77-82.

233. Yeagle P. L., Albert A. D., Boesze-Battaglia K., Young J. and Frye

J. (1990); Cholesterol dynamics in membranes; Biophysical Journal, 57 (3): 413–424

234. Young H. J., Lee H. W., Kim Y. H., Jeong D. W. and Lee H. S.

(2005); Simultaneous determination of piroxicam, meloxicam and tenoxicam in human plasma by liquid chromatography with tandem mass spectrometry; Journal of Chromatography B, 826 (1-2): 214-219.

235. Yarosh D. B. (2001); Liposomes in investigative dermatology;

Photodermatology Photoimmunology and Photomedicine, 17 (5): 203-12.

Page 274: Formulation and In-vitro/In-vivo Evaluation of Oral ... · Mr. M. Ali, Mr. Rizwan, Mr. Ibrahim shah and Munawar Ali for their constant prayers, help, co-operation and entertainment

243

236. Zaru M., Manca M. L., Fadda A. M. and Antimisiaris S. G. (2009); Chitosan-coated liposomes for delivery to lungs by nebulization; Colloids Surface B: Biointerfaces, 71 (1): 88-95.

237. Zaru M., Mourtas S., Klepetsanis P., Fadda A.M. and Antimisiaris S.

G. (2007); Liposomes for drug delivery to the lungs by nebulization; European Journal of Pharmaceutics and Biopharmaceutics, 67 (3): 655-66.

238. Zawilla N. H., Abdul-Azim M. M., El kousy N. M. and El-Moghazy

Aly S. M. (2003); Determination of meloxicam in bulk and pharmaceutical formulations; Journal of Pharmaceutical and Biomedical Analysis, 32 (6): 1135-1144

239. Zecheru T. , Zaharia C., Mabilleau G., Chappard D. and Cincu C.

(2006); New hema-based polymeric microbeads for drug delivery systems; Journal of Optoelectronics and Advanced Materials, 8 (3): 1312 - 1316

240. Zeljka P., Nataša Š. B., Jelena F. G., Anita M. and Ivan J. (2005);

Development and in vitro evaluation of a liposomal vaginal delivery system for acyclovir; Journal of Controlled Release, 106 (1-2): 34-43

241. Zhang L. J., Xing B., Wu J., Xu B. and Fang X. L. (2008);

Biodistribution in mice and severity of damage in rat lungs following pulmonary delivery of 9-nitro camptothecin liposomes; Pulmonary Pharmacology and Therapeutics 21 (1): 239-46.

242. Zhiwen Y., Songlin Y., and Dahua F. (2010); Preparation of

liposomes containing zedoary turmeric oil using freeze-drying of liposomes via TBA/water cosolvent systems and evaluation of the bioavailability of the oil; Journal of Drug Targeting, 18 (2): 148-155.

243. Zidan A.S., Spinks C., Fortunak J., Habib M. and Khan M. A.

(2010); Near-Infrared Investigations of Novel Anti-HIV Tenofovir Liposomes; AAPS 12 (2): 202-214.

244. Zucker D., Marcus D., Barenholz, Y. and Goldblum A. (2009);

Liposome Drug's Loading Efficiency: A Working Model Based on Loading Conditions and Drug's Physicochemical Properties; Journal of Controlled Release, 139:73-80.

245. Zuzanna D. K. and Agata D. J. (2010); Liposomes as delivery

systems for antibiotics, Review; International Journal of Pharmaceutics, 387: 187-198.