Você está na página 1de 237

DEVELOPMENT AND EVALUATION OF

THERMO-REVERSIBLE SUBCUTANEOUS AND


OPHTHALMIC DRUG DELIVERY SYSTEM

Ph.D Thesis

By

FAZLI NASIR

DEPARTMENT OF PHARMACY
UNIVERSITY OF PESHAWAR
2012
DEVELOPMENT AND EVALUATION OF
THERMO-REVERSIBLE SUBCUTANEOUS AND
OPHTHALMIC DRUG DELIVERY SYSTEM

Ph.D Thesis

By

FAZLI NASIR

THESIS SUBMITTED TO UNIVERSITY OF PESHAWAR IN


PARTIAL FULFILLMENT FOR THE DEGREE OF
DOCTOR OF PHILOSOPHY IN PHARMACY

DEPARTMENT OF PHARMACY
UNIVERSITY OF PESHAWAR
2012
Table of contents

ABSTRACT ................................................................................................................................... 1

1. INTRODUCTION ................................................................................................................ 4

2. BACK GROUND ................................................................................................................... 9

2.1 Drug Delivery System ............................................................................................................... 9

2.2 Delayed Release ......................................................................................................................10

2.3 Extended Release ....................................................................................................................11

2.4 Site Specific Targeting..............................................................................................................11

2.5 Receptor Targeting ..................................................................................................................11

2.6 Fast Dissolve Drug Delivery System (Flash)...............................................................................12

2.7 Drug Modification ...................................................................................................................13

2.8 Dosage Form Modification.......................................................................................................13


2.8.1 Reservoir Devices .........................................................................................................................13
2.8.2 Matrix Devices .............................................................................................................................13
2.8.3 Hybrid System ..............................................................................................................................14

2.9 Ophthalmic Drug Delivery:.......................................................................................................15


2.9.1 Eye ...............................................................................................................................................15
2.9.1.1 The Outer Layer...................................................................................................................16
2.9.1.2 The Inner Layer ...................................................................................................................18
2.9.1.3 Drug Kinetics through Eye ...................................................................................................18

2.10 Ocular Routes ..........................................................................................................................19


2.10.1 Topical Ocular ..........................................................................................................................19
2.10.2 Sub-conjunctival ......................................................................................................................20
2.10.3 Intra-vitreal ..............................................................................................................................20

i
2.11 Challenges in Ocular Drug Delivery ..........................................................................................21
2.11.1 Ophthalmic Dosage Forms ...................................................................................................... 23
2.11.1.1 Aqueous Solutions...............................................................................................................23
2.11.1.2 Suspensions .........................................................................................................................23
2.11.1.3 Ointments ...........................................................................................................................24
2.11.1.4 Pre-Formed Gels and Muco-adhesive Polymer Systems.....................................................24
2.11.1.5 Gel Forming Liquids.............................................................................................................24
2.11.1.6 Non-erodible Ocular Inserts ................................................................................................25
2.11.1.7 Prodrugs ..............................................................................................................................25
2.11.1.8 Microspheres and Nano-particles .......................................................................................26

2.12 Subcutaneous Drug Delivery .................................................................................................... 26

2.13 Parenteral Depot System .........................................................................................................29


2.13.1 Reasons for development of PDS (Parenteral Depot System).................................................29
2.13.1.1 Advantages:.........................................................................................................................29
2.13.1.2 Disadvantages: ....................................................................................................................30
2.13.1.3 Types of Depot Formulations: .............................................................................................30
2.13.2 Injectable Drug Delivery System.............................................................................................. 30
2.13.2.1 Emulsions ............................................................................................................................30
2.13.2.2 Liposomes ...........................................................................................................................30
2.13.2.3 Microspheres: .....................................................................................................................31
2.13.2.4 Micelles ...............................................................................................................................32
2.13.2.5 Thermoplastic Pastes .......................................................................................................... 32
2.13.2.6 Thermosets .........................................................................................................................33
2.13.3 Stimuli Sensitive Solution to Gel..............................................................................................34
2.13.3.1 Temperature Sensitive Hydrogels .......................................................................................34
2.13.3.2 pH Sensitive Hydrogels........................................................................................................ 34
2.13.3.3 In situ Solidifying Organogels ..............................................................................................35
2.13.4 Polymers in Drug Delivery System ...........................................................................................35
2.13.4.1 Classification of Polymers ................................................................................................... 36
2.13.4.2 Properties of Polymers as Candidate for Controlled Release .............................................40

2.14 Hydrogels ................................................................................................................................41


2.14.1 Environmental Sensitive Hydrogels .........................................................................................42
2.14.1.1 Temperature sensitive hydrogels ........................................................................................42

ii
2.14.1.2 pH Sensitive Hydrogels........................................................................................................ 42
2.14.1.3 Electric Signal Sensitive Hydrogels ......................................................................................42
2.14.1.4 Light Sensitive Hydrogels .................................................................................................... 42
2.14.1.5 Ion Sensitive Hydrogels ....................................................................................................... 42
2.14.1.6 Pressure Sensitive Hydrogels ..............................................................................................42
2.14.1.7 Temperature Sensitive Hydrogels .......................................................................................43
2.14.2 Pluronic (Poloxamers) .............................................................................................................44
2.14.3 Methyl Cellulose (MC) .............................................................................................................50
2.14.3.1 Biodegradation ....................................................................................................................51
2.14.3.2 Thermo-gelation..................................................................................................................51
2.14.3.3 Applications of Methyl Cellulose.........................................................................................52
2.14.4 Hydroxy propyl methyl cellulose (HPMC) ................................................................................53
2.14.4.1 Thermo-gelation..................................................................................................................54
2.14.4.2 Applications of Hydroxy propyl methylcellulose (HPMC) ...................................................55
2.14.5 Diclofenac Sodium ...................................................................................................................56
2.14.5.1 Chemical Name ...................................................................................................................56
2.14.5.2 Molecular Formula and Structure .......................................................................................56
2.14.5.3 Mechanism of Action .......................................................................................................... 56
2.14.5.4 Uses .....................................................................................................................................56
2.14.5.5 Dosage and Administration .................................................................................................57
2.14.5.6 Pharmacokinetics ................................................................................................................57
2.14.6 Timolol Maleate.......................................................................................................................60
2.14.6.1 Chemical Formula and Structure.........................................................................................60
2.14.6.2 Molecular Weight................................................................................................................60
2.14.6.3 Description ..........................................................................................................................60
2.14.6.4 Melting Point.......................................................................................................................60
2.14.6.5 Solubility..............................................................................................................................61
2.14.6.6 pH ........................................................................................................................................61
2.14.6.7 Uses .....................................................................................................................................61
2.14.6.8 Dosage.................................................................................................................................61
2.14.6.9 Pharmacokinetics ................................................................................................................61
2.14.6.10 Contraindication ............................................................................................................. 62
2.14.6.11 Side Effects......................................................................................................................62
2.14.6.12 Drug interactions ............................................................................................................ 62
2.14.7 Insulin ......................................................................................................................................63

iii
2.14.7.1 Insulin Production and Secretion ........................................................................................63
2.14.7.2 Effect of Insulin ...................................................................................................................64
2.14.7.3 Insulin Regulation................................................................................................................65

3. MATERIALS AND METHODS ........................................................................................ 68

3.1 Materials .................................................................................................................................68

3.2 Instrumentation ......................................................................................................................68

3.3 Study Design ...........................................................................................................................69


3.3.1 Preparation of Polymer Solutions ................................................................................................70
3.3.2 Measurement of the Sol–Gel Transition Temperature (Tsol-gel) ................................................72
3.3.3 Measurement of Steady Shear Viscosity .....................................................................................72
3.3.4 Clarity of the Formed Gel .............................................................................................................73
3.3.5 Autoclaving ..................................................................................................................................73
3.3.6 Preparation of Drug Polymer Solutions .......................................................................................73
3.3.7 Preparation of Diclofenac Sodium In-situ Gel Formulations .......................................................73
3.3.8 Preparation of Timolol Maleate In-situ Gel Formulations ...........................................................74
3.3.9 Preparation of Human Insulin (recombinant) In-situ Gel Formulations ......................................75
3.3.10 Measurement of Tsol- gel DS Formulations ............................................................................76
3.3.11 Measurement of Steady Shear Viscosity of Diclofenac Sodium Formulations ........................77
3.3.12 Clarity of the Formed Gel of DS Formulations .........................................................................77
3.3.13 Drug Content of DS Formulations............................................................................................77
3.3.14 Determination of In-vitro of Drug Release from DS Formulations ..........................................77
3.3.15 Measurement of Tsol- gel of TM Formulations .......................................................................78
3.3.16 Measurement of Steady Shear Viscosity of TM Formulations ................................................78
3.3.17 Clarity of the Formed Gel of TM Formulations........................................................................79
3.3.18 Drug Content of TM Formulations ..........................................................................................79
3.3.19 Determination of In-vitro of Drug Release from TM Formulations .........................................79
3.3.20 Measurement of Tsol- gel Insulin Preparations ......................................................................80
3.3.21 Measurement of Steady Shear Viscosity of Insulin Preparations ............................................80
3.3.22 Clarity of the Formed Gel of Insulin Preparations ...................................................................81
3.3.23 Drug Content of Insulin Preparations ......................................................................................81
3.3.24 Determination of In-vitro Drug Release from Insulin Preparations ........................................81
3.3.25 Drug Release Kinetics ..............................................................................................................82

iv
3.3.25.1 Zero Order Kinetic Model....................................................................................................82
3.3.25.2 First Order Kinetic Model .................................................................................................... 82
3.3.25.3 Higuchi Model .....................................................................................................................83
3.3.25.4 Hixson –Crowell Model .......................................................................................................83
3.3.25.5 Korsmeyer-Peppas Model ...................................................................................................83
3.3.26 In Vivo Evaluation of Formulations..........................................................................................84
3.3.26.1 Animal Handling ..................................................................................................................84
3.3.26.2 Diclofenac Sodium Administration and Sampling ...............................................................84
3.3.26.3 Timolol Maleate Administration and Sampling...................................................................85
3.3.26.4 Insulin Administration and Sampling ..................................................................................85
3.3.27 Pharmacokinetic Parameters ..................................................................................................86
3.3.28 HPLC analysis method .............................................................................................................86
3.3.28.1 Materials and methods .......................................................................................................86
3.3.28.2 Preparation of Standard Stock Solutions ............................................................................87
3.3.28.3 Sample Preparation.............................................................................................................87
3.3.28.4 Liquid-Liquid Extraction ...................................................................................................... 88
3.3.28.5 Chromatographic Conditions ..............................................................................................89
3.3.28.6 Chromatographic Conditions and Experimental Parameters Optimization ........................90
3.3.28.7 Validation of the Method ....................................................................................................91

4. RESULTS AND DISCUSSION ......................................................................................... 97

4.1 HPLC –UV Method for Analysis of Diclofenac Sodium and Timolol Maleate ..............................97
4.1.1 Optimization of HPLC Experimental Parameters .........................................................................98
4.1.1.1 Mobile Phase.......................................................................................................................98
4.1.1.2 Stationary Phase .................................................................................................................99
4.1.1.3 Flow Rate.............................................................................................................................99
4.1.1.4 Column Oven Temperature...............................................................................................100
4.1.1.5 pH ......................................................................................................................................100
4.1.1.6 Wave Length .....................................................................................................................101
4.1.1.7 Internal Standard ..............................................................................................................102
4.1.1.8 Sample Preparation........................................................................................................... 102
4.1.1.9 Method Validation ............................................................................................................ 103

4.2 Physical Evaluation of In-situ Solution to Gel Formulations .................................................... 108


4.2.1 Measurement of the Sol-Gel Transition Temperatures (Tsol-gel) .............................................108

v
4.2.2 Measurement of Rheological Parameters .................................................................................110
4.2.3 Clarity of the Formed Gel ...........................................................................................................112
4.2.4 Autoclaving ................................................................................................................................113
4.2.5 In-vitro Evaluation of In-situ Drug Formulations .......................................................................114
4.2.5.1 In-vitro Evaluation of In-situ Diclofenac Sodium Sol-Gel Formulations ............................114
4.2.5.2 In vitro Drug Release From DS In-situ Gels........................................................................117
4.2.5.3 Drug Release Kinetics from DS In-situ Gels .......................................................................121
4.2.5.4 In-vitro Evaluation of In-situ TM sol-gel Formulations......................................................131
4.2.5.5 Drug Release Kinetics from TM In-situ Gels ......................................................................137
4.2.5.6 In-vitro Evaluation of In-situ Insulin Sol-Gel Formulations ...............................................145
4.2.6 In-Vivo Evaluation of In-Situ Thermoreversible Gels .................................................................160
4.2.6.1 In-vivo Evaluation of DS In-situ gels ..................................................................................160
4.2.6.2 Subcutaneous Route .........................................................................................................160
4.2.6.3 Ocular Route .....................................................................................................................169
4.2.6.4 In-vivo Evaluation of Timolol Maleate In-situ Gels ...........................................................178
4.2.6.5 In-vivo Evaluation of Insulin In-situ Gels ...........................................................................187

5. CONCLUSION ................................................................................................................. 193

6. REFERENCES .................................................................................................................. 197

vi
DEDICATION

I would like to dedicate this thesis to my beloved mother who was

spring of spiritual, religious and moral support for me throughout

my life.

I also dedicate this thesis to my wife, my daughters Salwa, Meerab,

Aroosh and my son Muhammad for their sufferings and support.

vii
ACKNOWLEDGMENTS

All the praises to almighty Allah subhanatallah who gave me courage, knowledge,

patience and health to complete my Ph. D research work.

It is difficult to acknowledge everyone who has helped me over the last 5 ½ years in my

research work in earning my Ph.D. The work was difficult and I could not have done it

without their support and help.

First and foremost, I wish to thank my wife and family who have supported and prayed

for me throughout my research work. They have been steadfast and loving, made my

difficulties less difficult, and always been there when I needed them.

I am more than grateful to my supervisor Prof. Dr. Zafar Iqbal, B. Pharm., M. Pharm.

(Pak.), Ph.D, Post Doc. (UK). He is the reason that I decided to pursue a doctorate degree

and without his influence I would not be where I am now. From him I have learned the

value of continued excellence, education, and personal development throughout my

career.

I would like to thank the GSC and ASRB members; they have been invaluable mentors

in my doctorate progress. They have also been trusted advisors with me in my personal

endeavors and future career plans. I value their support and counsel and hope to be able

to continue our relationship for many years.

viii
I am also thankful to the HEC Pakistan for promoting the culture of higher education and

research. I am also grateful to the University of Peshawar for the financial support by

funding the research project and for providing all the facilities required. I am also

thankful for the support and prayers of the faculty members of the Department of

Pharmacy University of Peshawar.

I am thankful to whole of my research group and laboratory fellows Mr. Amirzada, Dr.

Abad Khan, Dr. Imran Khan, Mr. Yasir Shah, Mr. Latif Ahmed, Mr. Muhammad

Ismail, Mr. Amanullah, Ms. Naila, Ms. Shabnum Nazir, Ms. Saira, Mr. Abuzar

Khan, Mr. Muhammad Ismail, Mr. Fazli Khuda, Dr. Roohullah, Dr. Akhlaq

Ahmed, Mr. Abbas Khan, Mr. Khalid Javed and Mr. Hassan Qayum, for their

support and help. It was a pleasure to have you as colleagues and friends.

My thanks also go to Mr. Muzaffar Abbas, Mr Salar Ahmed and Mr. Gohar Ali who

helped me in carrying out the In-vivo studies. I have to say special thanks for their help

during my experimental work to Mr. Aman Gul, Mr. Ihtesham, Mr. Inamullah and

Mr. Saeed Shah. I am also thankful to the laboratory and clerical staff of the Department

of Pharmacy University of Peshawar for their co-operation.

In the last but not the least I wish to thank my parents who instilled in me perseverance

and faith in my abilities. I did not heed their advice until later in my life, but not too late

to make a difference. I am also grateful to my brothers, sisters and relatives who prayed

and supported me and shared the family responsibilities during my Ph. D studies.

ix
ABBREVIATIONS
AC Autoclaving
ACE Angiotensin converting enzyme
AH Aqueous Humour
API Active pharmaceutical ingredient
AUC Area under curve
AUMC Area under moving curve
CCD Charge coupled device
CL Clearance
Cmax Peak plasma concentration
CMC Critical micelle concentration
Cps Centipoise
CV Co-efficient of variation
DNA De-oxy ribonucleic acid
DS Diclofenac Sodium
DSC Differential scanning calorimetry
EDTA Ethylene diaminotetraamino acetic acid
G-CSF Glycosylated granulocyte colony stimulating factor
GI Gastro intestinal
GLUT Glutathione
HLB Hydrophilic lipophilic balance
HPLC High pressure liquid chromatography
HPMC Hydroxy propylmethyl cellulose
Hr Hour
IS Internal standard
IU International unit
LCST Lower critical solution temperature
LOD Limit of detection
LOQ Limit of quantification

x
MC Methyl cellulose
Min Minutes
ml Milliliter
MRT Mean residence time
MVL Multi vesicle liposomes
NCI Network chromatography interface
nm Nanometer
PCL poly(D,L-lactide) and poly(Ί-caprolactone)
PEG Polyethylene glycol
PEO Poly ethyleneoxide
pGH Porcine growth hormone
PK Pharmacokinetic
Pl Pluronic
PPO Poly propyleneoxide
PVC Polyvinyl chloride
RNA Ribonucleic acid
Rpm Revolution per minute
RSD Relative standard deviation
RST Rosuvastatin
S/N Signal to noise ratio
SD Standard deviation
TM Timolol Maleate
Tmax Time to achieved Cmax
UCST Uper critical solution temperature
UV Ultra violet
Vd Volume of distribution

xi
ABSTRACT

ABSTRACT

The aim and objective of the study was to develop and evaluate biodegradable polymer

drug delivery system that is capable of extended the drug release for prolong period of

time at predetermined rate and being a free flowing solution at room temperature and

converts to gel with increase in temperature.

In the current study four polymers (Poloxamer 407, Methyl cellulose, Hydroxypropyl

methylcellulose and polyethylene glycol) were used alone and in combination with each

other in different ratios. Pluronic PF-127 (Poloxamer 407) and methylcellulose (MC) gels

below 37oC at appropriate concentrations (P= 18% w/v, 20%w/v, MC=4%w/v,

PM=15/3%w/v, MPG3/2w/v and MPG1.5/10% w/v).

To evaluate the efficacy of the in-situ thermoreversible formulations in controlling the

drug release three drugs diclofenac sodium (hydrophobic), timolol maleate (hydrophilic)

and insulin (protein) were selected. The in-situ gels were evaluated for their physical

properties like Tsol-gel, viscosity, clarity of solution and gel. The drug delivery system

was also evaluated for drug content, in-vitro drug release and pharmacokinetic

parameters were also determine with in-vivo studies. The data obtained for drug content

after autoclaving the solutions indicates that autoclaving results in degradation of DS

while it has no significant effect on TM.

1
ABSTRACT

To predict the drug release kinetics and mechanism various mathematical models were

applied to the in-vitro dissolution data. To predict the pharmacokinetic parameters Pk-

Summit software was used.

Based upon the in-vitro and In-vivo data it was concluded that the system consisting of

the poloxamer 407 in concentration of 20% (DP20 and TP20) are the most capable

formulation for extending the drug release and maintaining therapeutic blood level of DS

and TM for longer duration. While the formulation IPM15/3 consisting of 15%w/v

poloxamer 406 and 3% MC w/v retarded the drug release and maintaining the plasma

insulin in steady state for longer duration of time.

The results obtained in current study suggests that the HPLC-UV method developed is

sensitive and rapid method for the determination of DS and TM in pharmaceutical

preparation and physiological fluids (plasma, AH). The data also suggests that the studied

polymers Poloxamer, MC and PG are good candidate to extend the drug release

possessing a unique thermoreversible property.

The physical data obtained during the experimental work indicates that the studied in-situ

thermorevesible sol-gel formulations were capable to retard the release of these drugs.

The drug delivery systems were smart enough to be used for the administration of the

studied drugs through subcutaneous and ophthalmic routes.

2
CHAPTER 1 INTRODUCTION

CHAPTER 1
INTRODUCTION

3
CHAPTER 1 INTRODUCTION

1. Introduction

“The delivery of a drug at predetermined rate and/or location according to the needs of

body and disease for definite time period is termed as controlled drug delivery”[1].

Various approaches have been adopted for controlling the release of the drug. The

common approaches are Drug modification and Dosage form modification[2].

Modification of dosage form is one of the most common approaches to retard the release

of the drug from the dosage form and thus extending the effective time period.

The applications of the polymers in pharmaceutical sciences particularly in drug delivery

systems and formulations are well known [3]. Some polymers separate from solution and

convert to gel upon solidification above a certain temperature. Natural polymers have

been used as food processing aids as well as in pharmacy. Thermo-reversible gelation has

been reported for gelatin and agarose [3-6].

Cellulose is water insoluble, but its hydrophilic derivatives are water soluble. When

cellulose derivatives have an optimum balance of hydrophilic and hydrophobic moieties,

they undergo a sol-to-gel transition in water. The sol–gel transition temperature depends

on the substitution of cellulose at the hydroxy group[7], Methyl cellulose and

Hydroxypropyl cellulose are typical examples[7]. With rise in temperature of solution of

these polymers the water becomes a poorer solvent and polymer–polymer interactions

become dominant resulting in a gel[8].

4
CHAPTER 1 INTRODUCTION

This transition temperature is known as the lower critical solution temperature (LCST).

Below this temperature, the polymers are water soluble. Above the LCST, these polymers

become hydrophobic and water insoluble that results in solidification [9]. Various

techniques such as spectroscopy [10], differential scanning calorimetry (DSC) and

rheology [9] can be employed to verify the sol–gel transition of such thermo-reversible

polymeric solutions [10].

Many polymers show a decrease in solubility as their hydrophobicity increases upon

temperature change. In such type of polymer solutions, three types of interactions are

observed, between the molecules of polymer, between water and polymer molecules and

between the molecules of water. For polymers exhibiting LCST, rise in temperature

results in a negative free energy of the system that decreases the interaction between

water and polymer molecules, facilitating rise in the other two types of associations. This

negative free energy is result of the increase in entropy. Due to water–water interactions

which are the governing associations in the system the entropy increases. This

phenomenon is called hydrophobic effect [10-13]. As a result of the increase in

hydrophobicity the polymer chains are linked by physical reversible linkage, and gels can

therefore return to solution after the temperature stimulus causing gelation is

removed[13].

The biodegradable thermo-reversible polymers have extensively been studied due to their

great compatibility, degradability within the biological system and temperature

sensitivity. Therefore, these polymers have been investigated as controlled in situ gel-

5
CHAPTER 1 INTRODUCTION

forming drug delivery system [14-17]. Different polymers like Poly (ethylene glycol)-

poly (propylene glycol)-poly(ethylene glycol) copolymer (PEG-PPG-PEG), known as

Poloxamer or Pluronic, has extensively been studied as a potential thermo-reversible drug

delivery system [9, 18].

Ketoprofen and spironolactone release kinetics were studied from Pluronics [19]. The

hydrophobicity of the drug markedly affected the drug release profile. The release of

more hydrophilic ketoprofen was maintained continuously over 2 weeks, and the release

rate was controlled by the initial polymer concentration. The more hydrophobic

spironolactone release was maintained over 2 months [19].

Block copolymers have been investigated intensively for the solubilization and

stabilization of water-insoluble drugs, such as cyclosporin A and paclitaxel, and various

protein pharmaceuticals, including Zn-insulin, porcine growth hormone (pGH), and

glycosylated granulocyte colony stimulating factor (G-CSF) [20].

This unique sol-to-gel transition has made the system attractive as an injectable drug

delivery system in an in situ gel-forming drug depot. Most applications are based on

Poloxamer PF-127 and include delivery of protein/peptide drugs, such as insulin,

epidermal growth factor, urease, interleukin-2, bone morphogenic protein, fibroblastic

growth factor and endothelial cell growth factor. Most release profiles show sustained

release kinetics over several hours [21-25]. Poloxamers have been suggested for use as an

ocular drug delivery carrier of pilocarpine, but an animal study of PF-127 showed marked

6
CHAPTER 1 INTRODUCTION

destruction of the retina [26]. The addition of poly(ethylene glycol) (PEG) or (poly vinyl

pyrrolidone) (PVP) accelerated pilocarpine release, while the addition of methylcellulose

slowed the release rate [27].

The solutions of these polymers are liquid at room temperature but when introduced into

the body will transform into gel, this phenomenon is known as sol-gel transition. It is

easy to manufacture, package and administer the drug quantitatively using liquid dosage

form compared with the gel/semisolid dosage form. Therefore, these polymers are useful

for the drug delivery through topical, subcutaneous, ocular routes. These formulations

may sustain the delivery of the drug for long period of time that will improve the

patient’s compliance.

7
CHAPTER 2 BACKGROUND

CHAPTER 2
BACKGROUND

8
CHAPTER 2 BACKGROUND

2. Back ground

2.1 Drug Delivery System

Active Pharmaceutical Ingredient (API) is rarely administered solely as pure substance

but almost always given in the form of dosage form or delivery system. The drug delivery

system is formulated invariably using drug and excipient(s) in appropriate proportions.

Drug delivery is the technique or process of administering active pharmaceutical

ingredient to achieve a therapeutic response in humans or animals [28]. The objective of

any drug delivery system is to make available therapeutic amount of drug to proper site in

the physiological system. Principally a dosage form is formulated to achieve predictable

therapeutic response of the drug included in the formulation. The drug delivery system

can be classified as:

 Conventional drug delivery system

 Modified or controlled drug delivery system.

It has been recognized that conventional drug delivery system for instance simple tablets,

solution and injections may not be the best mode of drug administration. To improve the

compliance towards drug administration and reduce the dosage fluctuation, more efforts,

novelty and innovations have been entrusted to designing effective delivery systems,

specially controlled drug delivery system. Significant milestones have been achieved in

the field of controlled drug delivery as a result of collaborative research work between

chemists, polymer scientists, engineers, pharmacologists, engineers and medical

researchers [29].The objectives of drug delivery system includes;

9
CHAPTER 2 BACKGROUND

 Spatial Placement: drug targeting to specific organ

 Temporal delivery: Controlling the rate of drug delivery to target site, thus

maintaining drug concentration.

The conventional delivery system has potential problems these are;

 Lack of specific targeted delivery (temporal delivery)

 Repeated dosage after specific interval. If the interval is not proper there will be

variable troughs and peaks.

 Patient non compliance

 Increased side effects

The above problems of conventional dosage form stimulate the formulator both in

industry and laboratory level to develop modified release dosage form. “The delivery of a

drug at predetermined rate and/or location according to the needs of body and disease for

definite time period is termed as modified/controlled drug delivery” [1]. Modified release

delivery system can be classified as [1]:

2.2 Delayed Release

These are type of dosage form that releases the drug at the time other than immediately

following the administration. The delayed release of the drug may be time dependant or

based on environmental conditions such gastric pH, enzymes or pressure etc. The

example includes enteric coated tablets, where a drug release is dependent on acid

resistant coating (CAP, Eudragit etc) repeated action tablets or spansules.

10
CHAPTER 2 BACKGROUND

2.3 Extended Release

These include delivery system that maintains therapeutic plasma levels of administered

drug for prolong time. Such dosage form reduces the dosage frequency compared to the

immediate/conventional dosage form. The drug release is in continuous manner over

prolong period of time to maintain the plasma concentration, the drug release is mostly

independent of the environmental conditions.

2.4 Site Specific Targeting

In such system the drug delivery is targeted to the surrounding or in the pathologic tissue

or organ. This is a technique of drug delivery in a manner that increases the drug

concentration in the targeted area of the body relative to other tissues. The prime goal is

to extend, localize and have confined drug interaction with the targeted tissue or organ.

The targeted drug delivery reduces the total incorporated dose in the dosage form thus not

only provides better disease control but also reduces the systemic untoward effects.

Targeted drug delivery is of specific importance for oncology products.

Targeted drug delivery may be active (antibody delivery) or passive (enhanced

permeability and retention effect). Targeted drug delivery can be achieved by the use of

drug carriers like liposomes, niosomes, dendrimers, resealed erythrocytes, biodegradable

nanoparticles and micro-particles etc.

2.5 Receptor Targeting

In such system the target is a particular receptor with an organ or tissue. Folate receptor is

commonly used as marker for drugs effective in the cancer (Doxurubicin, antibodies,

11
CHAPTER 2 BACKGROUND

interlukins, radio-pharmaceuticals). The beauty of the delivery system is that a specific

drug response is achieved with a minimal of side effects.

2.6 Fast Dissolve Drug Delivery System (Flash)

It is type of rapidly dissolving or disintegrating solid dosage form that delivers the drug

in the oral cavity and gets dissolve without the aid of water or chewing. Rapid dissolution

is achieved by forming porous slack network (Zydis, Eli Lilly), or by addition of some

effervescence producing ingredient (Oraslav, Cima) or with using a combination of

disintegrating agent and swelling phenomenon (Flash Tab, Prographarm)

Controlled drug delivery system offer various advantages over the conventional drug

delivery system, the advantages include; “avoid patient compliance problem, less total

drug incorporation, reduction, minimize or eradicate local side effects, diminishes or

curtail systemic side effects, reducing the drug potentiation with chronic use, minimize

accumulation of administered drug with chronic use, improve efficiency in treatment,

cure or control pathologic condition more promptly, minimizing drug plasma

concentration variation, improve bioavailability of some drugs, make use of special

effects (e.g. sustained release aspirin for morning relief of arthritis by dosing before

bedtime), economic savings”. [1, 2]

Various approaches have been adopted for controlling the release of the drug. The

common approaches are drug modification and dosage form modification [2].

12
CHAPTER 2 BACKGROUND

2.7 Drug Modification

In this approach the physical or chemical structure of drug is altered to achieve better

control over the drug release or drug targeting. The common methods in this system

include; Drug complexes, Drug adsorbates and Pro-drug.

2.8 Dosage Form Modification

Modification of dosage form is one of the most common approaches to retard the release

of the drug from the dosage form and thus extending the effective time period. The

common approaches adopted for dosage form modification include [2].

2.8.1 Reservoir Devices

In such system the drug is wrapped in by a polymeric membrane from which the drug is

released either through diffusion, dissolution or erosion, these systems can also be

formulated using osmotic pressure. Such system make use of formation of either totally

or partially encapsulating drug with rate controlling drug surface by formation of coat

using polymer as coating material. The drug reservoir can be drug solid particles

(nanoparticles), a dispersion of drug solid particles, or a concentrated drug solution in a

liquid (micro emulsion) or solid-type dispersing medium.

2.8.2 Matrix Devices

This approach utilizes the dispersion of the drug in the polymer bed. The drug release

follows dissolution, erosion of the polymer or diffusion through polymer matrix alone or

combination of these mechanisms may take place.

13
CHAPTER 2 BACKGROUND

2.8.3 Hybrid System

This system take advantages of both reservoir and matrix system and is term as the most

versatile system. This system enjoys the advantages of both systems. The drug is

wrapped in polymeric cover and then blended with polymer matrix, thus giving better

control and targeting of the drug.

14
CHAPTER 2 BACKGROUND

2.9 Ophthalmic Drug Delivery:

2.9.1 Eye

The human eye is the organ which gives us the sense of sight. As anatomical part the eye

allows us to see and construe the dimensions, shapes and colors of objects. The light

emitted by the object is focused onto the retina and at the back of the eye. When the light

rays strike the lens, it passes through the vitreous humour, a jelly-like substance and

arrives at the retina. The retina works like a camera film, or a charge-coupled device

(CCD) of a digital camera[30].

Figure 2.1: Anatomy of the eye

The retina consists of three types of light sensitive photoreceptors; rods, cones and

ganglion cells. These receptors convert light rays through electric transduction to the

15
CHAPTER 2 BACKGROUND

electrical signals. The impulses are transmitted through the optic nerve to the visual

cortex in the brain, where the image is processed and perceived.

Anatomically eye is divided into two segments i.e. the anterior and posterior segments

(Fig 2.1).

2.9.1.1 The Outer Layer

2.9.1.1.1 The Sclera and Cornea

Sclera is the white tough, fibrous outer layer that serves as protective cover. The front of

the sclera is covered by the conjunctiva, which is a thin, transparent membrane that

produces tear film that lubricates and protects the eye while it moves in its socket.

Cornea is a dome-shaped structure. It is transparent, and its function is to focus the

incoming light onto the retina.

2.9.1.1.2 Iris

Iris is coloured part that controls the size of the pupil. The iris sits between the anterior

chamber and the posterior chamber.

2.9.1.1.3 Pupil

Pupil is the black area in the centre of the iris. The size of pupil varies with the intensity

of the light.

16
CHAPTER 2 BACKGROUND

2.9.1.1.4 Aqueous Humour

It is a transparent fluid, which is constantly being produced by the ciliary body. It has a

refractive index of 1.337. Aqueous humour is important for nourishing the lens and

cornea. It is drained through canal of Schlem.

2.9.1.1.5 Lens

It is a clear, flexible structure that changes shape so that to focus on objects at varying

distances.

2.9.1.1.6 Ciliary Body

When contracted accommodate the lens for close up vision and when relaxed fix the lens

for long-range vision.

2.9.1.1.7 Vitreous Humour

It is a jelly-like substance filling the back of the eye behind the lens. The vitreous humour

maintains shape and transmits light to the back of eye.

2.9.1.1.8 Choroid

It is a highly pigmented and separates sclera and the retina, choroid is rich in blood

supply thus also works as transporter of oxygen and nutrients to the retina.

17
CHAPTER 2 BACKGROUND

2.9.1.2 The Inner Layer

2.9.1.2.1 Retina

Retina is the most sensitive part of the eye by virtue of containing millions of

photoreceptors cells that are linked to nerve fibers.

2.9.1.2.2 Blind spot

It is also known as the optic disc where all of nerve fibers converge to form the optic

nerve.

2.9.1.2.3 Fovea

It is the most sensitive area, providing the sharpest vision.

2.9.1.3 Drug Kinetics through Eye

Figure2.2: “Schematic presentation of the ocular routes of drug kinetics” [31].

1. “Trans-corneal permeation from the lachrymal fluid into the anterior segment

2. Non-corneal drug permeation across the conjunctiva and sclera into the anterior

segment

18
CHAPTER 2 BACKGROUND

3. Drug distribution from the blood stream into the anterior segment

4. Elimination of drug from the anterior segment by the aqueous humour

5. Drug elimination from the aqueous humour into the systemic circulation

6. Drug distribution from the blood into the posterior eye segment

7. Intra-vitreal drug administration,

8. Drug elimination from the vitreous humour through posterior route

9. Drug elimination from the vitreous humour via anterior route to the posterior

segment”

2.10 Ocular Routes

Drug to the eye can be administered through several possible routes. The selection of the

route depends primarily on the target tissue. The anterior segmented is targeted with

topical and sub conjunctival administrations and intra-vitreal administration for posterior

segment[31].

2.10.1 Topical Ocular

Ocular drug administration is accomplished by eye drops, the disadvantage associated

with the eye drops is the short contact time on the eye surface. The non-conventional

formulation design (preformed gels, in-situ sol-gel, ointments and inserts) will increase

the contact and residence time, thus enhancing the duration of drug action [32]. On

administration of eye drop the peak concentration in the anterior chamber is reached after

20–30 minutes [33]. Drug is eliminated from the aqueous humour by two main

mechanisms: by aqueous turnover and by the venous blood flow [34] (Figure 2.2). The

aqueous turnover has a rate of about 3μl/min.

19
CHAPTER 2 BACKGROUND

The other mechanism depends upon the drug permeability across the vessel walls.

Therefore the elimination of lipophilic drugs is faster as compared to hydrophilic drugs

from anterior chamber. The half-lives of drugs in the anterior chamber are typically short,

about an hour. Flow of aqueous humour from the posterior chamber to the anterior

chamber is another limiting factor.

2.10.2 Sub-conjunctival

Injection through this route is encouraged to deliver high drug concentration to the uvea.

The advancement and progress made by material scientists and pharmaceutical

formulator innovations have provided new stimulating possibilities to develop controlled

release drug delivery system to deliver drugs to the posterior segment of the eye [35].

After sub-Conjunctival injection drug is absorbed rapidly as it crosses the sclera which is

more permeable than the cornea. Interestingly the lipophilicity of drug has no pronounced

effect on drug permeability through sclera [36, 37]. Thus it seems easy to deliver drug to

sclera and choroid via this route.

2.10.3 Intra-vitreal

This route directly administers drug to the vitreous and retina (Figure 2.2). After intra-

vitreal injection the drug is eliminated by two main routes: anterior and posterior [34].

Drugs can be administered to the vitreous also in controlled release formulations

(liposomes, microspheres, implants).

20
CHAPTER 2 BACKGROUND

2.11 Challenges in Ocular Drug Delivery

The anatomy, physiology and biochemistry of the eye render this organ impervious to

foreign substances. It is a challenge to the formulator to overcome and to provide the

drug in sufficient effective concentration to the target tissues. The challenges and

problems encountered by the formulator are rapid and extensive drug elimination.

Figure 2.3: Drug elimination pathways from the pre-corneal area [38].

After instillation, the flow of lachrymal fluid removes instilled solution from the eye by

naso-lachrymal duct rapidly in a couple of minutes [39]. The eye maintains its residence

volume at 7-10μl while the instilled eye drop is in the range of 20-50μl that results in

higher drainage rate to maintain the residence volume [38]. The systemic absorption via

blood capillaries in the conjunctival sac or from the nasal cavity also reduces the

21
CHAPTER 2 BACKGROUND

availability of the drug from local absorption [40, 41]. This results in very low drug (1 -

6%) availability in the intra ocular tissues through cornea. The reason for this inefficient

drug delivery includes rapid tear turnover, lachrymal drainage and drug dilution by tears.

As most of the drug get systemically absorbed via the nose or gut after draining from eye.

This not only reduces the ocular bioavailability but also may lead to unwanted systemic

side effects and toxicity[38].

The following characteristics are required to optimize ocular drug delivery systems [38].

 “A good corneal penetration.

 A prolonged contact and residence time with corneal tissue.

 Ease of administration for the patient.

 A non-irritative, comfortable and biodegradable form”

Some common methods to prolong pre-corneal residence time include use of Hydrogels,

Liposomes, Inserts, micro and nano-carrier systems. In comparison with traditional

formulation, these systems have the following advantages [38].

 Increase contact time

 Prolonged drug release

 Reduced systemic side effects

 Reduced dosage frequency

 Better `patient compliance

22
CHAPTER 2 BACKGROUND

2.11.1 Ophthalmic Dosage Forms

Ophthalmic preparations (eye preparations) are sterile, liquid, semi-solid, or solid

preparations that may contain one or more active pharmaceutical ingredient(s) intended

for application to the conjunctiva, the conjunctival sac or the eyelids.

Despite severe limitations and problems significant improvement in Ocular drug delivery

have been made. The improvements have been with objective of providing and

maintaining the drug concentration for an extended period.

2.11.1.1 Aqueous Solutions

Aqueous solution offers many advantages including the simplicity of large scale

manufacture, uniform dosage and no irritation. The major disadvantage associated is the

minimum contact time with the eye tissue, loss of active drug through drainage and

frequent administration. The contact time may be increased with addition of suitable

viscosity enhancer but this may offer instillation problem.

2.11.1.2 Suspensions

Suspensions are important ophthalmic delivery system that offers distinct advantages.

Most of the recently developed new drugs are water insoluble and are required to be

formulated as suspensions. From pharmaceutical formulation and physical stability view

point suspension is more difficult and challenging to formulate compared to conventional

ophthalmic solutions. The major problem encountered by formulator is to overcome non-

homogeneity of the dosage form, cake formation, aggregation of the suspended particles,

redispersability, effective preservation, and ease of manufacture [42].

23
CHAPTER 2 BACKGROUND

2.11.1.3 Ointments

Ophthalmic ointments provide prolonged therapeutic actions. Major disadvantage of the

ophthalmic ointments is interference with the vision i.e. blurred vision due to refractive

index difference between the tears and the non-aqueous bases of the ointment [43].

2.11.1.4 Pre-Formed Gels and Muco-adhesive Polymer Systems

Muco-adhesive polymers have a property known as bio-adhesion meaning attachment of

a drug carrier to the epithelium and the mucosal surface. These polymers are able to

extend the contact time of the drug and thereby improve ocular bioavailability. There are

several bio-adhesive polymers now available with varying degree of muco-adhesive

performance [44].

2.11.1.5 Gel Forming Liquids

These are preparations which are clear free flowing liquid in packaging and can be

instilled in eye as drop but gel on contact with the eye by one of the following stimuli

[38];

 Physical stimuli like: Change in temperature, electric fields, light, pressure, sound

and magnetic fields.

 Chemical stimuli like: Change in pH and ion activation from biological fluid.

 Biological/ biochemical (bio-molecules) stimuli like: Change in Glucose level.

24
CHAPTER 2 BACKGROUND

These “intelligent” or “smart” polymers play important role in drug delivery. It is widely

accepted that increasing the viscosity of a drug formulation in the pre-corneal region will

lead to increased bioavailability, due to slower drainage rate from the cornea [45].

2.11.1.6 Non-erodible Ocular Inserts

Alza Corporation marketed the first ocular insert (Ocusert) as controlled drug delivery in

1975. The Ocusert is soft and flexible membrane and designed to be placed in the cul-de-

sac between the sclera and the eyelid that continuously release pilocarpine at a steady rate

for one week. The major disadvantage associated with the system is the insertion and

removal on a weekly basis.

2.11.1.7 Prodrugs

Prodrugs are pharmacologically in active that is bio-transformed chemically or

enzymaticaly to pharmacologically active compound. Some key advantages of prodrugs

are

 Higher bioavailability

 Enhanced potency

 Lesser dose

 Extended duration of action

 Lesser side effects

 Higher chemical and physical stability

25
CHAPTER 2 BACKGROUND

2.11.1.8 Microspheres and Nano-particles

Particulate polymeric delivery systems include microspheres and nanoparticles. Such

particles are commonly synthesized using biodegradable polymers such as polylactic co

polyglycolic acid. Drug delivery based on such system offer better control and

bioavailability of drug.

2.12 Subcutaneous Drug Delivery


The Parenteral administration route is the most effective and common for delivery of

active drug substances for which the bio-availability in limited by high first pass

metabolism.

Figure 2.4: skin tissue showing Hypodermis/Subcutaneous tissue

For the subcutaneous route, a needle is inserted into fatty tissue just beneath the skin. The

drug injected is absorbed in to the blood through the capillaries and thus introduced for

systemic effects. Macromolecules (Protein drugs), such as insulin, immunoglobulin that

are degraded by the oral route are administered via this route.

26
CHAPTER 2 BACKGROUND

Fig. 2.5: Subcutaneous route: needle inserted beneath the skin

Subcutaneous injectable products are administered as aqueous solutions, suspensions or

emulsions. The injection volume via this route is usually 2 ml. The rate of absorption

from this route is slow and constant thus providing sustained and extended

pharmacological effects [46]. This route also provides better tolerance and steady state

concentration for hydromorphine in cancer patients.

Subcutaneous drug delivery systems can be used both for systemic or localized effect.

Drug release kinetics can be altered by changing the character of injectable biomaterial

template or depot, and the properties of the drugs like size, hydrophobicity, etc. The

depot injection will avoid patient discomfort by avoiding surgical procedure as involved

with implants. Thus a long term drug delivery system through subcutaneous injection got

multiple benefits [47].

The development of sustained-release formulations for intramuscular/subcutaneous

administration has become an increasingly important issue for the last two decades [48].

27
CHAPTER 2 BACKGROUND

This will not only increase the patient compliance and comfort but also can minimize the

systemic side effects. Such type of parenteral drug delivery can be achieved by utilizing

drug delivery systems that will ensure continuous extended slow release of drug in

predictable manner.

28
CHAPTER 2 BACKGROUND

2.13 Parenteral Depot System

The injection of a substance in a form that tends to keep the drug at injection site will

prolong the drug absorption. Long acting parenteral drug formulations are designed,

ideally to provide slow constant, sustained, prolonged action.

2.13.1 Reasons for development of PDS (Parenteral Depot System)

The PDS is biodegradable system which is converted to non toxicological by-product

with in the physiological system and no surgical removal of depleted system is required.

The drug release from this system can be controlled by following mechanisms;

 Drug diffusion through the polymer

 Polymer surface erosion with concomitant release of drug.

 Cleavage of covalent bond between the polymer bulks or at the surface followed

by diffusion drug release.

2.13.1.1 Advantages:

 Patient convenience

 Potential for controlled release

 Avoiding the peaks and troughs, thus minimizing the risk of toxicity and

ineffectiveness

 Decreasing the frequency of dosing

 Efficient drug delivery

 Flexibility

29
CHAPTER 2 BACKGROUND

2.13.1.2 Disadvantages:

 Invasive

 Danger of device failure

 Commercial disadvantage

2.13.1.3 Types of Depot Formulations:

On the basis of different mechanism, depot formulation are categorized into four types

[49, 50]

 Dissolution controlled depot formulation

 Adsorption type depot formulation

 Encapsulation type depot formulation

 Esterification type depot formulation

2.13.2 Injectable Drug Delivery System

2.13.2.1 Emulsions

Emulsions are used extensively in parenteral products. Their use as depot type of

injection is limited because of physical instability. The possibility of coalescence and

dissolution in the surrounding body fluid has restricted the use of emulsion as choice for

long acting formulations.

2.13.2.2 Liposomes

Liposomes are bi-lipid vesicles which has internal aqueous core surrounded by

hydrophobic bi layer. Liposomes were employed as sustained release drug carriers,

30
CHAPTER 2 BACKGROUND

therapeutic response of the drug entrapped within the liposomes is longer compared to

free drugs, but they are rapidly cleared off from the general circulation by macrophages

which make it poor choice for PDS. Other problems, such as physical and chemical

stability, sterilization and low drug entrapment, have limited the utility of liposomes.

In the area of injectable drug delivery systems, research into liposomes played a major

role in the past few decades. Significant efforts in basic and applied research institutions

led to the clinical development and ultimate approval by regulatory agencies for human

use of a lipid complex (Abelcet®, Amphotec®) and three liposome formulations,

AmbiSome®, DaunoXome® and a Stealth® liposome (DOXIL®) [51, 52]. The

liposomes can provide extended release of the active moiety by utilizing multivessicle

liposomes (MVL). It has been found that using long chain triglycerides as lipid bilayer

result in slower release rates from the MVL formulations than short chain triglycerides

[53].

2.13.2.3 Microspheres:

Microspheres are easy to deliver to the site of action but they have several inherent

disadvantages. The major issue is manufacturing processing as they are difficult to

sterilize, reproduce, also physical stability is a problem. The microspheres may also

migrate from the site of injection.

Depot injectable formulation with microsphere technology of leuprorelin was

achieved[54]. The drug release from these depot formulations was fairly constant for a

31
CHAPTER 2 BACKGROUND

period of over one month or 3 months in animals and humans after subcutaneous or intra-

muscular routes.

2.13.2.4 Micelles

Micelles are amphiphilic in nature possessing both hydrophilic and hydrophobic ends,

being capable of delivering both water soluble and insoluble drugs. The formation of

micelles depends upon the concentration of polymer termed as critical micelle

concentration (CMC). The CMC has key role in the micelle stability, lower the CMC

more stable will be micelles. The micelles are also prone to migration as that of

microspheres. The dilution of micelles at the time of injection or within the body fluid

may results in burst release and dose dumping.

2.13.2.5 Thermoplastic Pastes

Thermoplastic polymers have low melting points i.e. between 25-65oC. These are

administered as solution at elevated temperature and solidify upon cooling to body

temperature. Bio-erodible thermoplastic pastes could be prepared from such monomers

as, caprolactone, trimethylene carbonate, dioxanone, D,L-lactide, glycolide, and ortho

esters [55-57].

As thermoplastic system is injected at temperature greater than 60 °C, the injection at

higher temperature can be very painful and increases the chance of necrosis and scar

formation at the site of injection is the major disadvantages associated with these delivery

system [58].

32
CHAPTER 2 BACKGROUND

For the local delivery of taxol in the form of thermoplastic pastes was developed by

Zhang et al, the system was composed of triblock copolymer system of poly(D,L-

lactide)-block-poly(ethylene glycol)-block-poly(D,L-lactide) and blends of low

molecular weight poly(D,L-lactide) and poly(Ί-caprolactone) (PCL)[59].

2.13.2.6 Thermosets

Thermoset polymers can flow and can be molded in any shape. Upon heating, they

acquire their final shape. The process of molding into any shape is termed “curing” and

involves the establishment of covalent linkages between polymer chains to form a

macromolecular network [60].

The advantage of using this system is its facile syringe ability. The major disadvantage

associated with this system is burst drug release. This burst is due to the lag time for

solidification of the polymer.

33
CHAPTER 2 BACKGROUND

2.13.3 Stimuli Sensitive Solution to Gel

These polymers also termed as smart or intelligent polymers changes from solution to gel

phase in response to external stimuli. Polymers possessing such ‘sensor’ properties can

undergo sol-gel phase transitions upon changes in the environmental condition [61].

These system deliver the drug in controlled manner for extended time period at targeted

tissues. The environmental sensitive polymeric drug delivery systems are also called

intelligent or smart hydrogels [62]. These polymers show a dramatic physical change

conversion from free flowing liquid to viscous gel in response to small environmental

changes. These smart polymeric systems can be classified into the following classes.

2.13.3.1 Temperature Sensitive Hydrogels

These polymers are capable to swell or de-swell as a result of temperature variation of the

surrounding fluid. These polymers may be positive or negative temperature sensitive and

thermally reversible. The most commonly used thermoreversible gels are these prepared

from poly ethylene oxide-poly propylene oxide (Pluronics®, Tetronics®, poloxamer)

[26, 63].

2.13.3.2 pH Sensitive Hydrogels

The pH-sensitive polymers contain acidic or basic functional groups and are known as

poly-electrolytes [63]. With increase in pH the polymers with acidic functional group

show increase in swelling, while the swelling decreases with in case of polymers with

basic functional group. The most of anionic pH-sensitive polymers are based on PAA

(Carbopol®, carbomer) or its derivatives, polyvinyl acetal diethylaminoacetate [61, 64].

34
CHAPTER 2 BACKGROUND

2.13.3.3 In situ Solidifying Organogels

Organogels or oleaginous gels are amphiphilic lipids, which form different types of liquid

crystals upon contact with water. The most commonly employed lipids include glycerol

monooleate, glycerol monopalmitostearate, and glycerol monolinoleate. These

compounds form a cubic liquid crystal phase upon injection. This liquid crystalline

structure is gel-like and highly viscous. In vitro release of levonorgestrel from these

organogels was observed for up to 14 days.

Advantage of these systems is that they are biodegradable. Purity of waxes and stability

of oils are the major issues that need to be addressed [60, 65].

2.13.4 Polymers in Drug Delivery System

Polymers are macromolecules that are formed by covalent linkage of many small

molecules termed as monomers. The field of synthetic polymers is currently one of the

fastest growing materials industries. “The synthetic polymers provide good

biocompatibility and allow them to perform many biomimetic tasks that cannot be

performed by other synthetic materials, which include drug delivery, use as grafts for

arteries and veins and use in artificial tendons, ligaments and joints. The polymer used for

controlled release can be biodegradable, nonbiodegradable, non-biodegradable and

soluble [29]”. As drug-carriers, these polymers exist in the form of microspheres,

matrices and membranes.

35
CHAPTER 2 BACKGROUND

2.13.4.1 Classification of Polymers

Polymers can be classified by number of ways for easy understanding.

2.13.4.1.1 Classification Based on Source of Availability

 Natural Polymers

The source of these polymers is natural (plants and animals) e.g. starch, protein,

cellulose, nucleic acid (DNA, RNA)

 Synthetic Polymers

The man made polymers which are synthesized in laboratories. For example polyethene,

PVC nylon, teflon, bakelite, terylene, synthetic rubber, pluronics, tetronics etc.

 Semi synthetic Polymers

These polymers are chemical derivatives of natural polymers synthesized in laboratories.

For example Ethyl cellulose, methyl cellulose, Hydroxy propyl cellulose,

hydroxylpropylmethyl cellulose, vulcanized rubber, Rayon etc.

2.13.4.1.2 Classification Based on Structure

 Linear polymers

These are polymers in which monomeric units are linked together to form linear chain.

Common examples are high density polyethylene, nylon, PVC etc.

36
CHAPTER 2 BACKGROUND

A B

Figure 2.6: Chemical structure of Poly venyl chloride (A), and Polyethylene (B) and
Nylon (C)

 Branched Chain Polymers

These are polymers in which the monomers are joined to form long chains with side

chains or branches. Examples are low density polythene, glycogen, starch etc.

(Amylopectin).

 Cross Linked Polymers

These are polymers in which monomers unit are cross linked together to form three

dimensional network polymers. e.g. Bakelite, melamine, formaldehyde resin etc.

2.13.4.1.3 Classification Based on Type of Monomer

 Homopolymers

These are formed from identical monomers. e.g. Polyvenyl Chloride, polyethylene,

polymethylmethacrylic acid.

37
CHAPTER 2 BACKGROUND

 Copolymers

These are formed from non-identical monomers. e.g. Nylon, terylene.

2.13.4.1.4 Classification Based on Monomer Arrangement

 Alternating Copolymers

A-B-A-B-A-B-A (polyester, polyamide)

 Random Copolymers:

A-A-B-A-A-A-B-B-A-B-B-A-A. (styrene, butadiene).

 Block Copolymers:

A-A-B-B-B-A-A-B-B-A-A-B-B

(Glycerol propoxylate-block-ethoxylate, Poly(styrene)-block-poly(acrylic acid)).

 Graft Copolymers:

A–A–A–A–A–A–A–A–A–A–A–A–
| | |
B B B
| | |
B B B
| | |
B B B

e.g. Polycarbonate-g-Polyacrylate

38
CHAPTER 2 BACKGROUND

2.13.4.1.5 Classification Based on Basis of Mode of Synthesis

 Addition Polymers

This type of polymer is formed by addition of monomer without by product elimination.

Examples include, Polypropylene, polyethylene, polystyrene.

 Condensation Polymers

Such polymers are formed by the condensation of monomers with the elimination of

simple molecule like water, ammonia, HCl, alcohol etc. e.g. Nylon

2.13.4.1.6 Classification Based on Molecular Forces

 Elastomers

The monomers of the polymers are attached by weak forces and applying on stress these

polymers can easily be stretched and regains their original shape upon removal of the

stress. e.g. natural rubber

 Fibers

These polymers are held together by strong intermolecular forces. These polymers have

high tensile strength and less elasticity .e.g. Nylon 66, dacron, silk etc.

 Thermoplastics

These polymers get softened with application of heat and hardened on removal of heat

with no or little change in their characteristics. These polymers have intermediate type of

forces as compared to elastomers and fibres. e.g. Polythene, polystyrene, PVC, teflon etc.

39
CHAPTER 2 BACKGROUND

 Thermosetting Polymers

These undergo permanent change on application of heat. Upon heating they get highly

cross linked to form hard infusible and insoluble products. Upon heating high cross

linking occurs in these polymers and they become permanently rigid. e.g. Bakelite,

melamine formaldehyde resin etc.

2.13.4.2 Properties of Polymers as Candidate for Controlled Release

There are several important Properties to be considered in selecting or developing a

polymer for controlled delivery [66] :

 Biocompatibility, safety and toxicology

 If the polymer is biodegradable then what is the degradation rate and byproducts

and their safety

 Cost

 Chemical, physical, and mechanical properties

 Solubility

 Processing requirements

 Compatibility with the active agent and other ingredients in the formulation.

 Required sterilization methods

 Thermal transition temperatures

40
CHAPTER 2 BACKGROUND

2.14 Hydrogels

The polymeric network system that absorb huge amount of water without becoming

soluble in aqueous solution by cross linking of individual polymer chain [67]. The

hydrogel technologies have variety of applications including, food additives[68],

pharmaceuticals[69], biomedical implants [70] tissue engineering and regenerative

medicines [71], diagnostics [72], cellular immobility[73], separation of biomolecules or

cells[74] and barrier materials to regulate biological adhesions[75], Biosensor and drug

carriers[76].

Hydrogels can be classified on the basis of source as [77] Natural hydrogels and

Synthetic hydrogels. Hydrogels from natural source offer several advantages as they are

biocompatible, biodegradable and support cellular activity but there are disadvantages

associated with the hydrogels from natural origin that they may provoke immune

response because of the presence of pathogens and also may not provide sufficient

mechanical properties. Synthetic hydrogels, on the other hand are free from pathogenic

activity and with efficient fabrication biodegradable and physiologically friendly

hydrogels can be synthesized [67].

Hydrogels offer the following benefits which make them very good carriers for drugs and

tissue engineering [67];

 “Biocompatible

 Can be injected

 Easy to modify

41
CHAPTER 2 BACKGROUND

 Timed release of growth factors and other nutrients to ensure proper tissue growth

 Entrapment of microbial cells to reduce the toxicity within polyurethane hydrogel

beads”

2.14.1 Environmental Sensitive Hydrogels

Environmentally sensitive hydrogels have the ability to sense changes of pH,

temperature, ions, light or the concentration of metabolite. Natural hydrogel materials are

being investigated for tissue engineering, which include agarose, methylcellulose and

hylaronan.

The polymeric solutions capable of changing their rheological behavior in response to

environmental stimuli are termed as smart or intelligent polymers. The environmental

stimuli may be physical or chemical and include;

2.14.1.1 Temperature sensitive hydrogels

2.14.1.2 pH Sensitive Hydrogels

2.14.1.3 Electric Signal Sensitive Hydrogels

2.14.1.4 Light Sensitive Hydrogels

2.14.1.5 Ion Sensitive Hydrogels

2.14.1.6 Pressure Sensitive Hydrogels

42
CHAPTER 2 BACKGROUND

2.14.1.7 Temperature Sensitive Hydrogels

These polymers are sensitive to environmental temperature and change their solution

phase to gel phase. The common feature shared by almost all temperature sensitive

polymers that they contain hydrophobic group i.e. methyl, ethyl and propyl [63]. These

components of polymers must be insoluble above or below certain temperature, called the

upper or lower critical solution temperature (UCST and LCST). For drug delivery system

based on this property the polymers with LCST are more promising[26] . At LCST, the

interaction forces (hydrogen bonding) between water molecules and polymer weakens

compared to polymer-polymer and water-water interaction that results in phase separation

and the polymer forms gels[78]. As a result of the increase in hydrophobicity the polymer

chains are linked by physical reversible linkage, and gels can therefore return to solution

after the temperature stimulus causing gelation is removed[13]. These polymeric

solutions have display low viscosity (free flowing) at ambient temperature but form a

semi-solid gel at body temperature[79]. The most commonly used thermosensitive

polymers for controlled drug delivery include, polysaccharides (e.g., cellulose

derivatives, xyloglycan, chitosan), N-isopropylacrylamide and Pluronics (poloxamers,

(PEG-PLA-PEG)[20].

43
CHAPTER 2 BACKGROUND

2.14.2 Pluronic (Poloxamers)

This is a group of nonionic surfactants consisting of polyethylene oxide (PEO) and

polypropylene oxide (PPO) copolymers. Commonly used Pluronics (Poloxamers) include

F-68 (188), F-87 (237), F-108 (338) and F-127 (407), all of them are freely water soluble.

Their chemical formula is

HO[CH2-CH2O]a [CH(CH3)-CH2O]b[CH2-CH2O]aOH,

“b” is higher than 14. Poloxamers are white, waxy, free-flowing granules practically

odorless and tasteless[80]. Different poloxamers and their physical properties are shown

in table 2.1.

PF-127 is the most commonly employed polymer among the group used as

thermorevesible vehicle as a carrier for most routes of administration including oral,

topical[81, 82], intranasal[83], vaginal, rectal[84, 85], ocular[86-92], and parenteral

routes[92]. The potential use of PF-127 as an artificial skin has also been reported[93].

Table 2.1: Types and physical properties of Pluronic/Poloxamers (USP 32)

Average
Pluronic Poloxamer A b Physical Form
molecular weight

L-44 124 12 20 Liquid 2090-2360

F-68 188 80 27 Solid 7680-9510

F-87 237 64 37 Solid 6840-8830

F-108 338 141 44 Solid 12700-17400

F-127 407 101 56 Solid 9840-14600

Where a= oxyethylene b= oxypropylene

44
CHAPTER 2 BACKGROUND

Poloxamer 407 is available in the registered trademark of Pluronic F127 (BASF

laboratories), and Synperonic F127 (ICI laboratories), has a molecular weight of about

12,600 (9,840-14,600)[94] a and b are equal to 95-105 and 54 - 60, respectively. Its HLB

is 22 at 22oC [94-96].

Aqueous solutions of Poloxamer 407 show gelation on rise in temperature that is thermo

reversible. Thermo gelling property is completely reversible characterized by sol to gel

transition temperature. Below the transition temperature the solution remains free flowing

liquid, while above this temperature the solution form highly viscous semi solid gel. The

sol-gel transition is result of interaction between the segments of the block

copolymer[97], the molecules aggregate forming micelles at LCST . This micellization is

due to the dehydration of hydrophobic poly Propylene oxide blocks (Figure 2.7) and

hydration of poly Ethylene oxide chains[98].

Figure 2.7: Schematic representation of the association mechanism of Poloxamer 407 in


water

45
CHAPTER 2 BACKGROUND

2.14.2.1.1 Solublization Property

Poloxamer 407 facilitates solubilisation of poorly water soluble molecules like

Indomethacin [99] or insulin[84, 100, 101]. Solubility of piroxicam in water was

increased by 11-fold by adding 22.5% w/w Poloxamer 407[102].

2.14.2.1.2 Stabilization Property

Pluronic F-127 enhances the stability of incorporated drugs in its matrix, particularly of

proteins [103-108]. The three dimensional structure of proteins is better preserved in the

formulations containing pluronic F-127 as ingredient [98, 103-106]. Poloxamer 407 is

also effectively been used with W/O/W multiple emulsion to stabilize the preparation

[109].

2.14.2.1.3 Gel Strength

Gel strength of pluronic solution increases with increase in temperature and

concentration. The gel strength may be altered in the presence of drugs or additives.

Additives like, diclofenac, ethanol and propylene glycol weaken Poloxamer 407 gel

while ingredients like sodium chloride, sodium mono hydrogen phosphate and glycerin

results in increasing the gel strength [110, 111].

2.14.2.1.4 Adhesive Properties

Adhesive characteristic is of great importance in topical formulations (e.g., rectal,

cutaneous or ophthalmic preparations) as prolonged residence-time is required.

Adhesiveness increases with gel strength.

46
CHAPTER 2 BACKGROUND

2.14.2.1.5 Solution to Gel Transition Temperature

Sol-gel transition temperature increases with decrease in Poloxamer 407 concentration

[112]. As the transition temperature increases or decreases with the addition of drugs or

various additives the bio-adhesive forces decreases or increases respectively [27]. The

added substances interfere with the micellization of the pluronic and alter the dehydration

of hydrophobic PO blocks [97, 113, 114].

Molecules like diclofenac, ethanol, propylene glycol, and HCl reduce the gel strength and

bio-adhesive force and increase Tsol-gel while others like NaCl, Na2HPO4, NaH2PO4

do the opposite[111, 115]. Adding sodium alginate, polycarbophil or carbopol has been

reported to lower gelation temperature [85], use of benzalkonium chloride also decreased

Tsol-gel [116].

2.14.2.1.6 Drug Release from Pluronic Polymers

At low temperatures depending upon the polymer concentration, the polymeric solution

embedding the drug to be released is a free flowing liquid that can be injected into the

body via a syringe. At a higher temperature, the polymer molecules aggregate forming

micelles with the core hydrophobic poly (propylene oxide) (PPO) surrounded by the

hydrophilic poly- (ethylene oxide) (PEO) [117-119]. Upon sub-cutaneous or

intramuscular injection the temperature of this micellar solution increases to body

temperature, the micelles begin to entangle with other micelles forming gel within the

body. For pluronic F127 with 70% ethylene oxide ratio this concentration is about 20%

w/w at 37oC[120, 121]. The gel dissolves within the body by withdrawing water from its

47
CHAPTER 2 BACKGROUND

surrounding, causing drug release that is dependent on the dissolution of the gel, which is

a zero-order process.

To predict the drug release from Pluronic gels for different drug and polymer

compositions, several aspects need to be investigated. The most common include, drug

solubility in pluronic solutions, diffusion of the drug through gel, and water penetration

rate into the gel [122]. Dissolution rate for concentrated gel is slower as compare to less

concentrated pluronic gels because the coefficient for water diffusion within the gel is

lower in the concentrated gels as than for the dilute polymer gels [122].

2.14.2.1.7 Applications of Pluronic F-127

The unique thermo reversible and controlled drug release characteristics of PF-127 make

it an attractive candidate as a pharmaceutical vehicle for drugs through different routes of

administration.

2.14.2.1.7.1 Topical and Dermal Applications

The pluronic gel is evaluated as topical drug delivery vehicle by several workers [123-

138]. Indomethacin was incorporated in 20% pluronic F-127 as a base for percutaneous

drug absorption[123]. Chi et al prepared and evaluated pluronic F-127 gel containing

ketotifen as drug, the preparation showed prolonged anti-inflammatory and analgesic

activities and less systemic side effects [126]. Fentanyl was successfully formulated in

PF-127 base to provide sustained release effect [129]. The effect of concentration of PF-

127 on the release rate of anticancer agents, 5-flurouracil and adriamycin was evaluated,

it was observed that with increase in polymer concentration the drug release decreases

48
CHAPTER 2 BACKGROUND

[139]. The ceftiofur followed zero order kinetic being formulated in PF-127 and was

correlated well with the weight percentage of PF-127[134], formulation of benzoic acid

used as antiseptic in PF-127 formulation showed similar results [140].

Exploring the unique properties of PF-127, it has also been used in bone wound healing

in calvarial defects[135] (64), PF-127 gels also sufficiently mimic normal functions of

the skin epidermis acting as an “artificial skin”[141].

2.14.2.1.8 Buccal applications

Pluronic F-127 has been studied as vehicle for many drugs like triamcinolone [136],

Insulin[137] and human gingival fibroblast[142] administered through buccal cavity.

2.14.2.1.9 Rectal Application

Utilizing the muco-adhesive properties of pluronic F 127, the polymer is applied as

vehicle for rectal route within the concentration range of 17%-20%[143] and sustaining

the drug release for 10-15 hours[144].

2.14.2.1.10 Ophthalmic Applications

Pilocarpine HCl was incorporated in pluronic gels, the drug release and meiotic response

was significantly improved as compare with aqueous pilocarpine solution. The pluronic

in-situ gel showed increase tissue contact time and bioavailability[145]. 25%w/w solution

of Timolol showed 2.5 fold higher ocular concentration in aqueous humour as compared

to aqueous Timolol solution[146]. Poloxamer 407 (PF-127) are also evaluated for the

ocular delivery of the quinolones[147, 148] i.e. ciprofloxacin, moxifloxacin etc.

49
CHAPTER 2 BACKGROUND

2.14.2.1.11 Other Routes

Number of workers has successfully investigated and evaluated poloxamers as vehicle for

the routes like, intramuscular[149-151] , intravenous [152], intraperitoneal [149]and

intranasal[153] routes.

2.14.3 Methyl Cellulose (MC)

Methyl cellulose also known as methyl cellulose ether is a white powder with yellow

cast. Methyl cellulose is a synthetic methoxy derivative of cellulose. The degree of

substitution is 1.6-1.9, on O-6, O-2 or O-3 position of glucose. This chemical

modification makes it soluble in cold water[154]. MC is available in number of viscosity

grades depending upon the molecular weight as given in Table 2.2.

Table 2.2: Methylcellulose viscosity grade with molecular weight (sigma-Aldrich)

Approximate Viscosity of Approximate


Product Number
2%, at 20 °C Molecular Weight

M 7140 15 cPs 14,000

M 6385 25 cPs 17,000

M 0262 400 cPs 41,000

M 0387 1500 cPs 63,000

M 0512 4000 cPs 88,000

50
CHAPTER 2 BACKGROUND

Methylcellulose is used as a binder or thickener in pharmaceutical, cosmetic, food and

ceramic applications. Upon heating, methylcellulose converts to translucent gel in water,

this gelation is thermo-reversible in nature [155-157].

2.14.3.1 Biodegradation

Methyl cellulose and other cellulose derivatives are biodegradable as they are degraded

by several fungi and bacteria capable of producing cellulase enzyme [158]. Methyl

cellulose is biocompatible cellulose that has triggered its use in biomedical devices [159,

160]. As animal do not produce cellulose enzyme hence no resorption of cellulose in

animal and human tissues does occur.

2.14.3.2 Thermo-gelation

Methyl cellulose aqueous solution in concentration of 1-10% by weight forms a

thermorevesible gel [161]. In cellulose that is the raw material for methylcellulose, when

a certain number methoxyl groups are replaced for the hydroxyl groups, some hydrogen

bonds are broken and MC becomes water-soluble. At low temperatures, water molecules

surround the hydrophobic methoxyl groups forming “cagelike” structure, resulting in

making the MC gel [156, 162]. On increasing the temperature, these structures distort and

break to expose the hydrophobic regions, inducing the formation of aggregates and

resulting in gelation.

51
CHAPTER 2 BACKGROUND

OR

H H H
OR H2C

RO O
O H O
H
O RO
H H
CH2 OR
H H H

OR n-2/2

Figure 2.8: chemical structure of methyl cellulose

The transition temperature depends on the degree of substitution, with increasing the

degree of substitution the hydrophobic character increases, thus lowering the transition

temperature. The degree of substitution can be properly adjusted (i.e. 1.9-2.6) to obtain

specific formulations gelling at 37 °C and thus potentially useful for biomedical

applications [163-165].

2.14.3.3 Applications of Methyl Cellulose

2.14.3.3.1 Oral Route

Methyl cellulose is commonly employed as tablet binder [166, 167], both in conventional

release and sustained release tablets depending upon its molecular weight and viscosity

grade [168]. Riboflavin was successfully incorporated in methyl cellulose matrix [169] to

provide sustained delivery of the drug. It is also used in oral pharmaceutical suspensions

as suspending agent [170, 171].

52
CHAPTER 2 BACKGROUND

2.14.3.3.2 Ocular Route

Methyl cellulose is used in combination with thermorevesible polymers as mucoadhesive

agent or viscosity enhancer [146, 172, 173]. The addition of methylcellulose to the

aqueous solution of pluronic, carbopol, chitosan not only reduced the total concentration

of these polymers but also increase the ocular residence time thus enhancing the drug

bioavailability. Ciprofloxacin and timolol are successfully formulated in thermorevesible

ocular drug delivery vehicle in combination with methyl cellulose [146, 172].

2.14.3.3.3 Vaginal/Rectal Route

To enhance the residence time of the dosage form in the vagina and rectum use of

methylcellulose as mucoadhesive and thermo-gelation agent is successfully explored by

the pharmaceutical scientists [174, 175]. Addition of 1% MC as mucoadhesive agent in

poloxamer 18 % solution effectively enhanced the peak plasma concentration of

carbamezipine and reduced the gelation temperature [176]. In situ suppository system

was developed for mebaverine HCl and acetaminophen [175, 177].

2.14.4 Hydroxy propyl methyl cellulose (HPMC)

HPMC is a synthetic cellulose derivative it is propylene glycol ether of methylcellulose.

The chemical structure is shown as Fig. 1. The R substitution represents a –CH3 or a

CH2CH (CH3) OH group, or a hydrogen atom. The physicochemical properties of HPMC

are dependent upon: (i) the methoxy group content; (ii) the hydroxypropoxy group

content; and (iii) the molecular weight. The USP classifies the HPMC in four different

grades according to their relative –OCH3 and –OCH2CH (CH3) OH content. The four

grades are HPMC 1828, HPMC 2208, HPMC 2906 and HPMC 2910. The first two digits

53
CHAPTER 2 BACKGROUND

in the nomenclature indicate the percentage of methoxy-groups while the last two

indicates percentage of hydroxypropoxy-groups [178].

The substitution comprises 19-30% of methoxy group and 3-12% hydroxypropoxy group.

HPMC is soluble in cold water and polyethylene glycols, but insoluble in alcohol. HPMC

is stable at wide range of pH i.e. 3-11. Aqeuous HPMC solution forms thermo-reversible

gel on heating [179] .

OR

H H H
OR H2C

RO O
O H O
H
O RO
H H
CH2 OR
H H H

OR n-2/2

Figure 2.9: Chemical structure of HPMC. The substituent R represents either a –CH3, or
a –CH2CH(CH3)OH group, or a hydrogen atom

2.14.4.1 Thermo-gelation

When aqueous solutions of HPMC are heated, they gel at higher temperatures. The

gelling temperature depends upon the HPMC grade and concentration. The solution to

gel transition is completely reversible i.e. upon cooling the gel liquefies to form free

flowing solution. The transition from sol-gel is primarily due to the hydrophobic

interaction between molecules [161]. At lower temperatures the molecules are

surrounded by water molecules and are in are hydrated form. There is little polymer-

polymer interaction in solution state. As the solution is heated and temperature raises, the

54
CHAPTER 2 BACKGROUND

molecules gradually lose their water of hydration and polymer to polymer interaction

prevails resulting in sharp rise of viscosity and the solution tends to gel [161, 162].

2.14.4.2 Applications of Hydroxy propyl methylcellulose (HPMC)

2.14.4.2.1 Oral Route

HPMC is the most common excipient used in the manufacturing of oral tablets as binder

both in conventional and sustained release matrix tablets. It has good compressibility and

can be used equally well in wet granulation and direct compression. The HPMC begins to

swell forming a protective gel like layer around the tablet when it comes in contact with

physiological fluids leading to a sustained release of drugs [180, 181]. HPMC K15 and

K100 are studied to develop sustained release matrix tablets of diclofenac sodium,

ketoprofen, carbamezipine, naproxen sodium, ketrolac, losartan potassium, zidovudine,

isoniazid, resperidone and olanzepine etc[182-189]. HPMC is an alternate and

comparable polymer to replace gelatin as hard shell capsules [190-192]. The

hydroxypropyl methylcellulose is used as tablet coating film former as it has a very high

tensile strength and high force is needed to break the HPMC film.

2.14.4.2.2 Ophthalmic Route

To improve the ocular dug availability the thermo reversible gelation of HPMC is

successfully utilized to enhance the residence time and bioavailability of the drugs

including ciprofloxacin once a day, ofloxacin, gatifloxacin [146, 148, 172, 193-195].

55
CHAPTER 2 BACKGROUND

2.14.5 Diclofenac Sodium

Diclofenac sodium a phenylacetic acid derivative is Nonsteroidal Anti-inflammatory

Agent [196-200].

2.14.5.1 Chemical Name

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

2.14.5.2 Molecular Formula and Structure

C14H11C12NO2.NaC20H24Cl2N2O3

Figure 2.10: Chemical structure of Diclofenac Sodium

2.14.5.3 Mechanism of Action

Diclofenac inhibits cycloxygenase -1 and 2.

2.14.5.4 Uses

symptomatic treatment of osteoarthritis [201, 202], ankylosing spondylitis [203, 204], for

symptomatic treatment of infusion-related superficial thrombophlebitis [205], relief of

pain, including postoperative (e.g., orthopedic, gynecologic) pain, acute pain due to

minor strains, sprains, and contusions [206].

56
CHAPTER 2 BACKGROUND

2.14.5.5 Dosage and Administration

2.14.5.5.1 Rheumatoid Arthritis

50 or 75 mg twice daily [207]

2.14.5.5.2 Ankylosing Spondylitis

100–125 mg daily, administer as 25 mg 4 times daily [203, 208].

2.14.5.5.3 Pain

50 mg 3 times daily [206]

2.14.5.5.4 Dysmenorrhea

50 mg 3 times daily [209]

2.14.5.6 Pharmacokinetics

2.14.5.6.1 Bioavailability

Diclofenac sodium is well absorbed following oral administration, it undergoes first pass

metabolism extensively and only 50-60% of drug is available unchanged systemically

[198, 210-214]. After administration of conventional oral tablets peak plasma

concentration is achieved within about 1 hour [210, 211], following topical application as

1% gel the absorption is low as compared to oral administration and peak plasma

concentration is reched in about 10-14 hours [215].

57
CHAPTER 2 BACKGROUND

2.14.5.6.2 Distribution

Diclofenac is widely distributed [210, 211], after oral administration the concentration is

low in plasma as compared to synovial fluid [214, 216-219], more than 99% of drug is

protein bound [220, 221].

2.14.5.6.3 Metabolism

Diclofenac is metabolized in the liver, the major biotransformation reactions are

hydroxylation and conjugation reaction [210, 211, 219, 222].

2.14.5.6.4 Elimination Route

65% of drug is excreted in urine while the remaining 35% is eliminated in feces [210,

222, 223].

2.14.5.6.5 Half-life

The biological half life of diclofenac after oral administration is about 1-2 hours [212,

213, 224].

2.14.5.6.6 Contraindications

Known hypersensitivity to diclofenac [209], History of asthma, urticaria, or other

sensitivity reaction [225-227]

2.14.5.6.7 Adverse Effects

Abdominal pain or cramps, constipation, diarrhea, GI bleeding, peptic ulcer, vomiting,

dyspepsia, nausea, dizziness, liver function test abnormalities, renal function

abnormalities, anemia, prolonged bleeding time, pruritus, rash and tinnitus [228-230].

58
CHAPTER 2 BACKGROUND

2.14.5.6.8 Drug Interactions

ACE inhibitors, Angiotensin II receptor antagonists, Antacids (magnesium- or aluminum-

containing), Anticoagulants (warfarin), Aspirin, Cyclosporine, Diuretics (furosemide,

thiazides), Lithium, Methotrexate, Quinolones (ciprofloxacin) [206, 231-241]

59
CHAPTER 2 BACKGROUND

2.14.6 Timolol Maleate

Timolol (TM) (Fig. 2.11) 22 (S)-1-[(1-1-dimethyl)amino]-3-[[4-(4-morpholinyl9-1,2,5-

thiadiazol-3-yl]oxy]-2-propanol, is a nonspecific β-adrenergic blocker. TM was the first

β-blocker to be used as an antiglaucoma agent.

2.14.6.1 Chemical Formula and Structure

 C13H24N4O3S

Figure 2.11: Chemical structure of Timolol Maleate

2.14.6.2 Molecular Weight

 316.42

2.14.6.3 Description

White odorless powder

2.14.6.4 Melting Point

201.5-202.5oC [242, 243].

60
CHAPTER 2 BACKGROUND

2.14.6.5 Solubility

Timolol maleate is soluble in 1 in 15 parts water, 1 in 21 of alcohol, 1 part in 40 parts of

chloroform and practically insoluble in ether [242, 243].

2.14.6.6 pH

2% aqueous solution has pH 3.8 to 4.3 [242, 243].

2.14.6.7 Uses

Hypertension, angina pectoris, cardiac arrhythmias, migraine and glaucoma

2.14.6.8 Dosage

2.14.6.8.1 Glaucoma

One drop of 0.25 % solution twice daily, the dose may be increased to 1 drop of 0.5%

solution twice daily.

2.14.6.8.2 Hypertension

15 to 60 mg daily

2.14.6.9 Pharmacokinetics

2.14.6.9.1 Absorption

After oral administration the absorption from the GI tract is almost complete i.e. 90%.

The peak plasma concentration is attained within 0.5 -3 hours [244, 245]. The first pass

effect for timolol is not significant [246]. The onset of action after ocular administration

is 10-20 minutes [247].

61
CHAPTER 2 BACKGROUND

2.14.6.9.2 Distribution

After oral administration timolol bioavailability is 60% [248], the apparent volume of

distribution is 1.30 to 1.70 l/kg [248, 249]. Approximately 10% of the drug is bound to

plasma protein. After ocular administration timolol is distributed in cornea, iris,

conjunctiva, aqueous humour, liver, lung and kidney. Following the cutaneous route

timolol is 50-60 % absorbed systemically [250].

2.14.6.9.3 Biological Half life

2.5-5.0 hour [246, 249, 251].

2.14.6.9.4 Metabolism

Timolol is principally metabolized in liver through hydrolysis of the morpholino ring

with subsequent oxidation. 20% of the drug is eliminated as such via urine [246].

2.14.6.10 Contraindication

Timolol is contraindicated in patients with 2nd and 3rd degree AV block, cardiogenic

shock and asthma. Also contraindicated with other β-blockers [252].

2.14.6.11 Side Effects

Fatigue, confusion, depression, hallucinations, dizziness, vertigo, lethargy insomnia,

bradycardia, edema [252, 253].

2.14.6.12 Drug interactions

Clonidine, Epinephrine, NSAIDs, Prazosin, Insulin, Verapamil and theophylines.

62
CHAPTER 2 BACKGROUND

2.14.7 Insulin

Insulin is a naturally occurring hormone, composed of two polypeptide chains with the

empirical formula of C254 H377 N65 O75 S6 and has a molecular weight of 5808 Da. The

two chains are connected to one another by three disulfide bonds as seen in Figure 2.12.

It is produced and secreted from the β-cells in the islets of Langerhans in the pancreas.

The primary role of insulin is to facilitate the transport of glucose from the bloodstream

to cells in the body.

Figure 2.12: Structure of human insulin; the amino acid diagram of human insulin,
showing the A and B chains and 3 disulfide bonds

2.14.7.1 Insulin Production and Secretion

In healthy humans, insulin is synthesized during the secretion from proinsulin containing

86 amino acid chain in the β-cells of the pancreas. In the β-cells as first step polypeptide

called pre-proinsulin containing 110 amino acid sequence is synthesized [254]. The pre-

proinsulin is cleaved at N-terminus in the endoplasmic reticulum upon translocation and

63
CHAPTER 2 BACKGROUND

24-peptide sequence is removed and the polypeptide pro-insulin is produced. The three

disulfide bonds characteristic of insulin are formed upon folding of the pro-insulin as

tertiary protein [254]. Pro-insulin is then converted to insulin upon secretion, individually

four amino acids are removed and co secreted along insulin as C-peptide (connector

peptide)[254]. The mediator for insulin secretion is blood glucose level which is critically

controlled during fed and fasted state in healthy humans. Insulin release is also initiated

by glucagon-like peptide 1 (GLP-1), gastrin, vasoactive intestinal peptide, secretin,

gastrointestinal inhibitory peptide (GIP), cholecystokinin, gastrin-releasing peptide, and

enteroglucagon [255].

2.14.7.2 Effect of Insulin

Insulin concentrations in plasma are commonly measured in international units. One

international unit (IU) of insulin is equal to 6 nmol, that is equivalent to 34.8 μg [256].

The insulin concentrations for healthy adults ranges between 5-15 μIU/ml (30-90 pmol/l)

for basal levels and 60-90 μIU/ml (360-540 pmol/l) for postprandial levels [257].

Circulating insulin binds to the insulin receptor in liver, muscle, and adipose tissues. As a

result of insulin binding to the receptors signals are mediated to initiate the translocation

GLUT4 transporters intracellular vesicles that are exposed to extracellular surface as

result of fusion with plasma membrane of the cells [255], this results in increase glucose

diffusion into cytosol and will be utilized as source of energy by the cells via glycolysis

or stored as gylycogen or triglyceride [258]. In addition to glucose translocation from

plasma to cell, insulin has several other significant roles in metabolic homeostasis. It also

inhibits the breakdown of fat, glycogen and protein and stimulates the synthesis of

64
CHAPTER 2 BACKGROUND

protein in muscle by increasing the amino acids uptake, regulates gene transcription,

increases glycogen and fatty acid synthesis, and inhibits the breakdown of glycogen, fat,

and protein for energy [255].

In fasting state plasma insulin level is low, in this state liver synthesizes glucose through

glycogenolysis and pyruvate pathway thus releasing the glucose [258]. Increased levels

of insulin after the meal in the hepatic portal vein, inhibits the glucose production via

these processes by the liver [259].

2.14.7.3 Insulin Regulation

Insulin has short plasma half -life of only 5- 6 minutes [255]. Insulin is degraded in liver,

kidney, and muscle tissues [260]. About 50 % of insulin secreted reaches general

circulation as it is extensively degraded by the liver. The Insulin is cleared off from blood

through renal clearance. Some insulin degradation occurs in peripheral tissues following

internalization into the cells, but this accounts for only a small amount of insulin

elimination. When injected subcutaneously, insulin first enters the general circulation,

thus having its initial effect on peripheral tissues. This does not mimic the natural process

of insulin secretion and the biological effect is different.

65
CHAPTER 2 BACKGROUND

Figure 2.13:Diagram of the insulin and glucose regulation model

66
CHAPTER 3 MATERIALS AND METHOD

CHAPTER 3
MATERIALS AND METHODS

67
CHAPTER 3 MATERIALS AND METHOD

3. Materials and Methods

3.1 Materials

Timolol maleate (TM) purity 97.5% was provided by Schazo Pharma Pvt. Ltd. Lahore,

Rosuvastatin (RST), purity 98.4% was kind gift of Ferozsons Laboratories Pvt. Ltd.

Nowshera, Recombinant Human insulin was a kind gift from Zafa Pharmaceuticals

Karachi, Diclofenac sodium (DS) purity 99.30% (manufactured by Suzhou Ausun

chemical company limited), Methylcellulose (MC) 4000 cps, hydroxy propylylmethyl

cellulose (HPMC, Methocel E5) manufactured by Dow chemical and polyethylene glycol

(PEG 6000) manufactured by Clariant GMBH were the kind gifts of Medicraft

Pharmaceuticals. Pvt, Ltd Peshawar. Voren injection (DS) manufactured by Asian

Continental Pharma, Blotim 0.5% (TM) ophthalmic solution manufactured by Remington

Pharmaceuticals Lahore and Dofnac 0.1% (DS) ophthalmic eye drops manufactured by

Ethical Laboratories Karachi were purchased from local market. Ciprofloxacin,

levofloxacin, naproxen sodium, atenolol, propanolol, and atorvastatin were obtained from

Fluka (Sigma–Aldrich, Oslo, Norway). Methanol, acetonitrile, diethylether,

tetrahydrofuran, chloroform, dichloromethane, n-hexane, ethanol, phosphoric acid, and

triethylamine (HPLC grade), Pluronic F-127 (Poloxamer406), were purchased from

Sigma–Aldrich (Oslo, Norway). Purified water was prepared using a Millipore ultra-pure

water system (Milford, USA).

3.2 Instrumentation

Perkin-Elmer HPLC system (Norwalk, USA), consisted of a pump (series 200), on-line

vacuum degasser (series 200), auto-sampler (series 200), Peltier column oven (series

68
CHAPTER 3 MATERIALS AND METHOD

200), linked by a PE Nelson network chromatography interface (NCI) 900 with UV/VIS

(series 200). The whole HPLC system was controlled by Perkin-Elmer Total chrom

Workstation Software (version 6.3.1). UV/Visible spectrophotometer (Lambda 25, Perkin

Elmer), Centrifuge (Centurion. Scientific Ltd), Shaking water bath B.S.11 Lab

Companion (Jelo Tech Korea), pH meter (Hanna instruments 8417, USA) and Autoclave

HS-60 (Hansuin Medical Co. Ltd Korea).

3.3 Study Design

The present study is designed to develop and evaluate thermo- reversible solution to gel

vehicle for sustained drug delivery through ocular and subcutaneous routes. The study

was carried out in two phases. In the first phase solutions of various concentrations of the

polymers including Poloxamer 407/Pluronic F-127 (P-127), Hydroxypropyl methyl

cellulose (HPMC), Methyl Cellulose (MC) and Polyethylene glycol were prepared alone

and in combination with each other. The formulations were evaluated for solution to gel

transition temperature. The selected formulations that gel below body temperature and

were free flowing at room temperature were further evaluated for rheological behavior

and gel clarity.

In second phase drugs (Diclofenac sodium, Timolol maleate and insulin) were

incorporated in the selected formulations, and were qualitatively evaluated both in-vitro

and in-vivo for sustained release profile. The in-vitro data was analyzed by Minitab® and

MS-Excel and fitted to various mathematical models, while the in vivo study was

processed by pk-summit® softwares.

69
CHAPTER 3 MATERIALS AND METHOD

3.3.1 Preparation of Polymer Solutions

The Polymer solutions of varying concentration (W/V%) alone and in different

combinations of PL-127, MC, HPMC and PEG 6000 were prepared by cold

method[261]. The formulations of the polymer alone and in combination are shown in

Table 3.1.Briefly, accurately weighed polymers were stirred in the calculated amount of

cold (4oC) distilled water. The dispersions were then stored in a refrigerator with

occasional stirring until clear solutions were obtained.

70
CHAPTER 3 MATERIALS AND METHOD

Table 3.1: Composition of polymer solutions for screening

PL F127 MC HPMC PEG Distilled


S.No Formulation
(gm) (gm) (gm) (gm) Water
1. P5 5.0 ----- ----- ----- qs to 100 ml
2. P10 10.0 ----- ----- ----- qs to 100 ml
3. P15 15.0 ----- ----- ----- qs to 100 ml
4. P18 18.0 ----- ----- ----- qs to 100 ml
5. P20 20.0 ----- ----- ----- qs to 100 ml
6. P23 23.0 ----- ----- ----- qs to 100 ml
7. P25 25.0 ----- ----- ----- qs to 100 ml
8. P30 30.0 ----- ----- ----- qs to 100 ml
9. H1 ----- ----- 1.0 ----- qs to 100 ml
10. H2 ----- ----- 2.0 ----- qs to 100 ml
11. H3 ----- ----- 3.0 ----- qs to 100 ml
12. H4 ----- ----- 4.0 ----- qs to 100 ml
13. H5 ----- ----- 5.0 ----- qs to 100 ml
14. H6 ----- ----- 6.0 ----- qs to 100 ml
15. H8 ----- ----- 8.0 ----- qs to 100 ml
16. H 10 ----- ----- 10.0 ----- qs to 100 ml
17. M1 ----- 1.0 ----- ----- qs to 100 ml
18. M2 ----- 2.0 ----- ----- qs to 100 ml
19. M3 ----- 3.0 ----- ----- qs to 100 ml
20. M4 ----- 4.0 ----- ----- qs to 100 ml
21. M5 ----- 5.0 ----- ----- qs to 100 ml
22. PM 10/3 10.0 3.0 ----- ----- qs to 100 ml
23. PM 15/3 15.0 3.0 ----- ----- qs to 100 ml
24. MPG 1.5/2 ----- 1.5 ----- 2.0 qs to 100 ml
25. MPG 1.5/5 ----- 1.5 ----- 5.0 qs to 100 ml
26. MPG 1.5/10 ----- 1.5 ----- 10.0 qs to 100 ml
27. MPG 3/2 ----- 3.0 ----- 2.0 qs to 100 ml
28. MPG 4/2 ----- 4.0 ----- 2.0 qs to 100 ml
29. HPG 1.5/2 ----- ----- 1.5 2.0 qs to 100 ml
30. HPG 1.5/5 ----- ----- 1.5 5.0 qs to 100 ml
31. HPG 1.5/10 ----- ----- 1.5 10.0 qs to 100 ml
32. HPG 3/2 ----- ----- 3.0 2.0 qs to 100 ml
33. HPG 6/2 ----- ----- 6.0 2.0 qs to 100 ml
34. HPG 10/2 ----- ----- 10.0 2.0 qs to 100 ml

P=Pluronic F127, M= Methyl cellulose 5cps, H= Hydroxy propyl methylcellulose 15 cps,


PG= polyethylene glycol 6000

71
CHAPTER 3 MATERIALS AND METHOD

3.3.2 Measurement of the Sol–Gel Transition Temperature (Tsol-gel)

The Tsol–gel of the preparations was determined by tube inversion method [27, 121]. 2

ml aliquot of each prepared solution was transferred to a 5 ml test tube in a digital

Shaking water bath B.S.11 Lab Companion (Jelo Tech Korea) maintained at 15oC and

sealed with a parafilm. The temperature of the bath was increased in increments of 3oC

(or 1oC in region of sol–gel transition temperature) upto 50oC. At each temperature point

the solution was allowed to equilibrate for 5 minutes. The samples were checked for

gelation by inverting the test tube at 90o, the solutions were said to be gel when no

fluidity was observed. Measurements were performed in triplicates and Student’s t-test at

P < 0.05 was performed for statistical significance.

3.3.3 Measurement of Steady Shear Viscosity

The rheological behavior before and after autoclaving of the solutions were studied using

Brookfield viscometer; type DVT-2. A 0.5 ml sample of the solution was placed at the

lower plate of the viscometer. The viscosity was determined at 35±0.1oC using spindle 52

at a shear rate ranging from 5 to 400 rpm. The shear rate (γ) in s−1 and the viscosity (η)

in centipoises (cps) were determined from the instrument readings and fitted to the power

law equation [262].

η = m γn−1 eq. 3.1

The consistency index (m) and the flow index (n) two dimensionless quantities for each

preparation were obtained. If the value for n=1 this indicates Newtonian behavior and if

the value of (n) is less than 1, this corresponds to shear thinning flow. The lower the

value of (n) the more shear thinning will be the preparation [113, 263, 264].

72
CHAPTER 3 MATERIALS AND METHOD

3.3.4 Clarity of the Formed Gel

The clarity of the solutions at various temperatures i.e. at 5oC, 25oC and gelling

temperature was observed visually.

3.3.5 Autoclaving

To study the effect of autoclaving on physical properties of the preparation, the selected

preparations were subjected to autoclaving sterilization conditions[178]. Briefly, screw

cap test tubes containing 10 ml of the preparation were placed in an autoclave HS-60

(Hansuin Medical Co. Ltd Korea). The autoclaving conditions were 121oC, 15 psi, and 20

minutes. The preparations were cool down to room temperature and were evaluated for

their physical properties i.e. Viscosity, clarity of solution and sol–gel transition

temperature, and compared to those before being autoclaved.

3.3.6 Preparation of Drug Polymer Solutions

Solutions of the drugs in selected polymer formulation (formulations that gels around

35oC) were prepared as below.

3.3.7 Preparation of Diclofenac Sodium In-situ Gel Formulations

The drug and polymers were accurately weighed according to the composition for the

formulations as shown in Table 3.2. Diclofenac sodium 5mg/ml solutions with different

polymers alone and in polymers combination were prepared using cold method [261,

265].

73
CHAPTER 3 MATERIALS AND METHOD

The polymer was dispersed in cold water with continuous stirring, the dispersion was

then stored in refrigerator for 24 hours to obtain clear polymer solution, to this solution

diclofenac sodium was added and dissolved with continuous stirring till clear solution is

obtained. The final solutions were sterilized by autoclaved and were evaluated for their

physicochemical properties i.e. Tsol-gel, viscosity, clarity, drug content, in-vitro drug

release and in-vivo pharmacokinetic parameters.

Table 3.2: Composition of Diclofenac Sodium thermoreversible gel

Diclofenac Sodium PL F127 MC PEG Distilled


S.No Formulation
(mg) (mg) (mg) (mg) Water

1. DP18 5.0 180 ----- ----- qs to 1 ml

2. DP20 5.0 200 ----- ----- qs to 1 ml

3. DPM 15/3 5.0 150 30.0 ----- qs to 1 ml

4. DMPG 1.5/10 5.0 ----- 15.0 100.0 qs to 1 ml

5. DMPG 3/2 5.0 ----- 30 20.0 qs to 1 ml

3.3.8 Preparation of Timolol Maleate In-situ Gel Formulations

The drug and polymers were accurately weighed according to the composition for the

formulations as shown in Tables 3.3. TM 5mg/ml solutions with different polymers alone

and in polymers combination were prepared using cold method [261, 265].

74
CHAPTER 3 MATERIALS AND METHOD

The polymer was dispersed in cold water with continuous stirring, the dispersion was

then stored in refrigerator for 24 hours to obtain clear polymer solution, to this solution

TM was added and dissolved with continuous stirring till clear solution is obtained. The

final solutions were sterilized by autoclaving and were evaluated for their

physicochemical properties i.e. Tsol-gel, flowability, clarity, drug content, in-vitro drug

release and in-vivo pharmacokinetic parameters.

Table 3.3: Composition of Timolol maleate thermoreversible gel

Timolol maleate PL F127 MC PEG Distilled


S.No Formulation
(mg) (mg) (mg) (mg) Water

1. TP18 5.0 180 ----- ----- qs to 1 ml

2. TP20 5.0 200 ----- ----- qs to 1 ml

3. TPM 15/3 5.0 150 30.0 ----- qs to 1 ml

4. TMPG 1.5/10 5.0 ----- 15.0 100.0 qs to 1 ml

5. TMPG 3/2 5.0 ----- 30 20.0 qs to 1 ml

3.3.9 Preparation of Human Insulin (recombinant) In-situ Gel Formulations

The drug and polymers were accurately weighed according to the composition for the

formulations as shown in Table 3.4. Insulin 1mg/ml (27.5 IU) solutions with different

polymers alone and in polymers combination were prepared using cold method [261,

265].

75
CHAPTER 3 MATERIALS AND METHOD

The polymer was dispersed in cold water with continuous stirring, the dispersion was

then stored in refrigerator for 24 hours to obtain clear polymer solution, to this solution

recombinant human insulin was added and dissolved with continuous stirring till clear

solution is obtained. The final solutions were sterilized by micro filtration and were

evaluated for their physicochemical properties i.e. Tsol-gel, viscosity, clarity, drug

content, in-vitro drug release and in-vivo pharmacokinetic parameters.

Table 3.4: Composition of Insulin thermoreversible gel

Insulin PL F127 MC Distilled


S.No Formulation
(mg) (mg) (mg) Water

1. IP18 1.0 180 ----- qs to 1 ml

2. IP20 1.0 200 ----- qs to 1 ml

3. IPM 15/3 1.0 150 30.0 qs to 1 ml

3.3.10 Measurement of Tsol- gel DS Formulations

The Tsol–gel of all thermorevesible solution was measured by tube inversion method [27,

121]. 2 ml of each solution was transferred to a 5 ml test tube in a digital Shaking water

bath B.S.11 Lab Companion (Jelo Tech Korea) maintained at gelation temperature and

sealed with a parafilm. The solution was allowed to equilibrate for 5 minutes to effect the

gelation. The samples were checked for gelation by inverting the test tube at 90o.

Measurements were performed in triplicates and Student’s t-test at P < 0.05 was

performed for statistical significance.

76
CHAPTER 3 MATERIALS AND METHOD

3.3.11 Measurement of Steady Shear Viscosity of Diclofenac Sodium Formulations

The steady shear viscosity of the DS in-situ gels was measured using cone and plate

viscometer. A 0.5 ml sample of the solution was applied to the lower plate of the

viscometer. The measurements were made at 37±0.1oC using spindle 52 at a shear rate

ranging from 5 to 400 rpm. The measurements were made before and after autoclaving.

All samples were analyzed in triplicate and for statistical significance student’s t-test at p

< 0.05 was performed.

3.3.12 Clarity of the Formed Gel of DS Formulations

The clarity of the DS solution and formed gels before and after autoclaving of the

formulations was observed visually at 5oC, 25oC and 37oC.

3.3.13 Drug Content of DS Formulations

All samples were analyzed in triplicate for the drug content using validated HPLC

method [266] before and after autoclaving. Only samples with drug content within 100 ±

10% of labeled amount were accepted.

3.3.14 Determination of In-vitro of Drug Release from DS Formulations

Membrane free models are recommended for in vitro evaluation of in-situ gels to be

employed as vehicle for ophthalmic and injectable formulations [100, 267, 268].

Membrane less model was used in the study[269] to evaluate the drug release from the

in-situ solution to gel formulations. The selected DS formulations were subjected to in

vitro drug release profile. From each preparation 2 ml solution containing DS was

transferred into 10 ml tubes. The test tubes containing the solutions were then

77
CHAPTER 3 MATERIALS AND METHOD

equilibrated for 5 minutes to effect the gelation in digital shaking water bath, maintained

at 37oC ± 1. Phosphate buffer pH 7.4 used as dissolution medium (2 ml) was poured

slowly on the surface of the gel not to disturb the surface layer. The water bath was

operated at 50 rpm. Whole of the dissolution medium was collected as sample after

predefined time intervals i.e.0.5, 1.0, 2.0, 4.0, 8.0, 12.0, 24, 36, 48, 72, 96 and 120 hours

depending upon formulation that remain intact for the specified time. As soon as samples

were collected fresh 2ml dissolution medium was added to the test tube.

The samples were suitably diluted and analyzed for diclofenac content using validated

HPLC method[266]. The dissolution samples were taken in triplicate for each

formulation.

3.3.15 Measurement of Tsol- gel of TM Formulations

The solution to gel transition temperature of the prepared formulation was recorded by

tube inversion method [27, 121]. For each formulation 2 ml of each solution was taken in

a 5 ml test tube sealed with parafilm in a digital Shaking water bath B.S.11 Lab

Companion (Jelo Tech Korea) maintained at gelation temperature. The solution was

allowed to equilibrate for 5 minutes to effect the gelation. The samples were checked for

gelation by inverting the test tube at 90o. Measurements were performed in triplicates and

Student’s t-test at P < 0.05 was performed for statistical significance.

3.3.16 Measurement of Steady Shear Viscosity of TM Formulations

The steady shear viscosity of the TM in-situ gels was measured using cone and plate

viscometer. From each formulation 0.5 ml of the solution was applied to the lower plate

78
CHAPTER 3 MATERIALS AND METHOD

of the viscometer. The measurements were made at 37 ± 0.1oC using spindle 52 at a shear

rate ranging from 5 to 400 rpm. The viscosity (η) in centipoises (cps) and the shear rate

(γ) in s−1 were noted and recorded from the instrument reading. The data was compared

with results obtained after sterilization by autoclaving. All samples were analyzed in

triplicate and for statistical significance student’s t-test at P< 0.05 was performed.

3.3.17 Clarity of the Formed Gel of TM Formulations

The clarity of the timolol solution and gel formulations before and after autoclaving was

determined visually at 5oC, 25oC and 37oC.

3.3.18 Drug Content of TM Formulations

All samples were analyzed in triplicate for the drug content using validated HPLC

method[266] before and after autoclaving. Only samples with drug content within 100 ±

10% of labeled amount were accepted.

3.3.19 Determination of In-vitro of Drug Release from TM Formulations

Timolol maleate release from the TM in-situ gel formulations was determined by

membrane less model [269]. The selected TM formulations were subjected to in vitro

drug release profile. 2ml of each preparation was taken in 10 ml test tubes. The test tubes

containing the solutions were equilibrated for 5 minutes to effect the gelation in digital

shaking water bath maintained at 37oC ± 1. Phosphate buffer pH 7.4 as dissolution

medium (2ml) was poured slowly on the surface of the gel not to disturb the surface

layer. The water bath was operated at 50 rpm. After predefined time interval i.e.0.5, 1.0,

2.0, 4.0, 8.0, 12.0, 24, 36, and 48 hours, whole of the dissolution medium was replaced

79
CHAPTER 3 MATERIALS AND METHOD

with fresh dissolution medium as sample. The samples were suitably diluted and

analyzed for TM content using validated HPLC method[266].

3.3.20 Measurement of Tsol- gel Insulin Preparations

The sol–gel transition temperature for the prepared formulations was measured by tube

inversion method [27, 121] before and after sterilization. 2 ml of each solution was

transferred to a 5 ml test tube in a digital Shaking water bath B.S.11 Lab Companion

(Jelo Tech Korea) maintained at gelation temperature and sealed with a parafilm. The

solution was allowed to equilibrate for 5 minutes to effect the gelation. The solutions

were checked for gelation by inverting the test tube at 90o. Measurements were

performed in triplicates and Student’s t-test at P < 0.05 was performed for statistical

significance.

3.3.21 Measurement of Steady Shear Viscosity of Insulin Preparations

The viscosity of the Insulin in-situ gels was measured using cone and plate viscometer

before and after sterilization. A 0.5 ml sample of the solution was applied to the lower

plate of the viscometer. The viscosity was determined at 37±0.1oC using spindle 52 at a

shear rate ranging from 5 to 400 rpm. The viscosity (η) in centipoises (cps) and the shear

rate (γ) in s−1 were recorded from the instrument reading.

All samples were analyzed in triplicate and for statistical significance student’s t-test at

P< 0.05 was performed.

80
CHAPTER 3 MATERIALS AND METHOD

3.3.22 Clarity of the Formed Gel of Insulin Preparations

The clarity of the formed gels before and after sterilization of the formulations was

observed visually.

3.3.23 Drug Content of Insulin Preparations

All samples were analyzed in triplicate for the drug content using HPLC method [270]at

a wavelength of 206 nm before and after sterilization. Only samples with drug content

within 100 ± 10% of labeled amount were accepted.

3.3.24 Determination of In-vitro Drug Release from Insulin Preparations

In vitro insulin release in dissolution medium was determined by membrane less model

[269]. The selected insulin formulations were subjected to in vitro drug release profile.

From each formulation 2ml solution was transferred to 10 ml test tubes. The test tubes

were equilibrated for 5 minutes to effect the gelation in digital shaking water bath

maintained at 37oC ± 1. Phosphate buffer pH 7.4 as dissolution medium was poured

slowly on the surface of the gel not to disturb the surface layer. The water bath was

operated at 50 rpm. Whole of the dissolution medium was collected as sample after

predefined time intervals i.e.0.5, 1.0, 2.0, 4.0, 8.0, 12.0, 24, 36, 48, 72, 96, 120, 144, 168,

192, 216, 240 and 264 hours depending upon formulation that remain intact for the

specified time. As soon as samples were collected fresh 2ml dissolution medium was

added to the test tube.

The samples were suitably diluted and analyzed by HPLC-UV method for insulin content

[270]. The dissolution samples were taken in triplicate for each formulation.

81
CHAPTER 3 MATERIALS AND METHOD

3.3.25 Drug Release Kinetics

The data obtained from in vitro experiments was fitted to various mathematical models

that are employed to assess the drug release kinetics. The results obtained were plotted as

drug released versus time.

3.3.25.1 Zero Order Kinetic Model

The dosage form that follows zero order release kinetics release same amount of drug by

unit of time, which is an ideal release profile independent of drug amount present in the

dosage form. The data obtained was analyzed by employing the equation 3.2. The data

was plotted as cumulative % drug release versus time.

(eq. 3.2)

Where Qt is the amount of drug dissolved in time t, Q0 is the initial amount of drug in the

solution and K0 is zero order release constant.

3.3.25.2 First Order Kinetic Model

This model relates decimal of logarithm cumulative % drug released versus time. The

drug release kinetics is said to be first order if the graphic is linear, indicating that the

drug release is dependent on the drug remaining in the dosage form. The equation for first

order kinetics is given as eq. 3.3.

(eq. 3.3)
.

Where Qt is the amount of drug dissolved in time t, Q0 is the initial amount of drug in the

solution and Kt is first order release constant.

82
CHAPTER 3 MATERIALS AND METHOD

3.3.25.3 Higuchi Model

The model was proposed by Higuchi (1961, 1963) that relates cumulative drug release

versus square root of time as shown in eq. 3.4.

√ (eq. 3.4)

3.3.25.4 Hixson –Crowell Model

The model was developed in 1931 by Hixson-Crowell, which relates cubic root of the

unreleased drug remaining in the dosage form versus time. As given by eq. 3.5.

/ /
(eq. 3.5)

3.3.25.5 Korsmeyer-Peppas Model

This model relates exponentially the drug release to the elapsed time. The equation is

given as eq. 3.6.

(eq. 3.6)

Peppas in 1985 use the following values given in the table 3.5 to interpret the release

mechanism from the polymeric system.

Table 3.5:Interpretation of diffusion release mechanism from polymeric system

Release exponent (n) Drug transport mechanism Rate as a function of time

0.45 Fickian diffusion t-0.5

0.45<n<1.0 Anomalous transport tn-1

1.0 Case II transport Zero order release

Higher than 1.0 Super case II transport tn-1

83
CHAPTER 3 MATERIALS AND METHOD

3.3.26 In Vivo Evaluation of Formulations

3.3.26.1 Animal Handling

Male New Zealand white rabbits weighing 2.5 ± 0.15 kg were used both for subcutaneous

and ophthalmic route. Rabbits were kept individually in standard stainless steel cages, fed

with a commercial laboratory rabbit diet and were freely allowed to water. The animals

were fasted for 12 hours prior to study.

3.3.26.2 Diclofenac Sodium Administration and Sampling

3.3.26.2.1 Subcutaneous Route

Six animals were divided into two groups. Each group was treated with either the

representative DS in situ gel or the commercial diclofenac sodium injection by injecting

2ml of each test formulation subcutaneously to each rabbit. Blood sample of 500 µl from

each animal was collected from marginal ear vein five minutes before injecting the

formulation and at predetermined time intervals i.e. 0.5, 1, 2,4, 8,12, 24, 36, 48, 72, 96

and 120 hours for each formulation. Blood samples were collected in

ethylenediamintetraacetic acid (EDTA) glass tubes and centrifuged at 1600× g for 10 min

at 4oC.The plasma was collected and stored at -20oC until analysis. Diclofenac sodium

content in plasma was determined by using a validated High Performance Liquid

Chromatography (HPLC) technique[266].

3.3.26.2.2 Ophthalmic Route

Rabbits were divided into two groups. Each group was instilled with either the

representative DS in situ gel or the commercial diclofenac sodium eye drop (0.1%, w/v)

84
CHAPTER 3 MATERIALS AND METHOD

by instilling 20 µl of each test formulation into the right eye of each rabbit. After

instillation of drug solution three animals were then killed at the following time intervals:

0.5, 1, 2, 3, 4 and 6 hours for each formulation. 200 µl aqueous humour was withdrawn

with a 27-gauge, 1.3 cm needle attached to 1 ml disposable syringe. The aqueous humour

was stored at -20oC prior to HPLC analysis for Diclofenac sodium content.

3.3.26.3 Timolol Maleate Administration and Sampling

3.3.26.3.1 Ophthalmic Route

Rabbits were divided into two groups. Each group was instilled with either the

representative TM in situ gel or the commercial TM eye drop (0.5%, w/v) by instilling 20

µl of each test formulation into the right eye of each rabbit. After instillation of drug

solution three animals were then killed at predetermined time intervals: 0.5, 1, 2, 3, 4 and

6 hours for each formulation. 200 µl aqueous humour was withdrawn with a 27-gauge,

1.3 cm needle attached to 1 ml disposable syringe. The aqueous humour was stored at -

20oC prior to HPLC analysis for TM content.

3.3.26.4 Insulin Administration and Sampling


3.3.26.4.1 Subcutaneous route

New Zealand White rabbits with initial body weight of 2– 2.5 kg and maintained on

standard laboratory food were injected with10%w/v alloxan (125 mg/kg body weight) via

a marginal ear vein. The diabetic animals were divided into two groups. Each group was

treated with either the representative Insulin in situ gel or the commercial insulin by

injecting 2ml (2U) of each test formulation subcutaneously to each rabbit. Blood sample

of 500 µl from each animal was collected from marginal ear vein five minutes before

85
CHAPTER 3 MATERIALS AND METHOD

injecting the formulation and at predetermined time intervals i.e. 0.5, 1, 2,4, 8,12, 24, 36,

48, 72, 96, 120, 144, 168, 192, 216 and 240 hours for each formulation. Plasma insulin

was determined using insulin enzyme immunoassay kit (Dainabot Co., Ltd., Tokyo,

Japan).

3.3.27 Pharmacokinetic Parameters

To assess different pharmacokinetic parameters like tmax, Cmax, Half-life (t ½), Area under

cure (AUC), Area under moving curve (AUMC), Mean residence time (MRT) and

Volume of distribution (Vd), a non-compartmental model approach was applied[271] to

the data obtained for all the formulations.

3.3.28 HPLC analysis method

3.3.28.1 Materials and methods

3.3.28.1.1 Materials and reagents

Timolol maleate (TM) purity 97.5% (Schazo Pharma Pvt. Ltd. Lahore), rosuvastatin

(RST), purity 98.4% (Feroz Sons Laboratories Pvt. Ltd. Nowshera), diclofenac sodium

(DS) purity 99.0% (Medicraft Pharma. Pvt. Ltd. Peshawar), were the kind gift of local

pharmaceuticals. Ciprofloxacin, levofloxacin, naproxen sodium, atenolol, propanolol, and

atorvastatin were obtained from Fluka (Sigma–Aldrich, Oslo, Norway). Methanol,

acetonitrile, diethylether, tetrahydrofuran, chloroform, dichloromethane, n-hexane,

ethanol, phosphoric acid, and triethylamine (HPLC grade), were purchased from Sigma–

Aldrich (Oslo, Norway). Purified water was prepared using a Millipore ultra-pure water

system (Milford, USA).

86
CHAPTER 3 MATERIALS AND METHOD

3.3.28.1.2 Instrumentation

Perkin-Elmer HPLC system (Norwalk, USA), consisted of a pump (series 200), on-line

vacuum degasser (series 200), auto-sampler (series 200), Peltier column oven (series

200), linked by a PE Nelson network chromatography interface (NCI) 900 with UV/VIS

(series 200). The whole HPLC system was controlled by Perkin-Elmer Total chrom

Workstation Software (version 6.3.1). The data was acquired and quantified by this

software.

3.3.28.2 Preparation of Standard Stock Solutions

Stock solutions of analytes and IS (ciprofloxacin, levofloxacin, naproxen sodium,

atenolol, propanolol, and atorvastatin), each (1 mg/ml) were prepared in acetonitrile and

stored in amber glass vials at −20oC until analysis. Working standards solutions were

prepared in volumetric flasks (10 ml), using mobile phase in the concentration range of

0.05–2 µg/ml of each analyte, keeping IS concentration 1 µg/ml in each sample. The

calibration curves were constructed at seven concentrations levels for the standard

solutions of each analyte. Similarly, a 1:1 mixture containing 1 µg/ml of each analyte and

IS was also prepared.

3.3.28.3 Sample Preparation

3.3.28.3.1 Plasma sample

Blood samples were collected from human volunteers, at Department of Pharmacy,

University of Peshawar (Pakistan), in ethylenediamintetraacetic acid (EDTA) glass tubes

and centrifuged at 1600× g for 10 min at 4oC. The study was approved by the concerned

ethical committee. The plasma was collected and stored at −20oC until analysis. For

87
CHAPTER 3 MATERIALS AND METHOD

sample preparation the plasma was first thawed at room temperature and a volume (200

µl) was spiked with the respective volume in the range of 10–400 µl of standard stock

solution (50 µg/ml), of TM, RST and DS each to prepare their respective dilutions in the

range of 0.05–2 µg/ml at 0.05, 0.100, 0.250, 0.500, 1.00, 1.50, and 2 µg/ml for each

analyte. The equal volume (10 µl) of IS (1 µg/ml), was added to each sample to make its

concentration 1 µg/ml in each sample and vortexed for 3 min. The liquid–liquid

extraction procedure was applied as given in section 3.3.28.4.

3.3.28.3.2 Aqueous Humour Sample

Aqueous humour was collected in borosilicate glass tubes from the bovine eyes. The

collected sample was stored in screw capped air tight glass vials at -20oC till analysis. At

the time of analysis aqueous humour was thawed and a volume (200 µl), was spiked with

the respective volume in the range of (10–400 µl), of the standard solutions of TM, and

DS each to prepare their respective dilutions in the range of 0.05–2 µg/ml at 0.05, 0.100,

0.250, 0.500, 1.00, 1.50, and 2 µg/ml for each analyte. An equal volume (10 µl) of IS (1

µg/ml) was added to each sample to make its concentration 1 µg/ml in each sample and

vortexed for 3 min. The extraction was carried out as given in Section 3.3.28.4.

3.3.28.4 Liquid-Liquid Extraction

Sample (200 µl) was transferred to plastic eppendorf tube (ca≈2 ml), and spiked with

each analyte in its respective concentration range and a constant amount of IS (1 µg/ml)

was added to each sample. The samples were vortex-mixed for 3 min and methanol (600

µl) was added for de-proteination. Extraction was carried out with 1 ml of mobile phase.

The samples were then centrifuged for 5 min at 2000×g and 4oC. After centrifugation the

88
CHAPTER 3 MATERIALS AND METHOD

clear supernatant was transferred to eppendorf tube and the volume was made to 1 ml

with mobile phase. Sample (50 µl) of the supernatant was injected into HPLC for

analysis.

Calibration curves were constructed for all the analytes in the range of 0.05–2 µg/ml at

0.05, 0.100, 0.250, 0.500, 1.00, 1.50, and 2 µg/ml for each analyte using naproxen

sodium (1 µg/ml) as IS in mobile phase, spiked plasma and spiked aqueous humour and

different columns like Hypersil BDS C18 column (250 mm ×4.6 mm,5 µm); Symmetry

C8 (250 mm ×4.6 mm, 5 µm); Discovery HS C18 column (150 mm ×4.6 mm, 5 µm),

Symmetry C8 columns (150 mm ×3.9 mm, 5 µm) were evaluated for separation of drugs

in the mixture.

3.3.28.5 Chromatographic Conditions

The HPLC analysis of the studied compounds was performed using ACN: 0.2% TEA

(60:40, v/v), pH 2.75 adjusted with 85% phosphoric acid, as mobile phase pumped at

flow rate of 1 ml/min, in isocratic mode on Hypersil BDS C18 column (250 mm×4.6

mm, 5 µm). The column oven temperature was kept at 45 oC and the peak response was

monitored at a wavelength of 284 nm. The sample (50 µl) was injected into HPLC

system and the data was acquired using Perkin-Elmer Total chrom Workstation Software

(version 6.3.1).

89
CHAPTER 3 MATERIALS AND METHOD

3.3.28.6 Chromatographic Conditions and Experimental Parameters Optimization

3.3.28.6.1 Selection of Stationary Phase (column)

Different particulate reversed-phase chromatographic columns (stationary phases) such

as; Hypersil BDS C18 column (250 mm×4.6 mm, 5 µm); Symmetry C8 (250 mm×4.6

mm,5 µm); Discovery HS C18 column (150 mm×4.6 mm, 5 µm), Symmetry C8 column

(150 mm×3.9 mm, 5 µm), protected by a Perkin Elmer C18 (30 mm×4.6 mm, 10 µm;

Norwalk, USA), pre-column guard cartridge were tried for the analysis of TM, RST and

DS.

3.3.28.6.2 Mobile Phase Composition

The mobile phase composition was optimized using various organic solvents including

methanol, acetonitrile, tetrahydrofuran, and water in different composition in isocratic

mode for the analysis of the above mentioned compounds. The mobile phase composition

that resulted in a better resolution and shorter analysis time of the studied compounds was

selected as mobile phase for the simultaneous analysis.

3.3.28.6.3 Mobile Phase Flow Rate

The mobile phase flow rate was adjusted in isocratic mode for the analysis of studied

analytes after applying various flow rats in the range of 0.9–2 ml/min.

3.3.28.6.4 Column Oven Temperature

The column oven temperature significantly affects the elution and resolution of different

compounds. The column oven temperature was therefore evaluated in the range of 25–

50oC to show its effects on the analysis of above mentioned compounds.

90
CHAPTER 3 MATERIALS AND METHOD

3.3.28.6.5 Internal Standard

Different compounds including ciprofloxacin, levofloxacin, naproxen sodium, atenolol,

propanolol, and atorvastatin were tried to be used as IS. The compound that showed

better compatibility, best recovery and shorter analysis time was selected as IS for the

suggested method.

3.3.28.6.6 Sample Size

The sample loop size was evaluated in the range of 20–50 µl to adjust the sample size and

minimize the problems like column loading and lack of sensitivity of the mentioned

compounds.

3.3.28.6.7 Detector Wavelength

For simultaneous determination of TM, RST, and DS using naproxen sodium as IS the

detector’s wavelength was evaluated in the range of 280–300 nm. The wavelength that

resulted in the optimal sensitivity and better resolution was chosen as the wavelength for

simultaneous analysis of studied compounds.

3.3.28.7 Validation of the Method

The proposed method was validated according to standard guidelines [272]. The

precision, specificity, sensitivity, linearity, recovery, robustness, stability of solutions and

system suitability parameters were evaluated. The proposed method was validated with

respect to the following parameters.

91
CHAPTER 3 MATERIALS AND METHOD

3.3.28.7.1 Specificity

The specificity of the suggested method was tested through separation of studied

compounds in the mobile phase, 1:1 mixture (containing 1 µg/ml of each analyte and IS),

plasma and aqueous humour samples spiked with appropriate concentration of each

analyte.

3.3.28.7.2 Percent Recovery

Percent recovery was tested to measure the accuracy of the suggested method. The %

recovery was determined from spiked plasma and aqueous humour samples at two

selected concentration levels of each analyte keeping IS concentration same in each

sample. Recovery was calculated according to the following equation:

(eq. 3.7)

Where [A] is the peak area response ratios of the analytes with reference to IS in the

mobile phase; C is the peak area response ratios of the analytes with reference to IS in

spiked plasma/aqueous humour.

92
CHAPTER 3 MATERIALS AND METHOD

Fig. 3.1.Overlay of different representative chromatograms. A = spiked aqueous humour,


B = spiked plasma sample and C = standard solution. Peaks: 1, timolol; 2, rosuvastatin; 3,
naproxen sodium; 4, diclofenac sodium. The chromatograms were obtained at column
oven temperature of 45 oC using mobile phase ACN:0.2% TEA in the ratio of 45:55 v/v
and flow rate of 1.0 ml/min.

3.3.28.7.3 Linearity

The linearity of the proposed method was determined from the calibration curves

constructed at seven concentration levels. Calibration curves were constructed for all the

analytes in the mobile phase, spiked plasma and spiked aqueous humour samples by

plotting their peaks response ratios (ratios of peak areas of analytes to IS) with respect to

their respective spiked concentrations using a linear least squares regression analysis. The

slope (m), intercept (b), and correlation coefficient (r) were calculated from their

respective regression equations.

93
CHAPTER 3 MATERIALS AND METHOD

3.3.28.7.4 Precision

Precision of the method was determined through injection repeatability and analysis

repeatability of spiked plasma and aqueous humour samples. Injection repeatability was

assessed through injecting 10 times the same plasma and aqueous humour spiked with 1

µg/ml of each analyte into the HPLC system. The retention time and peak area of each

analyte were expressed as mean, standard deviation (SD), and covariance (%RSD) as

precision of the suggested method. Analysis repeatability was evaluated from the analysis

of five spiked samples prepared from the same plasma/aqueous humour spiked with 1

µg/ml of each analyte, and the results were expressed as mean, standard deviation (SD),

and covariance (%RSD) of the recovered amount. Intra-day and inter-day studies were

carried out on spiked plasma/aqueous humour samples at 8:00, 16:00,and 24:00 h, for 1

week at alternate days to assess the intermediate precision. The results were expressed as

mean, standard deviation (SD), and covariance (%RSD) of the recovered amount.

3.3.28.7.5 Drug Concentration

The recovered amount was calculated in the form of concentration by the following

equation:

X A
C= × ×CS ×FD (eq. 3.8)
Y B

Where

X = Area of peak of analyte in sample i.e. Plasma/Aqueous humour/invite sample.

Y = Area of peak of internal standard added to the sample.

A = Area of peak of internal standard in 1:1 mixture.

B = Area of peak of the analyte standard in 1:1 mixture.

94
CHAPTER 3 MATERIALS AND METHOD

C = Amount of analyte in1:1 mixture.

FD=Dilution factor

3.3.28.7.6 Limit of Detection and Quantification

The limit of detection (LOD) and limit of quantification (LOQ) for all the analytes were

quantified at a concentration whose signal-to noise ratio (S/N) was three and ten,

respectively. For LOD and LOQ evaluation dilutions of the analytes were prepared in the

ranges of 0.5–5 ng/ml and 5–20 ng/ml for all the analytes. The LOD and LOQ were then

determined from the peaks by the software at signal-to noise ratio (S/N) of three and ten,

respectively.

3.3.28.7.7 Robustness

The robustness of the proposed method was tested through small deliberate changes in

the various chromatographic conditions, like mobile phase composition (±2%), column

oven temperature (±5oC), detector wave length (±2 nm) and flow rate of mobile phase

(0.2 ml/min).Stability studies of standard solutions and spiked plasma/aqueous humour

samples stored at 25oC, 4oC and −20oC were carried out for 1 month.

3.3.28.7.8 Percent Stability

The % stability was calculated by the following equation.

% 100 (eq. 3.9)

Where St is stability of analyte at time t, and S0 is stability at initial time.

95
CHAPTER 4 RESULTS AND DISCUSSION

CHAPTER 4
RESULTS AND DISCUSSION

96
CHAPTER 4 RESULTS AND DISCUSSION

4. Results and Discussion

The objective of the current study was to develop sustained release in-situ solution to gel

polymeric delivery system for the drugs to be administered through subcutaneous or

ocular route. Four polymers (Pluronic F-127, Methyl cellulose, hydroxypropyl

methylcellulose and polyethylene glycol 6000) were used in different concentrations

alone and in combination with each other in different ratios. Total of thirty four (34)

formulations were prepared and evaluated for physical and chemical attributes. All the

formulations were subjected to determine the Tsol-gel, out of which only seven

formulations were evaluated for rheological properties and clarity of the formed gels.

Five In-situ thermorevesible solution-gel formulations were selected for in-vitro drug

release by incorporating the selected drugs. On the basis of in-vitro evaluation selected

drug formulations were subjected to in-vivo evaluation. To quantify the amount of drug

(DS,TM) in the in-vitro and in-vivo samples HPLC –UV method was developed and

validated as per standard guidelines[272].

4.1 HPLC –UV Method for Analysis of Diclofenac Sodium and Timolol Maleate

The method developed was novel in the sense that simultaneous determination of TM,

RST, and DS was carried out for the first time using naproxen sodium as an IS. All the

analytes were separated applying the proposed method in standard mixtures, plasma

samples, and aqueous humour samples as presented in Fig. 3.1. The suggested method

was found accurate and quite specific for the simultaneous analysis of these compounds

in plasma and aqueous humour samples. Complete separation of the target compounds

was achieved in 7 min using the proposed method.

97
CHAPTER 4 RESULTS AND DISCUSSION

4.1.1 Optimization of HPLC Experimental Parameters

Different experimental parameters were optimized in the specified ranges to choose the

optimum mobile phase, stationary phase, mobile phase flow rate, detector’s wavelength,

column oven temperature and pH.

4.1.1.1 Mobile Phase

The mobile phase comprised of ACN: 0.2% TEA in the ratio of 60:40 (v/v), was selected

for the analysis of the above mentioned compounds. The retention times of the studied

compounds decreased with increasing the ratio of acetonitrile (ACN) in the mobile phase.

The overall analysis time decreased significantly with increasing the ACN content.

Figure 4.1:Influence of ACN ratios in the mobile phase on the elution of different analytes. A = having
45% ACN, B = having 50% ACN, C = having 55% ACN, D = having 60% ACN and E = having 70%
ACN. Peaks: 1, timolol; 2, rosuvastatin; 3, naproxen sodium; 4, diclofenac sodium. The chromatograms
were obtained at column oven temperature of 45 oC at a flow rate of 1.0 ml/min.

The effect of ACN on the retention time of DS was greater in comparison with TM and

RST. Increasing the ratio of ACN above 60% resulted in the co-elution of TM peak with

the solvent front although the analysis time decreased appreciably as presented in Fig 4.1.

98
CHAPTER 4 RESULTS AND DISCUSSION

4.1.1.2 Stationary Phase

The optimal stationary phase that resulted in better separation and resolution of the

studied compounds among the various tested stationary phases was selected for the

simultaneous determination of above mentioned analytes. The better separation was

achieved on Hypersil BDS C18 column among the tested columns. The poor resolution of

TM was obtained as TM was co-eluted with the peak of solvent front in case of other

tested columns.

4.1.1.3 Flow Rate

The flow rate of mobile phase greatly affected the analysis of the studied analytes.

Although run time decreased significantly at higher flow rates along with better

resolution of the peaks, however; sensitivities of the analytes decreased as shown in Fig.

4.2.

Figure 4.2:Influence of flow rate of the mobile phase on the elution of different analytes. A = at 0.8
ml/min, B = at 1.0 ml/min, C = at 1.25 ml/min, D = at 1.5 ml/min and E = at 2 ml/min. Peaks: 1, timolol; 2,

99
CHAPTER 4 RESULTS AND DISCUSSION

rosuvastatin; 3, naproxen sodium; 4, diclofenac sodium. The chromatograms were obtained at column oven
temperature of 45 oC using mobile phase ACN:0.2% TEA in the ratio of (60:40, v/v).

The flow rate greatly affected the retention of DS in comparison with other analytes. Co-

elution of TM with solvent front resulted at higher flow rates. The flow rate 1 ml/min was

chosen as optimal flow rate for the simultaneous analysis of these compounds.

4.1.1.4 Column Oven Temperature

Peak sensitivities were not affected significantly by column oven temperature, however,

run time was decreased and resolution of the peaks was increased at higher temperature.

The optimal temperature for the simultaneous analysis chosen was 45oC as shown in Fig.

4.3.

Figure 4.3:Influence of column oven temperature on the elution of different analytes. A = at 50oC, B = at
45oC, C = at 40oC, D = at 35oC, E = at 30oC and F = 25oC. Peaks: 1, timolol; 2, rosuvastatin; 3, naproxen
sodium; 4, diclofenac sodium. The chromatograms were obtained at a flow rate of 1.0 ml/min using mobile
phase ACN:0.2% TEA in the ratio of (60:40, v/v).

4.1.1.5 pH

Higher sensitivities and better resolution of the analytes were achieved at pH 2.75 of the

mobile phase among the different tested pH of the mobile phase as shown in Fig. 4.4. The

100
CHAPTER 4 RESULTS AND DISCUSSION

sensitivity of DS was greatly affected by pH of the mobile phase as compared with other

analytes. Variation in the retention times of the analytes was observed at different pH as

shown in Fig. 4.4.

Figure 4.4:Influence of various pH of the mobile phase on the elution of different analytes. A = at pH
2.5, B = at pH 2.75, C = at pH 3.0 and D = at pH 3.5. Peaks: 1, timolol; 2, rosuvastatin; 3, naproxen
sodium; 4, diclofenac sodium. The chromatograms were obtained at column oven temperature of 45 oC
using mobile phase ACN:0.2% TEA in the ratio of (60:40, v/v), and flow rate of 1.0 ml/min.

4.1.1.6 Wave Length

Detector’s wavelength has been selected after recording the sensitivities of the analytes at

various wavelengths. The greater sensitivities of TM, RST and DS were recorded at 284

nm. Small changes in the retention times of the analytes have been observed as evident in

Fig 4.5.

101
CHAPTER 4 RESULTS AND DISCUSSION

Figure 4.5:Influence of detector wavelengths on the elution of different analytes. A = at 280 nm, B = at
284 nm, C = at 290 nm, D = at 295 nm and E = at 300 nm. Peaks: 1, timolol; 2, rosuvastatin; 3, naproxen
sodium; 4, diclofenac sodium. The chromatograms were obtained at column oven temperature of 45 oC
using mobile phase ACN:0.2% TEA in the ratio of 60:40 (v/v), and flow rate of 1.0 ml/min.

4.1.1.7 Internal Standard

Internal standard (IS) that has shown better sensitivity, recovery, stability and

compatibility with other analytes was chosen among the various tested compounds.

Naproxen sodium was used as IS in these studies as it resulted in better recovery and

good compatibility in comparison with other tested compounds.

4.1.1.8 Sample Preparation

Standard stock solutions of TM, RST, DS and naproxen sodium were prepared in

acetonitrile and working dilutions were prepared in the mobile phase on daily basis.

Extraction from plasma and aqueous humour samples was carried out using different

organic solvents. De-proteination was carried out using methanol and the analytes were

then extracted applying various organic solvents.

102
CHAPTER 4 RESULTS AND DISCUSSION

The mobile phase resulted in better recoveries of the analytes as compared with other

solvents used for extraction. The supernatant separated after centrifugation was directly

injected into HPLC system and complete separation of all the target analytes was

achieved.

4.1.1.9 Method Validation

The developed method was validated in terms of linearity, selectivity, sensitivity,

recovery, precision, and robustness according to standard guidelines. The target

compounds were separated in standard mixtures, plasma and aqueous humour samples

using the proposed method with no extraneous peak. The method was found quite

suitable for the analysis of the above studied analytes.

103
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.1: Validation parameters of HPLC-UV Method

Parameters Analytes

Timolol Maleate Rosuvastatin Diclofenac Sodium

Linearity
0.05-2 µg/ml 0.05-2 µg/ml 0.05-2 µg/ml
Calibration range (µg/ml)
Standard solution
Regression equation y=1.523x+0.023 y=1.592+0.038 y=3.955x+0.010
Correlation coefficient (R2) 0.999 0.999 0.999
Spiked plasma
Regression equation y=1.483x+0.063 y=1.592+0.035 y=3.286x+0.040
Correlation coefficient (R2) 0.999 0.999 0.999
Spiked aqueous humour
Regression equation y=1.443x+0.054 y=3.274x+0.057
Correlation coefficient (R2) 0.999 0.998
Accuracy (%recovery) mean±SD; %RSD mean±SD; %RSD mean±SD; %RSD
Spiked sample (0.05µg/ml) 95.17±1.24; 1.30 97.15±1.03; 1.06 94.77±0.96; 1.01
Spiked sample (1.0µg/ml) 98.30±1.01; 1.08 94.99±1.11; 1.17 96.31±0.73; 0.75
Spiked sample (2.0µg/ml) 97.25±0.33; 0.34 94.03±0.12; 0.13 98.99±0.19; 0.20
Accuracy (amount recovered) mean±SD; %RSD mean±SD; %RSD mean±SD; %RSD
Spiked sample (0.05µg/ml) 0.049±0.0010;2.04 0.48±0.0008; 1.67 0.047±0.0009; 1.91
Spiked sample (1.0µg/ml) 0.986±0.0122; 1.23 0.951±0.0113; 1.19 0.965±0.0119; 1.23
Spiked sample (2.0µg/ml) 1.945±0.0101; 0.52 1.891±0.0090; 0.48 1.981±0.0106; 0.54
Percent recovery (relative)
mean±SD; %RSD mean±SD; %RSD mean±SD; %RSD
(spiked human plasma) spiked sample
98.72±1.19; 1.20 96.04±3.09; 3.22 95.14±1.19; 1.25
(1.0µg/ml)
Percent recovery (absolute) mean±SD; %RSD mean±SD; %RSD mean±SD; %RSD
Spiked Plasma 92.65±2.32; 2.50 94.43±1.58;1.67 93.54±1.38; 1.47
Percent recovery (relative)
mean±SD; %RSD mean±SD; %RSD
(spiked aqueous humor) spiked sample
94.99±0.98; 1.03 98.23±1.13; 1.15
(1.0µg/ml)
Percent recovery (absolute) mean±SD; %RSD mean±SD; %RSD
spiked aqueous humor 92.56±1.72; 1.86 95.85±1.52; 1.59
Repeatability mean±SD; %RSD mean±SD; %RSD mean±SD; %RSD
Injection repeatability
Spiked sample (2.0µg/ml) 2.9±0.02;0.69a 3.8±0.01;0.26a 6.2±0.02; 0.32a
Spiked sample (2.0µg/ml) 27437.33±109.20;0.40b 28328±110.49; 0.30b 77533.33±136.55;0.18b
Analysis repeatability
1.91±0.02; 1.04 1.92±0.02; 0.76
Spiked sample (2.0µg/ml) 1.90±0.01; 0.53
Sensitivity
Limit of detection (ng/ml) 0.800 0.500 0.250
Limit of quantification (ng/ml) 2.000 1.500 1.000

a; retention time (minutes of the analyte), b; peak area of the analyte

104
CHAPTER 4 RESULTS AND DISCUSSION

The linearity of the method was evaluated from the calibration curves of standard

mixtures, spiked plasma and aqueous humour samples constructed at seven concentration

levels of all the analytes in the range of 0.05–2 µg/ml. Calibration curves of standard

mixtures of TM, RST, and DS and their spiked plasma samples and aqueous humour

samples are shown in Fig 4.6a and b, respectively.

Figure 4.6:(a) Calibration curves of timolol, rosuvastatin and diclofenac sodium standard solutions and
spiked plasma samples. (b) Calibration curves of timolol and diclofenac sodium standard solutions and
spiked aqueous humour Note: Each point is a mean of triplicate injections.

The regression equation and their respective correlation co-efficient (r) are given in Table

4.1. Accuracy of the proposed method was determined on the basis of percent recovery at

three concentration levels (0.05, 1 and 2 µg/ml), for TM, RST, and DS. The results are

shown in Table 4.1.

105
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.2: Intra-day and Inter-day studies

Spiked concentration
Concentration recovered (µg/ml)
(µg/ml)
Intra-Day(mean±SD) %RSD Inter day (mean±SD) %RSD
Timolol maleate
1.0 0.9386±0.0095 0.6357 0.9166±0.0368 4.0104
1.5 1.5098±0.0063 0.4170 1.4527±0.0543 3.7410
2.0 1.8933±0.0035 0.1834 1.8552±0.0405 2.1821
Rosuvastatin
1.0 0.9499±0.0111 1.1729 0.9071±0.0334 3.6859
1.5 1.4332±0.0054 0.3779 1.4171±0.0205 1.4473
2.0 1.8675±0.0028 0.1492 1.8421±0.0197 1.0708
Diclofenac sodium
1.0 0.9404±0.0100 1.0586 0.9093±0.0196 2.1580
1.5 1.4800±0.0104 0.7052 1.4395±0.0284 1.9763
2.0 1.9166±0.0146 0.7622 1.8767±0.0197 1.0516

The precision of the method was evaluated through injection repeatability, analysis

repeatability, and intra-day, inter-day studies as shown in Table 4.2. The intra-day co-

efficient of variation (% RSD) was in the ranges of 0.183–0.635, 0.949–1.867, and

0.940–1.910 for TM, RST, and DS, respectively. Similarly, their respective values for

inter-day studies were in the range of2.182–4.010, 1.070–3.685, and 1.052–2.158 for TM,

RST, and DS, respectively.

Figure 4.7:Chromatograms representing LOD and LOQ values of TM, RST and DS.

106
CHAPTER 4 RESULTS AND DISCUSSION

LOD and LOQ values were determined at precision and accuracy of 20% variations for

the evaluation of sensitivity of the suggested method. The respective chromatograms of

LOD and LOQ are given in Fig 4.7. The respective values are given in Table 4.1.

Stability studies were conducted at room temperature (25oC), and at freezer temperature

(−20oC), for the standard mixtures and spiked plasma samples of TM, RST, and DS and

aqueous humour samples spiked with TM, and DS for 1 week, respectively. The results

obtained have shown that these compounds are stable at freezer temperature. The TM and

DS standard solutions as well as spiked samples were degraded at room temperature.

Ruggedness of the proposed method was determined through small deliberate changes in

various experimental parameters and the observed changes in the peak area and retention

times were found non-significant.

107
CHAPTER 4 RESULTS AND DISCUSSION

4.2 Physical Evaluation of In-situ Solution to Gel Formulations

4.2.1 Measurement of the Sol-Gel Transition Temperatures (Tsol-gel)

The Tsol–gel for all the formulations (34) was determined by tube inversion method [27,

121]. The data is shown in table 4.3. Out of 34 formulations seven formulation converts

to gel from free flowing solution (at 25oC) between 30 to 35oC. The transition

temperature obtained were 30oC, 31oC, 32oC, 33oC, 34oC, 34oC and 35oC for MPG 4/2,

P20, M4, P18, PM 15/3, MPG 3/2 and MPG 1.5/10 respectively. The lowest transition

temperature (30oC) was observed for MPG 4/2 and the highest (35oC) for MPG 1.5/10.

The selected formulations were evaluated in triplicate for precise Tsol-gel at observed

transition temperature ± 0.5oC; data is given in table 4.3. The results confirmed the

transition temperature is dependent on the concentrations for the selected polymers, the

transition temperature increases as the polymer concentration decreases [112].

The combination of the polymers results in the reduction of the polymer concentration

maintaining the transition temperature around the body temperature. Pluronic F-127 in

15% W/V concentration has the gelation point above 50oC, while addition of 3% MC

(PM15/3) decreases the Tsol-gel to 34oC. This might be due to increase in the viscosity as

MC is viscosity enhancer and also due to combined micellization phenomenon of the

polymers[273] thus reducing the critical micellization concentration. All the selected

formulation showed thermoreversible gelation i.e. reducing the temperature below the

transition temperature converts the gel formulations to free flowing liquid.

108
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.3: Tsol-gel of various Polymer Formulations

Temperature (oC)
S. No Formulation 15 20 25 28 30 31 32 33 34 35 36 37 40 45 50
1. P5 × × × × × × × × × × × × × × ×
2. P10 × × × × × × × × × × × × × × ×
3. P15 × × × × × × × × × × × × × × ×
4. P18 × × × × × × × ✔ - - - - - - -
5. P20 × × × × × ✔ × × × × × × × × ×
6. P23 × × ✔ × × × × × × × × × × × ×
7. P25 × ✔ - - - - - - - - - - - - -
8. P30 × ✔ - - - - - - - - - - - - -
9. H1 × × × × × × × × × × × × × × ×
10. H2 × × × × × × × × × × × × × × ×
11. H3 × × × × × × × × × × × × × × ×
12. H4 × × × × × × × × × × × × × × ×
13. H5 × × × × × × × × × × × × × × ×
14. H6 × × × × × × × × × × × × × × ×
15. H8 × × × × × × × × × × × × × × ×
16. H 10 × × × × × × × × × × × × × ✔ -
17. M1 × × × × × × × × × × × × × × ×
18. M2 15 20 25 28 30 31 32 33 34 35 36 37 40 45 50
19. M3 × × × × × × × × × × × × ✔ × -
20. M4 × × × × × × ✔ - - - - - - - -
21. M5 × ✔ - - - - - - - - - - - - -
22. PM 10/3 × × × × × × × × × × × × × × ×
23. PM 15/3 × × × × × × × × ✔ - - - - - -
24. MPG 1.5/2 × × × × × × × × × × × × × × ×
25. MPG 1.5/5 × × × × × × × × × × × × × × ×
26. MPG 1.5/10 × × × × × × × × × ✔ - - - - -
27. MPG 3/2 × × × × × × × × ✔ - - - - - -
28. MPG 4/2 × × × × ✔ - - - - - - - - - -
29. HPG 1.5/2 × × × × × × × × × × × × × × ×
30. HPG 1.5/5 × × × × × × × × × × × × × × ×
31. HPG 1.5/10 × × × × × × × × × × × × × × ×
32. HPG 3/2 × × × × × × × × × × × × × × ×
33. HPG 6/2 × × × × × × × × × × × × × × ×
34. HPG 10/2 × × × × × × × × × × × ✔ × × ×
P=Pluronic F127, M= Methyl cellulose 5cps, H= Hydroxy propyl methylcellulose 15 cps, PG=
polyethylene glycol 6000

109
CHAPTER 4 RESULTS AND DISCUSSION

4.2.2 Measurement of Rheological Parameters

The rheological behavior at 25oC and 37oC of the solutions was studied using cone and

plate viscometer (Brookfield viscometer; type DVT-2) results are depicted in table 4.4.

The highest viscosity (238 cps) was observed for the formulation MPG4/2 containing

methylcellulose 4% and 2% polyethylene glycol. The viscosity order was 238 ± 2.9

(MPG4/2), 222.7 ± 3.1 (M4), 130.3 ± 0.5 (PM15/3), 128.3 ± 2.5 (P20), 109.3 ± 0.5 (P18),

100 ± 0.8 (MPG3/) and 97.7 ± 1.2 (MPG1.5/10). The addition of viscosity enhancer 3%

MC to Pluronic F-127 produce solution of comparable viscosity while reducing the

Pluronic concentration from 20% to 15%. The increase in viscosity was linear with rise in

the temperature; all the formulations were in gel form at 37oC. The recorded viscosities

are 533 ± 2.1 (MPG4/2), 334.7 ± 1.2 (M4), 517.3 ± 1.9 (PM15/3), 310.7 ± 1.2 (P20),

341.7 ± 1.7 (P18), 292.7 ± 2.5 (MPG3/) and 283.7 ± 1.2 (MPG1.5/10).

The flow index (n) of all the formulations at 25oCwas almost one with very low

consistency index (m) values (Table 4.4), indicating Newtonian flow of the solutions.

Increase in the values of m and decrease in the values of n was observed at high

temperature i.e. at 37oC indicating shear thinning of the formulation. The lower the value

of (n) the more shear thinning will be the preparation [113, 263, 264]. The lowest n value

(0.1324±0.003) was obtained for the P18 formulation (Figure 4.8).

110
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.4: Rheological Data of Selected Formulations

25oC 37oC

Consistency Consistency
Viscosity Flow Index Viscosity Flow Index
S. No Formulation Tso-gel (oC) Index Index
Cps (n) Cps (n)
(m) (m)

1. MPG 4/2 29.8±0.05 238.0±2.9 0.8800±0.007 413.3±1.25 553.3±2.1 0.2859±0.003 200326±136

2. P20 31.0±0.08 128.3±2.5 0.9079±0.004 378.7±2.05 334.7±1.2 0.2567±0.004 170063±65

3. M4 32.1± 0.05 222.7±3.1 0.9040±0.006 388.7±1.25 517.3±1.9 0.2377±0.004 181026±48

4. P18 32.8±0.05 109.3±0.5 0.9278±0.003 322.3±1.89 310.7±1.2 0.1324±0.003 138948±47

5. PM15/3 33.9±0.05 130.3±0.5 0.9470±0.005 348.3±2.05 341.7±1.7 0.1542±0.003 122764±93

6. MPG3/2 33.7±0.05 100.0±0.8 0.9682±0.002 302.7±2.87 292.7±2.5 0.1945±0.004 118419±82

7. MPG1.5/10 35.0±0.09 97.7±1.2 0.9904±0.002 354.7±3.30 283.7±1.2 0.2690±0.002 149776±227

n=3

The addition of MC to pluronic solution and polyethylene glycol to MC supplemented

the hydrogen bonding thus comparable Tsol-gel and gel strength was obtained with

decreased concentration of pluronic (15%) and MC (3%) [268]. The formulation M4 and

MPG 4/2 were excluded from further studies as the solutions were highly viscous and

have poor syringe-ability.

111
CHAPTER 4 RESULTS AND DISCUSSION

25oC

37oC
1.0

Flow Index (n)


0.8

0.5

0.3

0.0

Figure 4.8: Flow index (n) of selected formulations

4.2.3 Clarity of the Formed Gel

Solutions were checked for clarity at various temperatures i.e. at 5ºC, 25ºC and 37ºC. All

the solutions were clear and transparent at 5ºC and 25ºC, the gel formed for pluronic

formulation was transparent and clear while the MC forms turbid gel, the gel formed by

combination of pluronic and MC (PM15/3) was semitransparent (Fig. 4.9)

1 2

Figure 4.9: Appearance of solution at 25oC=1 and 37oC=2. A=P18, B=PM15/3,


C=MPG1.5/10, D=MPG3/2 and E=P20.

112
CHAPTER 4 RESULTS AND DISCUSSION

4.2.4 Autoclaving

Viscosity, clarity and Tsol-gel of the preparations at 25ºC before and after autoclaving

were determined, data given in table 4.5.

Table 4.5: Physical Evaluation of Selected Formulation before and after autoclaving

Before Autoclaving After Autoclaving

Sol-gel Sol-gel
Viscosity Clarity of Viscosity Clarity of
S.No Formulation Temp Temp
At 25oC gel At 25oC gel
(oC) (oC)

1. P20 128.3±2.5 +++++ 31.0±0.08 129.1±1.5 +++++ 31.2±0.04

2. P18 109.3±0.5 +++++ 32.8±0.05 108.9±2.1 +++++ 32.6±0.07

3. PM 15/3 130.3±0.5 +++ 33.9±0.05 130.8±1.8 +++ 34.1±0.08

4. MPG3/2 100.0±0.8 ---- 33.7±0.05 101.3±0.8 ---- 33.8±0.07

5. MPG1.5/10 97.7±1.2 ---- 35.0±0.09 97.9±1.6 ---- 35.2±0.06

+++++= good Transparency, +++ =Fair Transparency, ----- Turbid

Autoclaving did not show any significant effect on the viscosity; clarity and Tsol-gel of

the solutions. The results are consistent with the earlier studies [99, 274, 275].The

preparations were free flowing at 25ºC before and after autoclaving.

113
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5 In-vitro Evaluation of In-situ Drug Formulations

4.2.5.1 In-vitro Evaluation of In-situ Diclofenac Sodium Sol-Gel Formulations

4.2.5.1.1 Tsol- gel of DS Formulations

Transition temperature data from sol-gel of the prepared in-situ thermoreversible gels is

depicted in Table 4.6. The data indicates that addition of diclofenac sodium in the

polymer solutions results in slight increase of the transition temperature. This might be

due to the interference in the micellization of these polymers by altering the dehydration

of the hydrophobic polyoxyethylene (PO) block [97, 113, 114]. The data in table 4.6

shows the effect of autoclaving on the Tsol-gel. The P-Values (n=3)obtained using

student’s t-test were 0.423, 0.225, 0.425, 0.478 and 1.00 for DPM18, DPM20, DPM15/3,

DMPG 3/2 and DMPG1.5/10 respectively, suggesting that autoclaving has no significant

effect on the transition temperature of the in-situ gels [274].

4.2.5.1.2 Viscosity of DS Formulations

Viscosity of DS in-situ gel (n=3) was measured using cone and plate viscometer

(Brookfield viscometer; type DVT-2), results are shown in Table 4.6.The addition of DS

to the polymer solutions has negligible effect on the viscosity of the solutions. The effect

of autoclaving was also determined which was not significant and is consistent with

previous studies [274], ensuring that autoclaving has no significant effect on the

rheological properties of the gelling polymers.

114
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.6: Physicochemical evaluation of Diclofenac sodium thermoreversible in-situ gels

Drug Drug Viscosity Clarity Clarity


P Viscosity P Tsol-gel Tsol-gel P
S. No Formulation content content Before Before After
value After AC value Before AC After AC value
Before AC After AC AC AC AC

1. DP18 100.63±0.49 95.17±0.59 0.003 119.0±0.8 117.67±1.5 0.057 33.33±0.24 33.67±0.24 0.423 +++++ +++++

2. DP20 100.49±0.47 94.32±0.10 0.003 132.0±0.8 130.7±0.5 0.270 31.0±0.41 31.50±041 0.225 +++++ +++++

3. DPM 15/3 100.63±0.21 94.23±0.35 0.001 145.7±0.9 145.0±0.8 0.529 34.85±0.24 35.17±0.24 0.425 +++ +++

4. DMPG 1.5/10 100.40±0.26 94.53±0.25 0.004 101.7±0.5 102.7±1.2 0.478 36.2±0.24 36.33±0.24 0.478 ---- ----

5. DMPG 3/2 100.30±0.40 94.00±0.70 0.005 113.3±1.2 112.7±0.9 0.270 35.3±0.24 35.33±0.24 1.00 ---- ----

(n=3, mean ±SD), Clarity = +++++ (good), +++ (Fair), ---- (poor)

115
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.1.3 Clarity of the DS Solutions and Formed Gels

Solutions of all the formulations were checked for clarity at 5ºC, 25ºC and gelling

temperature, all the solutions were clear indicating the solubility of the added ingredients

at all the temperature excepts for the solutions containing MC (DMPG 3/2 and DMPG

1.5/10) that formed turbid gels at the gelling temperature. The turbidity may be due to the

hydrophobic interaction of the methoxyl group of the polymer chain [161]. The gel

formed by the combination of pluronic and MC i.e. DMP 15/3 were semitransparent.

Autoclaving sterilization did not affect the clarity of these formulations. The clarity of the

pluronic solutions indicates the surfactant activity of these polymers which help in the

salvation [274], while the addition of PEG (co-solvent) in the MC formulations helps in

the solubility of the drug.

4.2.5.1.4 Drug Content of DS In-situ Gels

The diclofenac sodium contents for the formulations were between100.30±0.40 to

100.63±0.49 and 94.00±0.70 to 94.23±0.35 before and after autoclaving respectively. The

data in Table 4.6 suggests that the diclofenac content decreases significantly after the

sterilization through autoclaving as indicated by the P-Values which were between

0.001to 0.005 for the tested samples. The drug content decreases upto 6% for all the

formulation that is within the official limits [178]. The decrease in % labeled amount of

DS might be due to its degradation by exposure to high temperature during autoclaving.

The results are consistent with the previous studies [276]. Sterility is one of the prime

requirements for both ophthalmic and parenteral delivery system. It is important to

116
CHAPTER 4 RESULTS AND DISCUSSION

employ appropriate technique for sterilization to avoid the degradation of the DS, hence

filtration at room temperature will be an ideal alternate technique for the sterilization.

4.2.5.2 In vitro Drug Release From DS In-situ Gels

Drug release from the DS in-situ solution to gel formulations was determined using

membrane free models [100, 267, 268]. Phosphate buffer pH 7.4 was used as dissolution

medium. Samples were collected at the predefined time intervals and were analyzed for

drug content using validated HPLC method [266].

120.0

100.0
%Cummulative DS release

80.0
DP20
DP18
60.0
DPM15/3

40.0 DMPG1.5/10
DMPG3/2

20.0

0.0
0 12 24 36 48 60 72 84 96 108 120 132 144 156 168 180
Time (Hr)

Fig. 4.10: In-vitro release profile of Diclofenac sodium form various in-situ sol-gel

The release profiles of DS in- situ gels (Fig. 4.10) show that all the formulation retarded

the drug release above twelve (12) hours except for the formulation DMPG 1.5/10. The

t100% (time when 100% drug was released) was 4, 24, 72, 96 and 144 hours for DMPG

1.5/10, DMPG 3/2, DPM 15/3, DP18 and DP 20, respectively. Table 4.7 tabulates the in-

vitro release data of DS from different preparations at various time intervals. The slowest

drug release was observed for the formulation DP 20, that contains 20% pluronic F-127

117
CHAPTER 4 RESULTS AND DISCUSSION

as in-situ thermoreversible gelling polymer. The addition of 3% w/v MC to pluronic in

DPM 15/3 extended the drug release upto 72 hours.

118
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.7: Cumulative % In-vitro Diclofenac sodium release from diclofenac sodium in-

situ gels

Formulations DP18 DP20 DPM15/3 DMPG1.5/10 DMPG3/2

Time (Hr) %drug release %drug release %drug release %drug release %drug release

1 4.56±0.11 3.92±0.17 15.90±0.15 23.66±0.32 15.58±0.36

2 10.68±0.10 9.56±0.31 34.23±0.25 74.60±0.36 36.92±0.18

4 21.75±0.20 13.85±0.22 55.85±0.24 101.44±0.49 62.34±0.56

8 30.52±0.14 18.91±0.20 66.94±0.37 83.75±0.15

12 41.78±0.21 25.66±0.17 74.37±0.31 95.68±0.14

24 52.68±0.16 33.81±0.28 83.94±0.15 103.45±0.28

36 64.48±0.28 42.72±0.22 90.97±0.14

48 75.65±0.13 52.95±0.25 97.04±0.08

72 85.74±0.21 64.56±0.30 100.22±0.25

96 99.76±0.28 76.79±0.24

120 100.50±0.45 89.76±0.23

144 99.36±0.31

168 100.3±0.56

n=3

119
CHAPTER 4 RESULTS AND DISCUSSION

Formulation DMPG 1.5/10 containing 1.5% MC and 10% PEG as thermoreversible

gelling polymer could only extended the drug release for 4.0 hours.

Fig 4.11: Representative chromatogram of in-vitro DS release from DP 20 in-situ gel at


different time interval, A=1 hr, B=12 hrs, C=24 hrs, D=36 hrs, E=72 hrs, F=144 hrs,
G=168 hrs.

Initially no burst release of the drug was observed from the formulation DP 20 and DP18

after the first hour as only 3.92% ± 0.17 and 4.56% ± 0.11of the drug was released,

respectively. DP 20 released only 25% of the loaded drug by 12th hour, 50 % of the drug

was released after 48 hours and then 75% at 96 and 100% drug at 144 hours. The drug

release from DP 18 was 21.75 % (4 hr), 52.68% after 24 hours, 75 % of the drug was

released at 48 hours and t100 was 120 hours. A burst release at first hour for the

formulations DPM 15/3, DMPG 3/2 and DMPG 1.5/10 was observed the drug release

was 15.90%, 15.58% and 23.66%, respectively. The hydrophobic nature [277] of the DS

120
CHAPTER 4 RESULTS AND DISCUSSION

also played the role in retarding the release of drugs from the formulations, DS being less

water soluble will diffuse slowly from the hydrogel formed.

The results obtained showed that increase in the pluronic concentration have positive

effect on sustaining the drug release from the in-situ gels i.e. with increase in polymer

concentration the release profile reduces[278]. The higher the concentration of the

polymer longer the path length for diffusion, also the mesh size of the polymer network is

smaller with higher polymer concentration. With increasing the polymer concentration

the water penetration is slow within the hydrogel which causes reduction in the polymer

erosion and degradation. All these factors accounts for slow drug release from the

concentrated polymer system as compared to less polymer concentration [278].

4.2.5.3 Drug Release Kinetics from DS In-situ Gels

The drug release from the thermoreversible gels are governed by various parameters.

Solubility of the drug in the polymer and water, water diffusion rate into the polymer gel,

the drug diffusion rate from the polymer gel and dissolution of the polymer within the

experimental conditions are all important factors that affect the kinetics of drug release.

Initially the drug is released through Fickian diffusion followed by dissolution [122].

The data obtained from the in-vitro experiments were fitted to different mathematical

model i.e. Zero order, First Order, Higuchi, Hixson-Crowell, Korsmeyer Pappas, to

predict the kinetics and release mechanism of the drug.

121
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.3.1 DP 20 In-situ Gel

The in-vitro dissolution results obtained from membrane less technique for the

formulation DP20 was fitted to the above mathematic models. The release constant and

regression coefficient (R2) values obtained from the mathematical models Zero Order (eq.

3.2), First Order (eq. 3.3), Higuchi model (eq. 3.4), Hixson Crowell model (eq. 3.5) and

Korsmeyer Pappas model (eq.3.68) are shown in Table 4.8.

Table 4.8: Release Parameters of Diclofenac Sodium from DP20, DP18, DPM15/3,
DMPG 3/2 and DMPG 1.5/10 in-situ sol-gel formulations.

Formulation Zero Order First Order Higuchi Hixson Crowell Korsmeyer

R2 Koh-1 R2 K1h-1 R2 KH(h-0.5) R2 KHC(h-1/3) n R2

DP20 0.968 0.640 0.814 0.011 0.995 8.428 0.954 0.022 0.421 0.881

DP18 0.899 0.939 0.815 0.021 0.988 10.55 0.958 0.036 0.470 0.960

DPM15/3 0.717 1.373 0.966 0.027 0.839 10.13 0.961 0.060 0.455 0.978

DMPG3/2 0.711 3.389 0.992 0.113 0.870 22.39 0.997 0.249 0.670 0.978

DMPG1.5/10 0.871 24.13 1.000 0.477 0.925 75.65 0.990 1.825 1.291 0.969

It was found that the in-vitro DS release from the DP20 best fits to the Higuchi model

(Fig. 4.14) as the R2 value obtained was the highest (0.995) indicating the drug release

followed Fickian diffusion. Fig. 4.12 indicates that good linearity (R2=0.968) was

122
CHAPTER 4 RESULTS AND DISCUSSION

obtained for Zero Order indicating the drug release is independent of drug concentration.

Hixson – Crowell plot for the data was also constructed (Fig. 4.15), the R2 value was

0.954 indicating polymer erosion and dissolution.

120.0
y = 24.139x + 10.242
R² = 0.8712

100.0 y = 6.8251x + 21.999


R² = 0.8927

y = 1.3733x + 41.731 y = 0.9395x + 20.326


% cumulative DS release

y = 0.6408x + 14.042
R² = 0.7173 R² = 0.8992 R² = 0.9687
80.0

60.0
DP20

40.0 DP18

DMPG3/2

20.0 DMPG1.5/10

DPM15/3

0.0
0 20 40 60 80 100 120 140 160
Time (Hr)

Fig. 4.12: % Cumulative Diclofenac Sodium Release Vs Time (Zero Order Model) from
Diclofenac sodium in-situ thermoreversible sol-gel.

The value of release exponent “n” was 0.421 (Fig. 4.16) by plotting the fraction

diclofenac sodium released vs. log time (Korsmeyer-Pappas), indicating Fickian diffusion

for the release of DS from the in-situ gels.

In current study the in-vitro dissolution study was performed using membrane free

models, hence the drug mechanism is combined effect of Fickian diffusion and the

dissolution of the polymer. Water uptake through diffusion results in the subsequent

dilution of pluronic F-127 micelles governs the drug release. The drug diffusion and

123
CHAPTER 4 RESULTS AND DISCUSSION

polymer dissolution (90%) followed zero order kinetics [267]. Same results were

obtained for pilocarpine, metronidazole and cephelexin [177, 267] using Pluronic F-127

as thermorevesible polymer for sustaining of these drugs.

4.2.5.3.2 DP 18 In-situ Gel

The in-vitro dissolution drug release obtained for the formulation DP18 was fitted to

different mathematical models to explore the drug kinetics and release mechanism. The

values showed best linearity for the Higuchi model (R2= 0.988). Values for regression

coefficient for the various equations are summarized in Table 4.8. The highest value for

R2 (Fig. 4.14) was obtained for Higuchi Model (0.988) followed by Hixson-Crowell

(0.958) Fig. 4.15.

3.00

2.50
Log% DS remaining
Log(100) -Logt

y = 0.0214x - 0.1065
2.00 R² = 0.8151

y = 0.0271x + 0.1746 y = 0.0113x - 0.0858


R² = 0.9661 R² = 0.8148
y = 0.1137x - 0.0437
1.50 R² = 0.9924

DP20
1.00
y = 0.4778x - 0.3606 DP18
R² = 1
DMPG3/2
0.50 DMPG1.5/10
DPM15/3
0.00
0 20 40 60 80 100 120 140 160

-0.50
Time (Hr)

Fig. 4.13: % Log cumulative Diclofenac Sodium Remaining Log(100)-Logt Vs Time


(First Order Model) from Diclofenac sodium in-situ thermoreversible sol-gel.

The release data also showed reasonable linearity for zero order (R2= 0.899), indicating

the drug release is independent of the drug concentration within the gel. The value

124
CHAPTER 4 RESULTS AND DISCUSSION

(0.470) of release exponent “n” obtained with Korsmeyer-Pappas equation, suggests that

drug release from DP18 formulation followed anomalous transport. Hixson – Crowell

plot (Fig. 4.15) also confirms polymer erosion with the (R2= 0.958).

The “K” values for Zero order (0.939), First order (0.021), Higuchi equation (10.55) and

Hixson-Crowell equation (0.036) were higher compared with the DP 20 which were

0.640, 0.011, 8.428, 0.022 respectively. These values indicate that lower the

concentration of the pluronic F-127 the higher will be the diffusion of the drug from gel

matrix and rapid will be the degradation rate of the polymer.

140.0

y = 22.395x + 8.039
R² = 0.8704
120.0 y = 75.657x - 44.755
y = 10.136x + 27.139
R² = 0.8391
R² = 0.9259 y = 10.553x - 0.6132
R² = 0.988
y = 8.4289x - 4.6873
R² = 0.9959
100.0
% cumulative DS release

80.0

60.0 DP20

DP18
40.0 DPM15/3

DMPG3/2
20.0 DMPG1.5/10

0.0
0.0 2.0 4.0 6.0 8.0 10.0 12.0 14.0
Time (Sq.Rt)

Fig. 4.14: % cumulative Diclofenac Sodium release Vs Sq. Rt. Time (Higuchi Model)
from Diclofenac sodium in-situ thermoreversible sol-gel.

Reduction in pluronic F-127 concentration in case of DP 18 results in weak gel

compared with the DP 20, hence increase in the rate of water diffusion and rapid water

125
CHAPTER 4 RESULTS AND DISCUSSION

uptake results in rapid dissolution of the drug and faster diffusion as the path length is

reduced [279]. The decrease in concentration of the pluronic results in the increase in the

number and size of the water channels, hence results in high drug release [280, 281].

4.2.5.3.3 DPM 15/3 In-situ Gel

Formulation DPM 15/3 contains 15 % Pluronic P-F127 and 3% methyl cellulose. Methyl

cellulose was added to reduce the Tsol-gel below the physiological temperature. The

formulation showed reasonable gel strength and sustained the drug release upto 72 hours.

The mathematical models zero order, first order, Higuchi model, Hixson-Crowell model

and Korsmeyer-Pappas model were applied to the dissolution data obtained from in-vitro

dissolution testing, data is shown in Table 4.8. The results inferred that the drug release

follow first order (Figure 4.13) as the value (0.966) of the regression co-efficient was

highest, indicating the drug release was dependent on the drug concentration remaining

within the system. The data also showed good linearity (R2=0.961) for Hixson-Crowell

(Figure 4.15), indicating surface erosion of the in-situ gel. The data was also fitted to

Korsmeyer-Pappas equation by plotting log% drug release as a function of log time (Fig.

4.16), the value of release exponent was 0.455 indicating the drug followed almost

Fickian diffusion from the gel.

Methyl Cellulose is a high molecular weight water soluble polymer, when dissolved in

water the MC solutions are highly viscous which is concentration dependent, the

viscosity of MC is higher compared with the Pluronic solutions. The addition of 3% MC

to 15% Pluronic yield high viscosity solutions (Table 4.5 and 4.6). The resultant high

viscosity is comparable to that of 20% pluronic solutions might be the reason for

126
CHAPTER 4 RESULTS AND DISCUSSION

retarding the drug release from the formulation DPM 15/3. MC forms micelles in the

aqueous solution upon heating thus it has junction of the micelle zone with the pluronic

F-127 [282].

7.0
y = 0.2499x + 0.1223
y = 1.8253x - 1.6349 R² = 0.9976
R² = 0.9902
6.0
y = 0.0607x + 0.6292
R² = 0.9613

5.0
Cube Root % DS remaining

4.0
Wo-Wt

y = 0.036x + 0.1371
R² = 0.9588
y = 0.022x + 0.0441
R² = 0.9547
3.0

DP20
2.0
DP18
DPM15/3
1.0 DMPG3/2
DMPG1.5/10
0.0
0 20 40 60 80 100 120 140 160
Time (Hr)

Fig. 4.15: Cube root% Diclofenac Sodium remaining (Wo-Wt) Vs Time (Hixson-Crowell
Model) from Diclofenac sodium in-situ thermoreversible sol-gel.

The data suggests that the drug diffusion rate was high that may be due to rapid water

uptake that enhance the drug release rate [277] from the formulation compared with the

DPM20 and DPM18.

4.2.5.3.4 DMPG 3/2 In-situ gel

Solutions of 3% MC has Tsol-gel at 40ºC, the addition of 2% polyethylene glycol to the

solution reduces the transition temperature to 34ºC with good viscosity of 110 cps. The

formulation extended the in-vitro release of diclofenac sodium for 24 hours. The

127
CHAPTER 4 RESULTS AND DISCUSSION

regression coefficient “R2” values obtained for various models indicates highest values

for Hixson-Crowell equation (0.997) showing that the drug release follows diffusion as

well as erosion. Good linearity with R2 value of 0.992 (Figure 4.13) for first order was

obtained inferring the concentration dependent drug release from the in-situ

thermorevesible gel.

The value for release exponent (n=0.670) reveals that the drug release follows anomalous

diffusion. The values of release constant “K” for all the equations applied were high

indicating rapid drug release from the combined polymers formulation.

4.2.5.3.5 DMPG 1.5/10 In-situ Gel

This formulation could only hold its integrity in the in-vitro dissolution medium for 4

hours. The viscosity of the solution was upto 100 cps but forms a soft delicate gel, this

might be due to reduction in the MC content. The Tsol-gel (36ºC) was also high.

The R2 value for the Hixson-Crowell was 0.990 suggesting the drug release through

polymer erosion in the dissolution medium. The “N” values obtained by applying the

Korsmeyer-Pappas equation were 1.291 which is above 1 indicating a super case II

release. The value of release exponent suggests that the polymer degrades rapidly as a

result of its dissolution and erosion. The formulation was not suitable for subcutaneous

route but can be used for ocular route.

128
CHAPTER 4 RESULTS AND DISCUSSION

1.20
y = 0.4456x + 0.2249
y = 0.6706x + 0.1861 R² = 0.978 y = 0.4705x - 0.0457
y = 1.2918x + 0.2768 R² = 0.9789 R² = 0.9607
R² = 0.969
1.00

Fraction (Mt/M) DS released


0.80
y = 0.4211x - 0.1036
R² = 0.8813
0.60
DP 20
DP 18
0.40
DMPG 3/2

0.20 DMPG 1.5/10


DPM 15/3

0.00
0.0 0.5 1.0 1.5 2.0 2.5
Log Time

Fig. 4.16: Fraction (Mt/M) Diclofenac Sodium released Vs Log Time (Korsmeyer-
Pappas Model) from Diclofenac sodium in-situ thermoreversible sol-gel.

4.2.5.3.6 Conclusion

In-vitro evaluations of the formulations divulge that autoclaving has no significant effect

on the Tsol-gel, viscosity and clarity of the solutions. The sterilization through

autoclaving significantly decreased the drug content of all the formulations. The in-vitro

drug release data of the prepared formulation indicates that t100 (144 hrs) was obtained for

DP20 while the lowest (4.0 Hrs) was obtained for DMPG1.5/10. The application of

mathematical release models suggest that the formulation DP 20, DP18 follow the

Higuchi model for the drug release suggesting that major portion of the drug is released

through Fickian diffusion. Formulation DPM 15/3, DMPG 3/2 and DMPG 1.5/10 showed

high linearity for Hixson-Crowell model suggesting that the drug release from these in-

situ gels followed diffusion of the drug along with drug dissolution as a result of polymer

erosion.

129
CHAPTER 4 RESULTS AND DISCUSSION

The drug release from the DP 20 and DP 18 was independent of drug concentration as

highest R2 value was obtained for zero order equation. The formulations can deliver the

drug at constant rate for the predetermined time period. While the drug release was

dependant on the drug concentration present in the gel for the formulations DPM 15/3,

DMPG 3/2and DMPG 1.5/10.

The data obtained from the Korsmeyer-Pappas equation suggests that the drug release

from DP20 and DPM15/3 follow diffusion, formulations DP18 & DMPG 3/2 followed

anomalous transport, while drug release from DMPG 1.5/10 followed super case II

mechanism.

130
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.4 In-vitro Evaluation of In-situ TM sol-gel Formulations

4.2.5.4.1 Tsol- gel of TM Formulations

The results obtained for the transition temperature of the TM in-situ gel formulations are

shown in Table 4.9. It can be seen from the data obtained that addition of TM slightly

decreases gelling temperature of the solutions that may be due to hydrophilic nature of

TM, TM might cause modification of the micellization process of poloxamer gels thereby

increase their Tsol–gel [27, 175]. This may be due to the dehydration of polyoxyethylene

(PO) block thus reducing the micellization process and increase polymer entanglement

with each other [97, 113, 114]. Autoclaving has no significant effect on the Tsol-gel of

the TM in-situ thermoreversible gel formulations (p< 0.4-1.0).

4.2.5.4.2 Viscosity of TM Formulations

Addition of TM to the polymer solution has no significant effect on the viscosity of the

solutions as indicated from the data in Table 4.9. P-Values obtained for the effect of

terminal sterilization were 0.300, 0.250, 1.00, 0.472 and 0.86 for the formulations TP18,

TP 20, TPM15/3, TMPG3/2 and TMPG1.5/10, respectively. All the P-Values are non-

significant showing that autoclaving has no significant effect on viscosity of the

formulations. The results obtained are in accordance with previous studies [274].

131
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.9: Physicochemical evaluation of Timolol Maleate thermoreversible in-situ gels

Drug Content Drug content P Viscosity Viscosity P Tsol-gel Tsol-gel P Clarity Clarity
S. No Formulation
Before AC After AC value Before AC After AC value Before AC After AC value Before AC After AC

1. TP18 98.18±0.306 97.22±0.293 0.002 114.0±1.0 112.3±1.5 0.300 33.2±0.3 33.0±0.3 0.423 +++++ +++++

2. TP20 98.63±0.740 98.10±0.830 0.153 127.0±2.0 124.7±0.6 0.250 31.0±0.5 30.7±0.3 0.184 +++++ +++++

3. TPM 15/3 98.88±0.530 98.47±0.470 0.016 135.7±1.2 135.7±0.6 1.000 34.8±0.3 34.8±0.3 1.000 +++ +++

4. TMPG 1.5/10 98.50±0.514 97.90±0.312 0.086 100.0±1.0 99.7±0.6 0.742 35.5±0.5 35.8±0.3 0.529 ---- ----

5. TMPG 3/2 98.67±0.108 98.21±0.226 0.040 109.3±2.1 109.0±1.0 0.860 35.2±0.3 35.2±0.3 1.000 ---- ----

n=3

132
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.4.3 Clarity of TM Solutions and Formed gels

Solutions of all the formulations were checked for clarity at 5ºC, 25ºC and gelling

temperature, Timolol maleate is water soluble drug the solutions obtained for all the

formulations were clear. The gels obtained at the gelling temperature were clear [274]

except for the formulations TMPG3/2 and TMPG 1.5/10 that showed some turbidity.

Terminal sterilization by autoclaving has no effect on the clarity of these formulations.

The results obtained indicate that the addition of TM in the polymer solution has no

effect on the physical appearance of these solutions.

4.2.5.4.4 Drug Content of TM Formulations

The formulations prepared were analyzed with validated HPLC [266] method in triplicate

for the drug content. The assay of the formulations showed that the drug contents were in

the range of 98.18%±0.306 to 98.88%±0.530. Autoclaving has negligible effect on the

TM content of the formulations. The drug content after sterilization by autoclaving were

well above 97%. The data presages that TM is stable at autoclaving conditions.

4.2.5.4.5 In vitro Drug Release from TM Formulations

Timolol maleate release from the prepared formulations was determined by membrane

less model [269]. The TM content was determined using validated HPLC-UV method

[266].

The in-vitro drug release profiles of TM in- situ gels (Fig. 4.17) indicates that formed

gels were strong enough to extend the release of hydrophilic timolol maleate. The

formulation TPM 15/3 extended the release over the longer period of time, while TMPG

133
CHAPTER 4 RESULTS AND DISCUSSION

1.5/10 retarded for about 8 hours. The t100% was 8, 8, 12, 12 and 24 hours for TMPG

1.5/10, TP 18, TMPG 3/2, TP20 and TPM 15/3, respectively.

120.0
% cummulative Timolol Release
100.0

80.0 TP 20
TP18
60.0
TPM 15/3
TMPG 3/2
40.0
TMPG 1.5/10

20.0

0.0
0 4 8 12 16 20 24 28
Time (Hr)

Fig. 4.17: In-vitro release profile of Timolol Maleate form various in-situ
thermorevesible sol-gel (n=3)

The drug release from the in-situ thermoreversible gels after the first hour was in the

order of TMPG 1.5/1>TP18>TP20>TMPG3/2>TPM15/3. All the formulation released

less than 25% (Fig.4.17) of the drug after first hour except for the formulation TMPG

1.5/10 for which a burst release at first hour was 44.90%. The in-vitro release data at

different time intervals depicted in Table 4.10; indicates that the slowest release was

observed for the formulation (TPM 15/3) containing 15% pluronic and 3%

methylcellulose. This might be due to the bonding between the TM and MC polymer and

swelling of the MC in aqueous medium, thus increasing the path length for the diffusion

of both penetrating water and eluting drug from the hydrogel network [283].

The results obtained for the formulation reveals that increase in the pluronic

concentration has positive [278] effect on sustaining the drug release from the in-situ gels

134
CHAPTER 4 RESULTS AND DISCUSSION

i.e. with increase in poloxamer concentration the release profile reduces as the number

and size of the pores decreases within the network.

135
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.10: % Cumulative In-vitro drug release from Timolol Maleate in-situ gels

Formulation TP18 TP20 TPM15/3 TMPG1.5/10 TMPG3/2

%drug release %drug release %drug release %drug release %drug release
Time (Hr)
Mean ± SD Mean ± SD Mean ± SD Mean ± SD Mean ± SD

1 24.82±0.22 23.19±0.25 21.65±0.34 44.90±0.21 22.07±0.25

2 56.02±0.22 49.39±0.38 47.53±0.32 73.55±0.46 50.66±0.44

4 85.88±0.17 73.87±0.34 67.74±0.54 93.20±0.79 73.19±0.57

8 100.45±0.56 88.11±0.65 78.47±0.47 102.27±0.38 89.92±0.81

12 99.36±0.42 86.07±0.46 ---- 100.71±0.89

24 98.00±0.65 ---- ----

(n=3)

136
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.5 Drug Release Kinetics from TM In-situ Gels

The release kinetics and mechanism of drug release from the in-situ gels was evaluated

by fitting the dissolution data shown in Table 4.10 obtained from the in-vitro experiments

to different mathematical model i.e. zero order, first order, Higuchi, Hixson-Crowell,

Korsmeyer Pappas.

4.2.5.5.1 TP 20 in-situ Gel

The release constant (k) and regression coefficient (R2) values obtained from zero order

(eq. 3.2), first order (eq. 3.3), Higuchi model (eq. 3.4), and Hixson-Crowell model (eq.

3.5) and (n) values from Korsmeyer Pappas model (eq. 3.6) are shown in Table 4.11.

Table 4.11: Release Parameters of Timolol Maleate from TP20, TP18, TPM15/3, TMPG
3/2 and TMPG 1.5/10 in situ sol-gel formulations.

Formulation Zero Order First Order Higuchi Hixson Crowell Korsmeyer

R2 Koh-1 R2 K1h-1 R2 KH(h-0.5) R2 KHC(h-1/3) n R2

TP20 0.834 6.147 0.933 0.175 0.924 29.25 0.978 0.286 0.695 0.986

TP18 0.761 7.156 0.999 0.242 0.911 40.38 0.994 0.711 0.852 0.976

TPM15/3 0.698 2.704 0.986 0.065 0.850 17.82 0.933 0.118 0.537 0.967

TMPG3/2 0.836 6.330 0.987 0.123 0.926 30.10 0.932 0.424 0.715 0.987

TMPG1.5/10 0.820 9.811 0.999 0.301 0.864 29.78 0.994 0.723 0.637 0.950

137
CHAPTER 4 RESULTS AND DISCUSSION

Highest linearity with R2 value of 0.978 for the formulation was obtained for Hixson-

Crowell model (Figure 4.22), indicating the drug release follows both erosion and

diffusion. The Korsmeyer-Pappas power law also supplements the release mechanism

being anomalous transport as the value for release exponent is 0.695. Fig. 4.18 and 4.20

represent zero order and first order kinetics fit, respectively for the dissolution data

obtained, indicating good linearity for first order as compared with the zero order, the

value for regression co-efficient “R2” is 0.933 and 0.834, respectively.

120.0
y = 7.1567x + 51.643 y = 6.33x + 33.134
R² = 0.7612 R² = 0.8366

y = 6.1479x + 33.59 y = 2.7048x + 43.586


y = 9.8118x + 30 R² = 0.6985
100.0 R² = 0.8202
R² = 0.8343
%cumulative TM release

80.0

60.0
TP 20

40.0 TP 18

TPM15/3
20.0 TMPG3/2

TMPG1.5/10
0.0
0 5 10 15 20 25 30
Time (Hr)

Fig. 4.18: % Cumulative Timolol maleate Release Vs Time (Zero Order Model) from
TM in-situ thermoreversible sol-gel.

The dissolution data was also plotted for Higuchi model Fig. 4.21, which also showed

good linearity with the R2 value of 0.924 indicating diffusion of the drug from the

formulation.

138
CHAPTER 4 RESULTS AND DISCUSSION

Fig 4.19: Representative chromatogram of in-vitro TM release from TP 20 in-situ gel at


different time interval, A=1 hr, B=4 hrs, C=8 hrs, D=12.

4.2.5.5.2 TP 18 In-situ Gel

The R2 values obtained from the mathematical models indicates that the drug release

from the formulation depends on the concentration of TM remaining in the system, as

best linearity (R2=0.999) was observed for first order equation (Fig. 4.20). The R2value

(0.994) for the Hixson-Crowell (Fig. 4.22) suggests the drug release occurs due to erosion

of the polymeric system. The data was plotted for Korsmeyer-Pappas power law (Fig.

4.23) to better understand the release mechanism. The value of release exponent “n”

obtained was 0.852 suggesting anomalous transport i.e. the drug release from the system

follows diffusion of the drug from the in-situ gels and erosion of the polymer. The

R2value of 0.976 also suggest good linearity for the power law equation.

139
CHAPTER 4 RESULTS AND DISCUSSION

2.5

y = 0.1754x - 0.1236
R² = 0.9337
2.0

% log drug remaining


(Log100-logt)
y = 0.3017x - 0.036
1.5 R² = 0.9996
y = 0.2427x - 0.1227
R² = 0.9998
y = 0.1233x + 0.0337 y = 0.065x + 0.1303
R² = 0.9873 R² = 0.9863 TP20
1.0
TP 18
TPM15/3
0.5 TMPG3/2
TMPG1.5/10
0.0
0 5 10 15 20 25 30
Time (Hr)

Fig. 4.20: % Log cumulative Timolol Maleate Remaining Log(100)-Logt Vs Time (First
Order Model) from TM in-situ thermoreversible sol-gel.

4.2.5.5.3 TPM 15/3 In-situ Gel

Formulation TPM 15/3 contains 15 % Pluronic P-F127 and 3% methyl cellulose. The

formulation extended the drug release upto 24 hours, which is the most among the TM in-

situ gel formulations prepared. The dissolution data compiled in Table 4.10 was fitted to

the mathematical models zero order, first order, Higuchi, Hixson-Crowell and

Korsmeyer-Pappas. Highest linearity (R2=0.986) was obtained for first order equation,

suggesting concentration dependant drug release. The data also showed good linearity

(R2=0.933) for Hixson-Crowell (Fig. 4.22), indicating surface erosion of the in-situ gel.

The value of release exponent (n=0.537) obtained by plotting the fraction of drug

released vs. log time indicates the drug followed anomalous transport from the in-situ gel.

140
CHAPTER 4 RESULTS AND DISCUSSION

120.0
y = 40.383x - 6.3257y = 30.105x + 2.8499
R² = 0.9114 R² = 0.9264
y = 29.252x + 4.1488
100.0 y = 29.785x + 24.551 R² = 0.9246

% cumulative TM Released
R² = 0.8649
y = 17.825x + 20.213
R² = 0.8501
80.0

60.0
TP20
TP18
40.0
TPM15/3
TMPG3/2
20.0
TMPG1.5/10

0.0
0.0 1.0 2.0 3.0 4.0 5.0 6.0
Time (Sq.Rt)

Fig. 4.21: % cumulative Timolol maleate release Vs Sq. Rt. Time (Higuchi Model) from
TM in-situ thermoreversible sol-gel.

The addition of Methyl Cellulose to the pluronic solution increases the viscosity. MC

forms micelles in the aqueous solution when heated thus it has junction of the micelle

zone with the pluronic F-127 [282]. The MC establishes hydrogen bonding with pluronic

[146] and the drug, thus retarding the diffusion and the erosion of the polymer surface.

7.0
%Cube root TM remaining (Wo-Wt)

6.0 y = 0.7232x + 0.0885 y = 0.7111x - 0.3748


R² = 0.9948 R² = 0.9945

5.0 y = 0.424x - 0.0598


R² = 0.9322

4.0
y = 0.2865x + 0.2832
R² = 0.9785 TP20
3.0
y = 0.1189x + 0.6882 TP18
R² = 0.9336
2.0 TPM15/3

1.0 TMPG3/2

TMPG1.5/10
0.0
0 5 10 15 20 25 30
Time (hr)

Fig. 4.22: Cube root% Timolol maleate remaining (Wo-Wt) Vs Time (Hixson-Crowell
Model) from TM in-situ thermoreversible sol-gel.

141
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.5.4 TMPG 3/2 In-situ Gel

This formulation extended the TM release for twelve (12) hours. Table 4.11 shows

regression coefficient “R2” values obtained for various models. The graph obtained by

plotting the log % drug remaining vs. time (Fig 4.20) shows the R2 value (0.987), which

is the highest among the various release models, indicating the drug release is dependent

on drug remaining within the polymer system.

The plot for Higuchi and Hixson-Crowell equations also show good linearity with the R2

values of 0.926 and 0.932, respectively. It can be inferred from the values of regression

co-efficient of Higuchi and Hixson-Crowell that the drug release follows diffusion and

surface erosion of the polymers.

The value for release exponent (0.715) suggests anomalous transport. The values of

release constant “K” for all the equations applied were high indicating rapid drug release

from the combined polymers formulation.

4.2.5.5.5 TMPG 1.5/10 In-situ Gel

The formulation could only hold its integrity in the in-vitro dissolution medium for eight

(8.0) hours. The data presented in table 4.11 obtained from the application of

mathematical models suggests the drug release follow First order kinetics (R2= 0.999).

The R2value for the Hixson-Crowell was 0.994 suggesting the drug release through

surface erosion of the polymer in the dissolution medium. The “n” value obtained by

applying the Korsmeyer-Pappas equation was 0.637. The value of release exponent is

below one (1) indicating anomalous transport from formulation to dissolution media.

142
CHAPTER 4 RESULTS AND DISCUSSION

1.2 y = 0.6957x + 0.2664


R² = 0.9864 y = 0.7158x + 0.2601
R² = 0.9877
y = 0.8529x + 0.2828
R² = 0.9767
1

fraction Mt/M TM released


y = 0.637x + 0.4972 y = 0.537x + 0.284
R² = 0.9504 R² = 0.9675
0.8

0.6
TP 20
TP 18
0.4
TMPG 3/2
TMPG 1.5/10
0.2
TPM 15/3

0
0.0 0.2 0.4 0.6 0.8 1.0 1.2 1.4 1.6
Log Time

Fig. 4.23: Fraction Mt/M Timolol Maleate release Vs Log Time (Korsmeyer-Pappas
Model) from TM in-situ thermoreversible sol-gel.

4.2.5.5.6 Conclusion

In-vitro evaluations of the TM in-situ thermorevesible gel formulations demonstrate that

autoclaving have no significant effect on the Tsol-gel, viscosity, drug content and clarity

of the solutions. The in-vitro drug release data of the prepared formulation indicates that

formulation TPM 15/3 extended the drug release for the longer period of time with t100 of

24 hours. While the formulation TP18 and TMPG 1.5/10 could only retard the drug

release for 8 hours. The application of mathematical release models suggests that all the

formulations followed first order release kinetics. The formulations also showed good

linearity for Hixson-Crowell model indicating the drug release from these in-situ gels

followed diffusion of the drug along with polymer dissolution.

The release exponent “n” values (Fig. 4.23) for all the formulations obtained by applying

Korsmeyer-Pappas equation were higher than 0.5 and lower than 1.0, indicating that the

drug release for all the formulation follows anomalous transport.

143
CHAPTER 4 RESULTS AND DISCUSSION

It can also be determined from the in-vitro release data that all the formulations extended

the drug release between 8-24 hours, suggesting the formulation can be good candidates

for the ocular drug release.

144
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.6 In-vitro Evaluation of In-situ Insulin Sol-Gel Formulations

4.2.5.6.1 Tsol- gel of Insulin Formulations

The sol–gel transition temperature for the prepared formulations was measured by tube

inversion method [27, 121] before and after filtration. Tsol-gel of insulin in-situ gel

formulation IP20, IP18 and IP15/3 is shown in Table 4.12. It can be observed from the

data that the addition of insulin to the polymer system significantly increased the Tsol-gel

[27, 175]. The transition temperature of Pluronic 18%(P18), pluronic 20% (P20) and PM

15/3, MPG 3/2 and MPG 1.5/10 was increased from 32.1ºC, 31ºC, 33.9ºC, 33.7ºC and

35ºC to 34.20ºC, 31.8ºC, 35.5ºC, 40.7ºC and 43.7ºC, respectively.

The Tsol-gel for the formulation IMPG 3/2 and IMPG 1.5/10 was above 37ºC. These

formulations were excluded from further evaluation and study. The increase in transition

temperature is due to the dehydration of polyoxyethylene (PO) block thus reducing the

micellization process and increase polymer entanglement with each other [97, 113, 114].

The results are in accordance with the early studies [115], which suggests that addition of

hydrophobic ingredients to the aqueous solution of polymers forming thermorevesible in-

situ gels increases the Tsol-gel. The filtration has no significant effect on the Tsol-gel of

the insulin in-situ thermoreversible gel formulations.

4.2.5.6.2 Viscosity of Insulin Formulations

The viscosity shows no significant difference on the addition of insulin to the polymer

solutions. The viscosities of the formulations IP18, IP20 and IPM15/3 were in the range

of 115.3, 127.7 and 143.3 before filtration and 115, 127.7 and 144.0 cps after filtration,

145
CHAPTER 4 RESULTS AND DISCUSSION

respectively. The results illustrates that filtration has no significant effect on the viscosity

of the in-situ thermorevesible gels. The P-Values obtained for the viscosity before and

after the filtration were insignificant as shown in Table 4.12. The results obtained are in

accordance with previous studies [274].

4.2.5.6.3 Clarity of Insulin Solutions and Formed Gels

Solutions of all the formulations were checked for clarity at 5ºC, 25ºC and at gelling

temperature, Human Insulin is water insoluble soluble drug the solutions obtained for all

the formulations were clear, confirming the surfactant effect of the pluronic which

favours solubility of insulin [84, 100, 101].The gels obtained at the gelling temperature

were clear [274] except for the formulation IPM 15/3 which formed turbid gel on

transition from solution phase. Filtration sterilization has no effect on the clarity of these

formulations.

146
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.12: Physicochemical evaluation of Recombinant Human Insulin thermoreversible in-situ gels

P P P
Assay Viscosity Tsol-gel Clarity
Value Value Value

Before After Before After Before After Before After


Formulation
Filtration Filtration Filtration Filtration Filtration Filtration Filtration Filtration

IP18 98.72 ±0.42 98.6±0.40 0.192 115.3±1.15 115.0±1.00 0.423 34.2± 0.29 34.2±0.29 1 +++ +++

1
IP20 98.35±0.62 98.3±0.57 0.423 127.7±0.58 127.7±0.58 31.8±0.29 32.0±0.00 0.423 +++ +++
(NS)

IPM 15/3 98.96±0.19 98.8±0.28 0.314 143.3±2.08 144.0±1.73 0.184 35.5±0.00 35.3±0.29 0.423 + +

IMPG 1.5/10 43.7±0.58 43.7±0.58 1

IMPG 3/2 40.7±0.59 40.8±0.29 0.423

(n=3)

147
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.6.4 Drug Content of Human Insulin Preparations

The formulations prepared were analyzed with HPLC-UV method [270] in triplicate for

the drug content at a wavelength of 206 nm. The assay of the formulations IP18, IP20

and IPM15/3 was 98.72 ± 0.42, 98.35 ± 0.62 and 98.96 ± 0.19, respectively. The assay

was also performed after the filtration of the formulation that did not showed any

significant effect on the insulin content of formulations. The P-Values were insignificant

for all formulations.

4.2.5.6.5 Determination of In-vitro Drug Release from Insulin Formulations

In vitro insulin release in dissolution medium was determined by membrane less model

[269]. The selected insulin formulations were subjected to in vitro drug release profile

using phosphate buffer pH 7.4 as dissolution medium. Samples were collected after

predefined time intervals i.e.0.5, 1.0, 2.0, 4.0, 8.0, 12.0, 24, 36, 48, 72, 96, 120, 144, 192,

216, 240 and 264 hours depending upon formulation that remain intact for the specified

time.

The samples were suitably diluted and analyzed for insulin content by HPLC technique

.The dissolution experiment was repeated in triplicate for each formulation. The

formulations with insulin content 100 ± 10% were included in the in-vitro dissolution

studies.

The in-vitro drug release/time profiles of insulin thermoreversible in-situ gels (Fig. 4.24)

indicate that the formulation IPM 15/3 is the most efficient formulation in terms of

148
CHAPTER 4 RESULTS AND DISCUSSION

sustaining the drug release. The formulation IPM 15/3 extended the insulin release for

264 hours (11 days). While the formulations IP18 and IP20 extended the drug release for

36 and 72 hours only.

The drug release after second hour was 3.56%, 10.50% and 32.12% from IPM15/3, IP20

and IP18, respectively. The formulation IP 18 showed a burst release of insulin after first

hour. This burst release is due to the higher transition temperature and week gel strength

as insulin is hydrophobic drug that increases the Tsol-gel and also reduces the gel

strength.

The slowest drug release was attained for the formulation IPM15/3(Figure 4.24), which is

composed of combination of pluronic F-127 15% and methylcellulose 3%. This might be

due to the bonding between the insulin and MC polymer and swelling of the MC in

aqueous medium, thus increasing the path length for the diffusion of both penetrating

water and eluting drug from the hydrogel network [283]. Insulin is also hydrophobic and

large molecular weight drug as compared to diclofenac and timolol, and it might be

entangled in the polymeric matrix.

4.2.5.6.6 Drug Release Kinetics from Insulin In-situ Gels

The release kinetics and mechanism of insulin from thermoreversible sol-gel formulation

was predicted by fitting the dissolution data obtained (Table 4.13) to various

mathematical models. The models applied were zero order, first order, Higuchi time

square root equation, Hixson-Crowell cube root model, and Korsmeyer Pappas power

law.

149
CHAPTER 4 RESULTS AND DISCUSSION

The drug release from the thermoreversible gels are governed by various parameters.

Solubility of the drug within the polymer and water, water diffusion rate into the polymer

gel, the drug diffusion rate from the polymer gel and dissolution of the polymer within

the experimental conditions are all important factors that will affect the kinetics of drug

release.

Pluronic F-127 attains core-shell structure in an aqueous medium. The hydrophilic PEO

block occupies the outer shell region and the hydrophobic PEO block is embedded in the

shell as core.

120.0
% Cummulative Insulin Released

100.0

80.0

IP20
60.0
IP18
40.0 IPM15/3

20.0

0.0
0 24 48 72 96 120 144 168 192 216 240 264 288
Time (hr)

Fig. 4.24: In-vitro release profile of Recombinant Human Insulin form various in-situ
thermorevesible sol-gel (n=3)

Insulin is a hydrophobic drug and mostly resides inside the hydrophobic hydrogel

network. The drug release from the PEO hydrophilic block is through diffusion, while the

150
CHAPTER 4 RESULTS AND DISCUSSION

rest of the drug released is governed by the erosion and degradation of the hydrophobic

PPO domain of hydrogel.

Insulin in aqueous solution exists in various association states such as monomer, dimer,

hexamer and aggregate [284]. It is thought that insulin without zinc exists in aggregate

form inside the in-situ gel. Thus this form of insulin may not diffuse fast from in-situ

gels.

151
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.13: % Cumulative In-vitro drug release from Insulin in-situ gels

Formulation IP18 IP20 IPM15/3

%drug release %drug release %drug release


Time (Hr) Mean ± SD Mean ± SD Mean ± SD

1 32.12±0.12 10.50±0.21 3.56±0.22

2 48.3±0.25 18.24±0.23 6.07±0.34

4 66.6±0.15 25.41±0.28 11.68±0.42

8 83.4±0.31 32.51±0.17 17.42±0.18

12 89.0±0.25 43.09±0.18 23.49±0.15

24 94.1±0.14 52.47±0.21 29.24±0.18

36 100.2±0.17 66.97±0.28 35.03±0.28

48 83.23±0.41 40.81±0.51

72 99.48±0.22 46.11±0.29

96 51.40±0.22

120 56.76±0.24

144 62.33±0.27

168 67.49±0.18

192 74.42±0.09

216 82.43±0.26

240 91.47±0.24

264 99.24±0.18

(n=3)

152
CHAPTER 4 RESULTS AND DISCUSSION

4.2.5.6.7 IP 18 insulin In-situ Gel

The drug release time profile of insulin in-situ gel formulation in Fig 4.24, illustrates that

the formulation IP18 extended the drug release for 36.The regression coefficient “R2” and

release exponent “n” values obtained for the formulations are depicted in Table 4.14. As

evident from the data best linearity (R2=0.930) for drug release from IP 18 was obtained

for Hixson-Crowell model, suggesting that the drug release is the result of degradation of

the polymer. Figure 4.26 indicates that good linearity with “R2” value of 0.900 was

obtained for first order, suggesting drug release dependence on drug remaining within the

hydrogel network.

Table 4.14: Release Parameters of Human Insulin from IP18, IP20 and IPM15/3 in situ
sol-gel formulations.

Model Zero Order First Order Higuchi Hixson-Crowell Korsmeyer

Formulation R2 KoH-1 R2 K1H-1 R2 KH(h-0.5) R2 KHC(h-1/3) n R2

IP 18 0.689 1.22 0.900 0.040 0.826 10.09 0.930 0.109 0.239 0.902

IP 20 0.970 1.142 0.839 0.027 0.991 11.41 0.955 0.046 0.467 0.992

DPM15/3 0.966 0.309 0.723 0.004 0.982 5.580 0.898 0.010 0.503 0.982

Plots for zero order and Higuchi equations are shown in Figure 4.25 and 4.27

respectively. The value of regression co-efficient “R2” is 0.689 and 0.826, respectively.

The value of release exponent (n=0.239) obtained from Korsmeyer-Pappas equation

153
CHAPTER 4 RESULTS AND DISCUSSION

indicates that the drug release follows Fickian diffusion that is in agreement with the first

order.

120.0 y = 1.222x + 62.732


y = 1.1421x + 23.268 y = 0.309x + 17.929
% Cummulative Insulin Released R² = 0.6895 R² = 0.9663
R² = 0.9706
100.0

80.0
IP20
60.0
IP18
40.0
IPM15/3
20.0

0.0
0 24 48 72 96 120 144 168 192 216 240 264 288
Time (hr)

Fig. 4.25: % Cumulative Recombinant Human Insulin Release Vs Time (Zero Order
Model) from different insulin in-situ thermoreversible sol-gel.

4.2.5.6.8 IP 20 insulin In-situ Gel

The in-vitro insulin release at 37ºC from the formulation is compiled in Table 4.13. The

cumulative % released of insulin after 2 and 12 hours from the formulation was 10.50%

and 43.09%, respectively. The slope and regression co-efficient obtained by plotting the

release data to different release models is shown in Table 4.14.The insulin release from

formulation showed best linearity for Higuchi model with R2 value of 0.991, the

formulation also showed good linearity (R2= 0.970) for zero order equation, suggesting

that the drug release is independent of the drug within the polymer network. The value of

release exponent obtained from Korsmeyer-Pappas model is 0.467, indicating the drug

release follow anomalous transport and also supports the drug release is independent of

drug concentration. The R2 (0.955) obtained from Hixson-Crowell suggests that portion

154
CHAPTER 4 RESULTS AND DISCUSSION

of the drug is released through drug diffusion and drug dissolution due to polymer

degradation.

2.5

2.0
y = 0.0408x + 0.3372
%loginsulin remaining

R² = 0.9006
y = 0.0271x - 0.1356 y = 0.0049x - 0.0533
1.5 R² = 0.8396 R² = 0.723
(log100-logt)

IP20
1.0
IP18
IPM15/3
0.5

0.0
0 50 100 150 200 250 300

-0.5
Time (hr)

Fig. 4.26: % Log cumulative Human Insulin Remaining Log (100)-Logt Vs Time (First
Order Model) from Insulin in-situ thermoreversible sol-gel.

4.2.5.6.9 IPM 15/3 insulin In-situ Gel

The formulation consists of 15%w/v Pluronic F-127 and 3%w/v methylcellulose. The

formulation was the most promising among the prepared insulin thermoreversible in-situ

solution to gel formulations. Figure 4.24 indicates that the formulation extended the drug

release for 264 hours (11 days). The values of regression coefficient “R2”, slope and

release exponent “n” are tabulated in Table 4.14. The Figure 4.27 indicates that the

release data best fits to Higuchi model as the R2 value obtained was 0.982 which is the

highest among the released model applied to the data. The R2 value suggests the drug is

released through diffusion. The Zero order release model also showed good linearity

155
CHAPTER 4 RESULTS AND DISCUSSION

indicating the drug release is independent of insulin concentration within the delivery

system.

120.0 y = 10.095x + 45.577


R² = 0.8265
%Cummulative Insulin Released

100.0 y = 11.412x + 1.2957


R² = 0.9915
y = 5.5802x + 0.1792
R² = 0.9822
80.0

60.0

40.0
IP20

20.0 IP18
IPM15/3

0.0
0.0 2.0 4.0 6.0 8.0 10.0 12.0 14.0 16.0 18.0
Sq.Rt Time

Fig. 4.27: % cumulative Human Insulin release Vs Sq. Rt. Time (Higuchi Model) from
Insulin in-situ thermoreversible sol-gel.

The value obtained for the release exponent “n” by application of Korsmeyer-Pappas

equation was higher than 0.45 indicating the anomalous transport of the drug from

hydrogel system.

156
CHAPTER 4 RESULTS AND DISCUSSION

Fig 4.28: Representative chromatogram of in-vitro Insulin release from IPM 15/3 in-situ
gel at different time interval, violet=1 hr, black=12 hrs, blue=96 hrs, orange=264 hrs.

In general, the PF127 gel formation occurs due to the progressive dehydration of the

polymer micelles as temperature increases, leading to increased chain entanglement. This

entanglement was increased by the addition of methylcellulose to the polymer system,

resulting in increase in gel strength and thus reducing the drug release [285].

4.2.5.6.10 Conclusion

The in-vitro release of insulin from the in-situ gel formulation at 37ºC shown in Fig. 4.24,

indicates that the formulation IPM15/3 retarded the drug release for the greater time

period.

As shown in Fig. 4.24 and Table 4.13, IP18 produced the fastest release of insulin among

the formulations studied. The alteration of transition temperatures may alter the diffusion

of drugs in the gels, hence influencing release rates.

157
CHAPTER 4 RESULTS AND DISCUSSION

6.0
y = 0.109x + 0.910
R² = 0.930

5.0

CRT Insulin Remaining


4.0
y = 0.0465x + 0.112
y = 0.0102x + 0.1172

(Wo-Wt)
R² = 0.9557
R² = 0.8984
3.0

2.0
IP20

IP18
1.0
IPM15/3

0.0
0 50 100 150 200 250 300
Time (hr)

Fig. 4.29: Cube root% Human Insulin remaining (Wo-Wt) Vs Time (Hixson-Crowell
Model) from Insulin in-situ thermoreversible sol-gel.

The physicochemical properties of drugs also affect the drug release characteristics from

formulations [283]. As insulin is hydrophobic large molecular protein drug it is slowly

soluble in the penetrating water and thus released slowly and gradually from the in-situ

thermoreversible gel.

The inter-micellar gap decreases with increasing the concentration of PF127, the degree

of swelling also increases with increase in polymer concentration. This results in the

reduction of Tsol-gel and therefore altering the viscosity and the consistency

characteristics of the formulation. This is probably the most important factor influencing

the release characteristics of the drug [261, 286, 287].

158
CHAPTER 4 RESULTS AND DISCUSSION

2.50
y = 0.2391x + 1.66 y = 0.4679x + 1.1121

Log % cumulative Insulin Released


R² = 0.902 R² = 0.9926
2.00
y = 0.5035x + 0.7422
R² = 0.9827

1.50

1.00

IP20
0.50 IP18

IPM15/3
0.00
0.00 0.50 1.00 1.50 2.00 2.50 3.00
Log Time

Fig. 4.30: log %cumulative Human Insulin release Vs Log Time (Korsmeyer-Pappas
Model) from Insulin in-situ thermoreversible sol-gel.

The IPM15/3 formulation was selected for the in-vivo study based on the results obtained

from the in-vitro study.

159
CHAPTER 4 RESULTS AND DISCUSSION

4.2.6 In-Vivo Evaluation of In-Situ Thermoreversible Gels

4.2.6.1 In-vivo Evaluation of DS In-situ gels

The in-situ thermorevesible gels preparations containing diclofenac sodium was

evaluated in-vivo using rabbits as experimental animals to determine various

pharmacokinetic (PK) parameters. The PK parameters were evaluated following

administration through ocular and subcutaneous route.

4.2.6.2 Subcutaneous Route

Formulations DP20, DP18, DPM 15/3, DMPG3/2, DMPG1.5/10 and conventional

commercial diclofenac sodium solutions were injected (5mg/kg) through subcutaneous

route. The plasma drug concentration time profile is shown in Figure 4.23.The data

obtained for the preparations was analyzed using non-compartmental model by PK-

summit® software. The pharmacokinetic parameters determined include; Peak plasma

concentration (Cmax),Time (Tmax) to attain Cmax, area under plasma drug concentration –

Time curve time zero to last sample drawn (AUC0-t), area under curve plasma drug

concentration –Time curve time zero to infinity(AUC0-∞), area under the movement curve

(AUMC0-∞), mean residence time (MRT), volume of distribution (Vd), clearance and

half-life (t1/2).The formulations were compared statistically with diclofenac sodium

conventional injection using student’s t-test at95% significance level.

4.2.6.2.1 DP18 and DP20 DS In-situ gel

The plasma drug concentration at different time intervals after the subcutaneous injection

of different in-situ gel preparations and the conventional parenteral diclofenac solution is

160
CHAPTER 4 RESULTS AND DISCUSSION

shown in Figure 4.31. The pharmacokinetic parameters of DS of these preparations are

shown in Table 4.15.

3
DPL20
DPL18
2.5
DMPG3/2
Plasma Drug Conc. µg/ml

DPM 15/3
2
Ref

1.5

0.5

0
0 20 40 60 80 100 120
Time (h)

Fig 4.31: Plasma Concentration (µg/ml) of Diclofenac sodium at various time intervals
after subcutaneous injection of commercial diclofenac sodium injection and DS in-situ
gels.

The Cmax for DP18, DP20 and conventional parenteral solution was 2.3798 ± 0.16, 2.4017

± 0.109 and 0.671 ± 0.061, respectively. The Cmax values obtained for the formulation

DP18 and DP20 were significantly high. The P-Values of DP 18 and DP 20 with respect

to reference formulation were 0.003 and 0.004, respectively. The Cmax of the in-situ gels

was 3.5 fold higher than the conventional parenteral solution. The higher Cmax may be

due to the gel that remains intact and remains for the longer duration at the site of

absorption releasing the drug continuously at constant rate.

The Tmax was 48 hours for both DP18 and DP20 in-situ gels preparations compared with

the conventional parenteral solution which was 2 hours. The plasma drug concentration

161
CHAPTER 4 RESULTS AND DISCUSSION

vs. time of different in-situ gels and commercial parenteral solution is shown in Figure

4.30. The plasma drug concentration-Time plot of formulations DP18 and DP 20 give

two peak concentrations, the first peak was observed at first hour indicating the burst

release of drug [60], which is due to transition lag time from solution to gel of the in-situ

gels. The second high concentration peak is observed at 48 hours that is the Tmax for

both formulations. This second high concentration peak is due to the dissolution [122]

and degradation of the polymer hydrogel. The in-vivo drug absorption is in accordance

with the in-vitro drug release and confirms that the drug release is anomalous transports

and follows both diffusion and erosion of the polymeric system.

Fig 4.32: Representative chromatogram of in-vivo plasma DS release from DP 20 in-situ


gel at different time interval, A=1 hr, B=24 hrs, C=48 hrs, D=72 hrs, E=96 hrs.

162
CHAPTER 4 RESULTS AND DISCUSSION

The P-Values are also significant for the pharmacokinetic parameters AUC0-t, AUC0-∞

and Cl (Clearance). The AUC, Cl, MRT and biological half-life values clearly suggests

that the in-situ gels have higher bioavailability as compared to reference solution.

The AUC0-t values indicate that bioavailability of diclofenac from DP 18 and DP 20 was

21.5 and 34 fold, respectively, higher than the conventional solution that indicates the

longer systemic release and absorption of the DS from the in-situ-gel formulations. The

relative bioavailability of DS from DP 18 and DP 20 was 227% and 367% higher

compared with reference formulation.

The elimination half-life obtained was in the order of 23.109 ± 9.70, 18.363 ±1.03 and

4.038 ± 0.19 for DP20, DP18 and reference formulation, respectively indicating the

prolong release of the drug from these formulations. Mean residence time (MRT) is the

ratio of AUMC/AUC, the value of MRT was 70.901 ± 13.19 and 50.77 ± 1.36 for DP20

and DP18,respectively, which is higher compared to the conventional preparation (6.953

± 0.31), indicating that the drug is slowly eliminated from the body after administration

of the in-situ gel compared to conventional preparation. The higher MRT value suggests

that the steady state plasma drug concentration was maintained during the studied time

period.

The plasma drug concentration-Time plots of the formulation DP 18 and DP 20 showed

that the plasma drug concentration was in the range of 1.985 ± 0.16 - 2.379 ± 0.17 and

2.045 ± 0.055 - 2.401 ± 0.11 µg/ml through-out the maintenance period i.e. 0-72 hours

163
CHAPTER 4 RESULTS AND DISCUSSION

and 0-96 hours, respectively, which is well above MEC of diclofenac sodium [288]. On

the other hand the conventional commercial parenteral solution could only maintain the

plasma drug concentration for 12 hours.

The rapid decline in the drug plasma concentration of the formulation DP 18 depicted in

the Figure 4.31 as compared to DP20 may be due to rapid degradation rate of the polymer

as the polymer concentration is less in DP18 compared to DP20.

4.2.6.2.2 DPM 15/3 DS In-situ gel

The plasma drug concentration at different time intervals after the subcutaneous injection

of 2 ml (10mg) DPM15/3 in-situ gel preparations and the conventional parenteral

diclofenac solution is shown in Figure 4.31. The formulation DP 15/3 maintained the

steady state drug plasma concentration for 72 hours. It is evident from the Figure 4.31

that the drug concentration at all points is higher in case of in-situ gel compared to the

reference solution. The Figure 4.31 also indicates that there is initial burst release in the

first hour, which is characteristic to in-situ gels because of the solution to gel transition

lag time[60] and the second burst release observed is due to the degradation of the

polymer in the physiological system.

The pharmacokinetic parameters calculated using PK-Summit is given in Table 4.15.

High Cmax (2.724 ± 0.06) was achieved by the formulation compared to the reference

formulation (0.671 ± 0.061).

164
CHAPTER 4 RESULTS AND DISCUSSION

The Tmax observed for the formulation was 48 hours compared to 2 hours of the

commercial conventional formulation. As illustrated in Figure 4.31 there is initial burst

release followed by constant plasma drug concentration. The Cmax attained at 48 hours is

due to the second high release of the drug from the in-situ gels that may be due to the in-

vivo degradation of the polymers.

Moreover higher values for AUC0-t, AUC 0-∞, AUMC0-∞ and MRT were attained for the

in-situ gel compared to reference parenteral solution (Table 4.15). The P-Values were

0.001, 0.001, 0.001, 0.001, 0.001, 0.001 and 0.013 for AUC0-t, AUC0-∞, AUMC0-∞, MRT,

Vd, CL, and Half-life, respectively.

The higher values of these parameters suggest higher bioavailability of the drug from the

thermorevesible gels. The bioavailability from the formulation is 18.6 fold higher than

the reference conventional parenteral solution. The relative bioavailability was 161%

higher for the formulation DPM 15/3 in comparison with reference conventional

formulation.

The volume of distribution (Vd) is significantly high for the reference formulation

compared with the DPM 15/3 in-situ gel formulation; the high Vd value indicates that

extensive distribution of the drug into the body tissues as it was absorbed rapidly due to

availability of high drug concentration at the site of absorption from the conventional

parenteral solution.

165
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.15: Pharmacokinetic parameters of diclofenac sodium commercial formulation and in-situ gel after subcutaneous
administration
Formulation DPL 20 DPL 18 DMPG3/2 DPM15/3 Reference

P P P P
Pk Parmeter Unit Mean ± SD Mean ± SD Mean ± SD Mean ± SD Mean ± SD
Value Value Value Value

E Half-life Hr 23.109±9.70 0.008 18.363±1.03 0.003 9.071±0.76 0.003 6.785±0.07 0.013 4.038±0.19

Cmax (obs) µg/ml 2.4017±0.109 0.004 2.3798±0.16 0.003 2.640±0.01 0.001 2.724±0.06 0.001 0.671±0.061

Tmax (obs) Hr 48±0.0 ---- 48±0.0 ---- 1±0.0 ---- 48±0.0 ---- 2±0.0

AUC(0-t) (obs area) µg-hr/ml 156.61±11.71 0.002 99.197±6.11 0.001 32.50±0.77 0.001 85.757±0.21 0.001 4.608±0.15

AUC (0-∞) µg-hr/ml 202.22±44.68 0.017 124.65±6.95 0.001 35.78±0.44 0.001 88.058±0.12 0.001 5.493±0.09

AUMC (0-∞) µg-hr*hr/ml 14731 ±6097.78 0.005 6329.5±389.96 0.001 815.65±35.86 0.001 3370.8±19.18 0.001 38.19±12.43

MRT (area) Hr 70.901±13.19 0.015 50.775±1.36 0.001 22.793±1.001 0.001 38.279±0.27 0.001 6.953±0.31

Vd (area) / kg ml/kg 802.39±152.59 0.002 1065.3±87.52 0.001 1829.5±161.7 0.000 555.94±6.42 0.001 5304.60±4.56

CL (area) / kg ml/hr/kg 25.486±5.19 0.001 40.19±2.27 0.001 139.735±1.72 0.001 56.780±0.08 0.001 910.190±0.10

Half-life from Vd and CL Hr 23.109±9.70 0.008 18.36±1.03 0.002 9.071±0.76 0.003 6.7851±0.07 0.013 4.038±0.19

n=3

166
CHAPTER 4 RESULTS AND DISCUSSION

The Vd was small in case of DPM 15/3 in-situ gel formulation as the drug concentration

was maintained at steady state this may be due to the equilibrium between the drug

absorption and elimination. DS was slowly released from the gel formulation compared

with the conventional reference formulation that provides 100% of the drug immediately

at the site of absorption.

The elimination rate of the drug from the in-situ gel was significantly low compared to

the reference conventional solution. The MRT was statistically significant; the MRT

value was 38.279 ± 0.27 that was 5.5 times greater than the conventional reference

preparation. The gel remains intact for 48 hours maintaining the steady state

concentration and then degraded rapidly resulting in depletion of the drug and rapid

decline in the plasma concentration.

4.2.6.2.3 DMPG 3/2 DS In-situ Gel

Formulation DMPG 3/2 maintained the plasma drug concentration for 48 hours (Figure

4.31). The drug plasma – time profile shows characteristic initial burst release [60].

Plasma drug concentration as a function of time plot shows (Figure 4.31) that after 12

hours there is rise in the plasma concentration suggesting the dissolution of the polymer

within the physiological system.

The Cmax 2.640±0.01was observed after one hour. The Cmax obtained for the formulation

was significantly high (p< 0.001) compared with commercial parenteral solution. The in-

situ gel formulation shows greater values of AUC0-t, AUC 0-∞, AUMC0-∞, MRT and t1/2

that were statistically significant.

167
CHAPTER 4 RESULTS AND DISCUSSION

The relative bioavailability of the DMPG 3/2 was 65% greater than that of the reference

formulation. This indicates that the bioavailability of the drug from the formulation

DMPG3/2 was 6.5 fold higher compared with the commercial conventional solution. The

half-life was 9.071 ± 0.76, and 4.038 ± 0.19 for DMPG3/2 and reference formulation,

respectively. The half-life of the gel formulation was statistically significant, indicating

availability of the drug for prolong period of time.

The formulation DMPG 3/2 has lower Cmax, AUC, AUMC and MRT values compared

with the other prepared in-situ gel formulations.

4.2.6.2.4 Conclusion

The bioavailability of all in-situ gels was significantly high compared with the reference

conventional parenteral solution. The elimination half-life of the drug obtained for all in-

situ gel formulations was significantly greater than that of conventional solution. The

volume of distribution (Vd) for all the prepared in-situ gels were lower than the Vd of the

conventional reference parenteral solution.

Consequently, the diclofenac sodium content from the administration of conventional

parenteral preparation could not be detected in the plasma after twelve (12) hours

indicating that it was eliminated from the body or below the limit of detection.

The plasma drug–time profile (Fig: 4.31) values suggested that the in-situ

thermorevesible gels prepared with Pluronic, MC and their combination increased the

168
CHAPTER 4 RESULTS AND DISCUSSION

absorption and prolong the elimination half-life of diclofenac sodium resulting in better

bioavailability as compared with the conventional parenteral solution. Among various in-

situ gels formulations DP20 was the most robust to extended the in-vitro drug release for

120 hours that was fully supported by the in-vivo data obtained. The study also revealed

that the combination of pluronic and methylcellulose (DPM15/3) show comparable drug

control and sustainability as that of the pluronic formulations.

4.2.6.3 Ocular Route

The formulations DP20, DP18, DPM 15/3, DMPG3/2 and DMPG1.5/10 were evaluated

for different pharmacokinetic parameters after instillation 50 µl (25µg DS) of each

formulation into the rabbit’s eye. The pharmacokinetic parameters determined include;

peak aqueous humour drug concentration (Cmax),time to attain Cmax (Tmax), area under

curve aqueous humour drug concentration-Time Curve (AUC0-t), area under curve

aqueous humour drug concentration-time curve from time (T = 0) to infinity (AUC0-∞),

area under the movement curve (AUMC0-∞), mean residence time (MRT), volume of

distribution (Vd), clearance and half-life (t1/2).

The concentration of DS determined in rabbits’ aqueous humour samples is shown in

Figure 4.34. The data obtained was analyzed using non-compartmental analysis by PK-

summit® software to calculate the different pharmacokinetic parameters.

The in-situ thermorevesible formulations were compared statistically with diclofenac

sodium 0.1% conventional eye drops using student’s t-test with 95% significance level.

169
CHAPTER 4 RESULTS AND DISCUSSION

4.2.6.3.1 DP18 and DP20 DS In-situ Gel

Aqueous humour drug concentration-Time profile of the DP18 and DP20 following

instillation of eye drops is shown in Figure 4.34.The pharmacokinetic parameters of DS

in aqueous humour of these preparations are depicted in Table 4.16. The data was

statistically evaluated using Minitab 14 and the difference between in-situ gel

preparations and reference formulation was conducted using student t-test at 95%

confidence interval.

Fig 4.33: Representative chromatogram of in-vivo DS release in AH from DP 20 in-situ


gel at different time interval, A=0.5 hr, B=1 hrs, C=2 hrs, D=3 hrs, E=4 hrs, F=6 hrs.

The Cmax obtained was 2.95 ± 0.08, 4.07 ± 0.04 and 0.94 ± 0.07 for DP18, DP20 and

conventional eye drops, respectively. The P-Values were 0.003 and 0.004 for DP18 and

DP20, respectively that shows highly significant difference of Cmax between the in-situ

gel formulations and Reference drug preparation. The Cmax for the in-situ gels were 3.12

and 4.2 folds greater than the conventional eye drops for DP18 and DP20, respectively.

170
CHAPTER 4 RESULTS AND DISCUSSION

The Time to attain Cmax was one hour for DP18 and two hours for DP20 in-situ gels

compared with the conventional eye drop which was half (0.5) hour. The AUC0-t, AUC0-∞

and AUMC0-∞ values for the formulation DP18 was 6.84 ± 0.11, 8.88 ± 0.43 and 36.10 ±

4.65, respectively. The relative bioavailability was 40% and 56% higher for the

formulation DP 18 and DP 20 respectively compared with reference eye drops.

The values of these parameters obtained for the formulation DP20 were not only higher

than the commercial eye drops but also than the DP18 in-situ gel, indicating that higher

the concentration of the polymer more stronger and adhesive the gel will be, thus

increasing the residence and contact time of the formulation within the eye [289].

The aqueous humour drug concentration-Time profile shown in Figure 4.34, indicates the

absorption, distribution and extended elimination phase for the in-situ gels formulations.

The rapid decline of the DS concentration in AH confirms the in-vitro data obtained,

which suggests the release through drug diffusion and polymer erosion [290].

The diclofenac concentration (µg/ml) in aqueous humour for DP18 and DP20 was 0.450

and 0.524 µg/ml, respectively, after 6 hours indicating that the gels are able to maintain

the steady state concentration within the aqueous humour for the period.

171
CHAPTER 4 RESULTS AND DISCUSSION

4.5
4

Aq-Homour Drug Conc. (µg/ml)


3.5
3 Refernce
DMPG3/2
2.5
DP18
2
DP20
1.5
DPM15/3
1
0.5
0
0 1 2 3 4 5 6 7
Time (h)

Fig 4.34: Concentration (µg/ml) of diclofenac sodium in rabbits’ aqueous humour at


various times after instillation of a commercial diclofenac sodium eye drops and DS in-
situ gel formulations

4.2.6.3.2 DPM 15/3 DS In-situ Gel

The formulation consist of 15% Pluronic F-127 and 3% methyl cellulose. The DS

concentration at all points was higher compared with reference eye drops. The data

obtained various Pharmacokinetic parameters calculated with PK-summit software is

shown in Table 4.16.

Aqueous humour drug concentration-Time profile of the DPM 15/3 formulation

following instillation of eye drops is shown in Figure 4.34. The pharmacokinetic

parameters of DS in aqueous humour of the preparation are given in Table 4.16. The data

was statistically evaluated using Minitab 14 and the difference between in-situ gel

preparations and reference formulation was conducted using student t-test at 95%

confidence interval.

172
CHAPTER 4 RESULTS AND DISCUSSION

The Cmax (2.95 ± 0.08) attained at one hour (Tmax) is higher in comparison to the

reference conventional eye drops. The values for AUC0-t, AUC0-∞, AUMC0-∞ and MRT

were significantly high for the in-situ gel as compared with the conventional eye drops.

The relative bioavailability was 32% higher compared with the reference formulation.

This higher bioavailability might be due to the bio-adhesive character of the gel imparted

by MC[291].

The value of MRT was 3.33 ± 0.34 which is higher than the reference formulation (1.83

± 0.11). The greater MRT value also confirms the increased contact time of the gel

formulation compared to the reference formulation. The elimination rate of the drug from

the in-situ gel was significantly low indicating low drainage and high residence of the gel

in the ocular tissue. The elimination half-life were 2.14 ± 0.69 and 0.82 ± 0.14for the

formulation and reference, respectively. The half-life of the in-situ gel formulation was

significantly high with the p-value of 0.013.The formulation DPM15/3 has low relative

bioavailability compared with DP 18 and DP 20 i.e. 0.80 and 0.57 respectively.

4.2.6.3.3 DMPG 3/2 DS In-situ Gel

Aqueous humour drug concentration-Time profile of the DMPG 3/2 following instillation

of eye drops is shown in Figure 4.34.The pharmacokinetic parameters of DS in aqueous

humour of the preparation are given in Table 4.16. The data was statistically evaluated

using Minitab 14 and the difference between in-situ gel preparations and reference

formulation was conducted using student t-test at 95% confidence interval.

173
CHAPTER 4 RESULTS AND DISCUSSION

The drug content-time profile Figure 4.34 shows that the formulation is capable of

maintaining the drug concentration above the MEC of diclofenac for six (6) hours. The

pharmacokinetic data obtained for the in-situ formulation is shown in Table 4.16.The

Figure 4.34 suggests that there is initial burst release resulting in peak plasma

concentration (Cmax) at half (0.5) hour. It can be seen from the in-vitro evaluation of the

formulation that the Tsol-gel was above 35oC, thus the lag time for conversion of solution

to gel was high which may be the cause of the burst release.

The Cmax of the formulation was 1.84 ± 0.008 µg/ml compared with 0.94 ± 0.07 µg/ml

reference formulation. The Cmax value was statistically high, with p-value of 0.001.

The values of AUC0-t, AUC0-∞, AUMC0-∞, MRT and t1/2 were statistically significant

compared with the reference formulation (Table 4.16) that indicates the bioavailability of

the drug from the in-situ gel formulation was higher in comparison to the commercial eye

drops. The relative bioavailability was 22% more for the in-situ gel formulation

compared with the reference formulation.

The MRT was higher for the formulation indicating the high retention time of the

formulation in the eye as compared to the conventional eye drops, providing longer

absorption duration. The clearance rate of diclofenac sodium from the ocular tissue with

the administration of the gel formulation was low compared with the commercial eye

drops, the values are 111.91 and 50.97 for commercial eye drops and DMPG3/2. The bio-

adhesive characteristic of the methyl cellulose increases the residence and contact time of

174
CHAPTER 4 RESULTS AND DISCUSSION

the formulation with in the eye resulting in greater bioavailability from the formulation

compared with the conventional eye drops which are rapidly drained off from the ocular

tissues.

175
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.16: Pharmacokinetic parameters of Diclofenac sodium commercial formulation and in-situ gel formulation after ocular
administration
Formulation DP 18 DP 20 DPM 15/3 DMPG3/2 Reference

Pk Parmeter Unit Mean ± SD P-Value Mean ± SD P-Value Mean ± SD P-Value Mean ± SD P-Value Mean ± SD

E Half-life Hr 3.13±0.36 0.003 2.45±0.40 0.008 2.14±0.69 0.013 1.55±0.07 0.003 0.82±0.14

Cmax µg/ml 2.95±0.08 0.003 4.07±0.040 0.004 2.95±0.08 0.001 1.84±0.08 0.001 0.94±0.07

Tmax Hr 1.00±0.00 ---- 2.00±0.00 ---- 1.00±0.00 ---- 0.50±0.00 ---- 0.5±0.00

AUC(0-t) µg-hr/ml 6.84±0.11 0.001 10.66±0.35 0.002 6.10±0.39 0.001 4.48±0.33 0.001 1.89±0.01

AUC(0-∞) µg-hr/ml 8.88±0.43 0.001 12.51±0.11 0.017 7.19±0.62 0.001 4.94±0.39 0.001 2.24±0.08

AUMC(0-∞) µg-hr*hr/ml 36.10±4.65 0.001 42.00 ±2.41 0.005 24.03±4.33 0.001 13.44±1.26 0.001 4.11±0.39

MRT Hr 4.05±0.33 0.001 3.36±0.22 0.015 3.33±0.34 0.001 2.72±0.04 0.001 1.83±0.11

Vd ml/kg 126.75±8.60 0.001 70.67±12.14 0.002 106.44±27.17 0.001 112.96±3.67 0.001 132.25±17.21

CL ml/hr/kg 28.19±1.35 0.001 19.99±0.18 0.001 34.95±2.93 0.001 50.79±3.98 0.001 111.91±3.89

Half-life from Vd and CL Hr 3.13±0.36 0.002 2.45±0.40 0.008 2.14±0.69 0.015 1.55±0.07 0.003 0.82±0.14

n=3

176
CHAPTER 4 RESULTS AND DISCUSSION

4.2.6.3.4 Conclusion

The bioavailability of all in-situ gels was significantly high compared with the reference

conventional ophthalmic solution. The elimination rate of the drug from all the in-situ gel

formulations was significantly low than that of conventional eye drops.

The diclofenac sodium content from the instillation of conventional commercial eye drop

could not be detected in the aqueous humour after three (3) hours indicating that it was

below the limit of detection or eliminated from the aqueous humour, while the DS

content for the gels was detected for six (6) hours after the instillation. The concentration

of the DS determined with HPLC method for DP20, DP18, DPM15/3 and DMPG3/2

after 6 hours was0.450 ± 0.02, 0.524 ± 0.015, 0.348 ± 0.02 and 0.206 ± 0.02 (µg/ml),

respectively. The concentration of diclofenac achieved at sixth hour were above the MEC

level of the diclofenac sodium [288].

The in-vivo data correlates with the in-vitro release data. The in-vivo evaluation suggest

the in-situ thermorevesible solution to gel formulation of Pluronic, MC and their

combination could increase the diclofenac sodium content absorbed into the eyes and

prolong the elimination time, hence resulting in better bioavailability as compared to the

conventional eye drops.

177
CHAPTER 4 RESULTS AND DISCUSSION

4.2.6.4 In-vivo Evaluation of Timolol Maleate In-situ Gels

The drug release and pharmacokinetics studies were conducted following the instillation

of the formulations TP20, TP18, TPM 15/3, TMPG3/2 and TMPG1.5/10 onto rabbits’

cornea. The pharmacokinetic parameters determined include; Peak aqueous humour

concentration (Cmax),Time (Tmax) to attain Cmax, area under aqueous humour

concentration curve (AUC0-t), area under curve (AUC0-∞), area under the movement

curve (AUMC0-∞), mean residence time (MRT), volume of distribution (Vd), clearance

and Half life (t1/2).

The concentration of TM determined in the samples of rabbit’s aqueous humour was

analyzed by PK-summit® software to calculate the above mentioned pharmacokinetic

parameters.

After the administration of 50 µl (25µg) dose of TM commercial conventional (0.5%) eye

drops, used a s a reference formulation, and the prepared in-situ thermorevesible gel

formulations (TP20, TP18, TPM15/3, TMPG3/2 and TMPG1.5/10). The pharmacokinetic

parameters of TM in aqueous humour of these preparations are depicted formulations in

Table 4.17. Various pharmacokinetics parameters were statistically compared with the

reference formulation using t-test 95% confidence interval.

4.2.6.4.1 TP18 and TP20 TM In-situ Gel

Timolol (TM) aqueous humour concentrations was determined at different time intervals

following the instillation of TP 18 and TP 20 in-situ gel formulations (50 µl ≈25µg) and

178
CHAPTER 4 RESULTS AND DISCUSSION

conventional eye drops, used as reference formulation, onto the rabbits’ cornea. The TM

concentration in the aqueous humour was detected for 6 hours following drug instillation.

Aqueous humour drug concentration-Time profile of the TP 18 and TP 20 following

instillation of eye drops is shown in Figure 4.36. The pharmacokinetic parameters of TM

in aqueous humour of the preparation are given in Table 4.17. The data was statistically

evaluated using Minitab 14 and the difference between in-situ gel preparations and

reference formulation was conducted using student t-test at 95% confidence interval.

Fig 4.35: Representative chromatogram of in-vivo TM release in AH from TP 20 in-situ


gel at different time interval, A=0.5 hr, B=1 hrs, C=2 hrs, D=3 hrs, E=6 hrs.

The Cmax obtained was 3.637 ± 0.56, 6.126 ± 0.45 and 1.601 ± 0.12 for TP18, TP20 and

conventional eye drops respectively. The Cmax values for both in-situ formulations were

higher with significant P-Values compared with the reference conventional eye drops.

The Cmax was 2.7 and 3.83 fold higher than the conventional eye drops for TP18 and

179
CHAPTER 4 RESULTS AND DISCUSSION

TP20 respectively. The Tmax was one hour for both TP18 and TP20 in-situ gels as

compared to conventional eye drop which was half (0.5) hour.

The values of AUC0-t, AUC0-∞, AUMC0-∞, MRT and t1/2 were high for the formulation,

while the clearance rate was low for the in-situ gel formulations. The AUC, CL, MRT

and biological half-life values clearly suggest that the in-situ gels has higher

bioavailability as compared to conventional eye drop.

The AUC0-t values of TP18 and TP20 suggested that the bioavailability of TM was 4.0

and 6.8 fold respectively, higher than the conventional eye drops. As compared to the

reference formulation the relative bioavailability was 44% and 73%higher for DP 18 and

DDP 20 respectively. The MRT were 2.856 ± 0.84 and 2.872 ± 0.33 for DP 18 and DP 20

respectively, while the MRT value of the reference formulation was only 1.305. The

higher value of MRT suggests higher residence time of the in-situ gel formulation in the

ocular tissue in comparison to the conventional TM eye drops. The elimination rate of the

drug from the in-situ gel was significantly low indicating low drainage and high

residence of the gel in the ocular tissue. The elimination half-life was 1.266 ± 0.68, 1.44

± 0.15 and 0.668 ± 0.45 for TP 18, TP 20 and reference formulation respectively. The

half-life of the in-situ gel formulation was significantly high.

The pharmacokinetic parameters revealed that the TM was well absorbed from the TP18

and TP20 formulations, it might be due to the bio-adhesiveness and high consistency of

the gel that increase the residence and contact time [289] of the TM in the eye as

180
CHAPTER 4 RESULTS AND DISCUSSION

compared to the conventional eye drop that may be rapidly drained off and eliminated

from the eye tissue.

Aqueous humour drug concentration-time profile is depicted in Figure 4.36. The rapid

decline of the TM concentration in AH signify rapid erosion of the gel and diffusion of

the water soluble drug in aqueous humour[290].

The TM concentration in aqueous humour for TP18 and TP20 was 0.540 and

0.710µg/ml, respectively, after 6 hours indicating that the gels are able to maintain the

steady state concentration within the aqueous humour for the studied duration.

8.0
TPM 15/3
7.0
TP 20
6.0
Aq.Humour Drug Conc. (µg/ml)

TP 18
5.0 TMPG1.5/10
TMPG3/2
4.0
Reference
3.0

2.0

1.0

0.0

-1.0
0 1 2 3 4 5 6 7

-2.0
Time (h)

Fig. 4.36: Concentration (µg/ml, mean ± SD) of Timolol maleate in rabbits’ aqueous
humour at various times after instillation of a commercial diclofenac sodium eye drops
and TM in-situ gel formulations

The AUC0-t value of the TP20 was higher (p=0.008) than TP18. This significance

indicates that with increase in the pluronic concentration the bioavailability of TM

181
CHAPTER 4 RESULTS AND DISCUSSION

increases. The results obtained in current study are consistent with the earlier studies [91]

and also correlates with the in-vitro release profiles.

4.2.6.4.2 TPM 15/3 TM In-situ Gel

The formulation consists of 15% Pluronic F-127 and 3% methyl cellulose. The in-vitro

release profile indicated that the formulation sustained the drug release for 24 hours that

is higher as compared with TP20 that extended the release of TM for 12 hours. The mean

AH drug concentration–time profiles after administration of the conventional eye drops

and TPM15/3 in-situ gel of TM are illustrated in Figure 4.36. From the profile it is clear

that higher AH drug levels were achieved in case of the TPM15/3 gel compared with the

conventional eye drops. The pharmacokinetic and bioavailability data for the different In-

situ gels and conventional eye drops is shown in Table 4.17.

The Cmax values were 3.10 8 ± 0.70 µg/ml for the TPM15/3 in-situ gel while that of

conventional reference formulation was 1.601 ± 0.12 µg/ml. Statistical analysis revealed

that the Cmax was significantly (p<0.02) higher in case of the in-situ gel.

Moreover, the values for AUC0-t, AUC 0-∞, AUMC0-∞ and MRT were significantly high

for the in-situ gel compared with the conventional eye drops (Table 4.17); suggesting

high bioavailability of the drug from the thermorevesible gel. This might be due to the

bio-adhesive character of the gel imparted by MC[291]. The relative bioavailability of the

formulation compared with reference was 39% greater. The increase in bioavailability

might be due to highly viscose gel formed by the addition of the thickening agent MC to

182
CHAPTER 4 RESULTS AND DISCUSSION

the poloxamer solution. The results obtained in current study is consistent to the in the

early studies [146].

The MRT obtained for the formulation (2.988 ± 0.40) was higher than the reference

formulation (1.395 ± 0.25) the higher MRT value is due to the long residence and contact

of the formed gel as compared to the reference conventional eye drop that drains off

rapidly. The elimination rate of the drug from the in-situ gel was significantly low

indicating low drainage and high residence of the gel in the ocular tissue. The increased

residence and contact time in the eye is due to the adhesiveness of gel augmented by the

addition of MC.

4.2.6.4.3 TMPG 3/2 and TMPG 1.5/10 TM In-situ Gel

Aqueous humour drug concentration-Time profile of the TMPG 3/2 and TMPG 1.5/10

following instillation of these formulations is shown in Figure 4.36.The pharmacokinetic

parameters of TM in aqueous humour of the preparations are given in Table 4.17. The

data was statistically evaluated using Minitab 14 and the difference between in-situ gel

preparations and reference formulation was conducted using student t-test at 95%

confidence interval.

The pharmacokinetic data obtained for the in-situ formulations is shown in Table

4.17.The peak aqueous humour drug concentration for both the formulation was obtained

at one hour.

183
CHAPTER 4 RESULTS AND DISCUSSION

The Cmax value was in order of 3.145 ± 0.45µg/ml and 2.879 ± 0.41µg/ml and P-Values

were 0.018 and 0.035 for TMPG3/2 and TMPG 1.5/10, respectively. The Cmax obtained

for the in-situ gel formulations were significantly high compared with the commercial

ophthalmic solution.

The values of AUC0-t, AUC0-∞, AUMC0-∞, MRT and t1/2 were higher and statistically

significant. The higher values of these Parameters indicate that the bioavailability of the

drug from the in-situ gels was higher in comparison to the commercial eye drops. The

relative bioavailability of the formulations TMPG 3/2 and TMPG 1.5/10 was 33% and

26% more than that of the reference conventional eye drops.

The MRT values obtained using PK-Summit software was higher for the formulation

TMPG 3/2 and TMPG 1.5/10, suggesting high residence and slow drainage from the eye,

which is the prime objective of enhancing the ocular drug availability. The elimination

rates for TMPG3/2, TMPG 1.5/10 and conventional eye drops was 25.995 ml/hr/kg,

33.053 ml/hr/kg and 86.508 ml/hr/kg, respectively. The low elimination rate suggests that

the clearance of the TM was low after the instillation of the in-situ thermoreversible gel

compared with the conventional eye drops. The high Cmax, bioavailability and lower

elimination rate is due high residence and contact of the in-situ gels because of the muco-

adhesive character of the MC.

184
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.17: Pharmacokinetic parameters of Timolol maleate commercial formulation and in-situ gel formulation after ocular
administration

P P P P P Conv. Eye
Pk Parameter Unit TP20 TP18 TPM15/3 TMPG 1.5/10 TMPG3/2
value value value value value Drops

Cmax µg/ml 6.126±0.45 0.009 3.637±0.56 0.019 3.108±0.70 0.028 2.879±0.41 0.035 3.145±0.45 0.018 1.601±0.12

Tmax Hr 1.0±0.0 1.0±0.0 1.0±0.0 1.0±0.0 1.0±0.0 0.50±0.0

AUC(0-t) µg-hr/ml 14.984±1.29 0.004 8.870±0.50 0.002 8.049±0.21 0.001 5.327±0.28 0.004 6.352±0.28 0.001 2.202±0.18

AUC (0-∞) µg-hr/ml 16.458±1.86 0.007 9.856±3.50 0.005 8.815±0.28 0.001 5.900±0.33 0.001 7.502±0.47 0.006 2.254±0.22

AUMC (0-∞) µg-hr*hr/ml 47.263±11.01 0.023 28.143±10.50 0.06 26.339±3.88 0.009 14.991±1.51 0.014 24.010±1.21 0.001 2.943±0.83

MRT Hr 2.872±0.33 0.003 2.856±0.84 0.175 2.988±0.40 0.017 2.541±0.27 0.089 3.201±0.42 0.011 1.305±0.25

Vd ml/kg 24.617±5.37 0.063 36.135±16.00 0.111 35.439±2.29 0.062 101.810±13.53 0.288 93.108±7.46 0.188 83.429±39.86

CL kg ml/hr/kg 11.848±1.26 0.003 19.786±3.02 0.007 22.121±0.66 0.006 33.053±2.80 0.02 25.995±2.81 0.01 86.508±6.90

Half-life from
Hr 1.440±0.15 0.623 1.266±0.68 0.754 1.110±0.13 0.729 2.135±0.35 0.033 2.482±0.20 0.033 0.668±0.45
Vd and CL

(n= 3, mean ± SD)

185
CHAPTER 4 RESULTS AND DISCUSSION

It can be concluded from the data that higher the concentration of MC higher the

bioavailability, also it can be inferred from the data that gels (TMPG1.5/10) with low

concentration of MC and high concentration of PEG can reasonably increase the

bioavailability and also extend the drug release for the studied period of time.

4.2.6.4.4 Conclusion

The bioavailability of all the in-situ gels was significantly high as compared to the

reference conventional ophthalmic solution. The elimination rate of the drug was

significantly low than that of conventional eye drops.

The TM content after the instillation of conventional commercial eye drop could not be

detected in the aqueous humour after four hours indicating that it was below the limit of

detection or cleared from the aqueous humour. While the TM content for the gels was

detected after six hours after the instillation. The concentration of the TM determined

with HPLC method for TP20, TP18, TPM15/3, TMPG3/2 and TMPG1.5/10 after 6 hours

was 0.710, 0.540, 0.478, 0.186 and 0.321 (µg/ml) respectively.

These values suggested that the in-situ thermorevesible gels prepared with Pluronic, MC

and their combination could increase the TM content absorbed into the eyes and prolong

the elimination time, hence resulting in better bioavailability as compared to the

conventional eye drops.

186
CHAPTER 4 RESULTS AND DISCUSSION

4.2.6.5 In-vivo Evaluation of Insulin In-situ Gels

The drug release and pharmacokinetics studies were conducted following the

subcutaneous administration of the formulation IPM 15/3 and commercial insulin

preparation to rabbits. The pharmacokinetic parameters determined include; peak plasma

concentration (Cmax), Time (Tmax) to attain Cmax, area under plasma concentration curve

(AUC0-t), area under curve (AUC0-∞), area under the movement curve (AUMC0-∞), mean

residence time (MRT), volume of distribution (Vd) and clearance and Half-life (t1/2).

The concentration of insulin determined in the rabbits’ plasma samples (Figure 4.37 and

4.38) after the administration of 2ml (2U) dose of Insulin commercial conventional

preparation, used as a reference formulation, and the IPM15/3 in-situ thermorevesible gel

formulations was analyzed by PK-summit® software.

The pharmacokinetic parameters of Insulin in plasma of these preparations are depicted

in Table 4.18. Various pharmacokinetics parameters were statistically compared with the

reference formulation using student’s t-test 95% confidence interval.

4.2.6.5.1 IPM 15/3 Insulin In-situ Gel

Insulin plasma concentration was determined at different time intervals following the

subcutaneous administration of IPM15/3 and conventional insulin preparation, used as

reference formulation. The plasma Insulin concentration (Figure 4.37) was detected for

240 hours (10 days) following administration of in-situ gel formulation in comparison

with the conventional insulin preparation for which the insulin content in plasma was

detected for only 12 hours (Figure 4.38).

187
CHAPTER 4 RESULTS AND DISCUSSION

45.0

Plasma Insulin Level (µU/ml)


40.0
35.0
30.0
25.0
20.0
15.0
10.0
5.0
0.0
0 24 48 72 96 120 144 168 192 216 240 264
Time (hrs)

Fig 4.37: Plasma concentration (µU/ml) of Insulin in rabbits at various times intervals
after subcutaneous administration of Insulin in-situ gel formulations

30.0
Plasma Insulin level (µU/ml)

25.0

20.0

15.0

10.0

5.0

0.0
0 2 4 6 8 10 12 14
Time (hrs)

Fig 4.38: Plasma concentration (µU/ml) of Insulin in rabbits at various times intervals
after subcutaneous administration of a commercial insulin solution

The Cmax values attained after administration of the In-situ gel formulation and reference

formulation were 37.7 ± 5.1 and 26.0 ± 0.8, respectively. The Cmax of IPM15/3 was 1.45

times higher than the reference formulation. The Cmax for the in-situ gel was achieved

188
CHAPTER 4 RESULTS AND DISCUSSION

after 144 hours in comparison to one hour of the reference insulin formulation. A burst

release at 144 hours from the IPM15/3 was observed (Figure 4.37) indicating the

degradation of the polymer followed by decline plasma insulin level.

Higher AUC0-t, AUC0-∞, AUMC0-∞ and MRT values were obtained for the in-situ gel

compared to reference parenteral solution (Table 4.18). The P-Values for all the

pharmacokinetic parameters were significant at 95% confidence interval.

189
CHAPTER 4 RESULTS AND DISCUSSION

Table 4.18: Pharmacokinetic parameters of Human Insulin commercial formulation and


in-situ gel formulation after subcutaneous administration

IPM 15/3 Reference


PK Parameter Unit P Value
Mean ± SD Mean ± SD

E Half-life Hr 28.6±5.9 7.2±0.3 0.001

Cmax µg/ml 37.7±5.1 26.0±0.8 0.003

Tmax Hr 144.0±0.00 1.0±0.0 ----

AUC(0-t) µg-hr/ml 5794.0±940 123.8±4.3 0.001

AUC (0-∞) µg-hr/ml 6228.6±954 181.6±9.8 0.001

AUMC (0-∞) µg-hr*hr/ml 805888±117910 1881.8±178 0.001

MRT Hr 129.5±2.1 10.3±0.4 0.001

Vd ml/kg 1.0±0.4 8.6±0.3 0.001

CL ml/hr/kg 0.025±0.004 0.83±0.04 0.001

Half-life from
Hr 28.6±5.9 7.2±0.3 0.001
Vd and CL

(n=3, Mean ± SD)

190
CHAPTER 4 RESULTS AND DISCUSSION

The higher values of these parameters suggest higher bioavailability of the drug from the

thermorevesible gels. The relative bioavailability compared with reference formulation

was 360% higher for the in-situ gel formulation.

The volume of distribution (Vd) of the in-situ gel formulation was significantly low

compared with the reference formulation. The Vd was small in case of IPM 15/3 in-situ

gel formulations as the drug concentration was maintained at steady state this may be due

to the equilibrium between the drug absorption and elimination. Insulin was slowly

released from the gel formulation compared with the conventional reference formulation

that provides 100% of the drug immediately at the site of absorption.

The elimination rate of the drug from the in-situ gel was significantly low compared to

the reference conventional solution. The MRT was statistically significant; the MRT

value was 129±2.1 that was 12.5 times greater than the conventional reference

preparation.

The formulation IPM 15/3 maintains the steady state plasma insulin level between 10-40

µU/ml (Figure 4.37) throughout the study period which is the normal required basal

insulin level [292].

191
CHAPTER 5 CONCLUSION

CHAPTER 5
CONCLUSION

192
CHAPTER 5 CONCLUSION

5. Conclusion
The objective of the current study was to develop sustained release in-situ solution to gel

polymeric delivery system for the drugs to be administered through subcutaneous or

ocular route. Four polymers (Pluronic F-127, Methyl cellulose, hydroxyl

propylmethylcellulose and polyethylene glycol 6000) were used in different

concentrations alone and in combination with each other in different ratios. Total of thirty

four formulations were prepared and evaluated for Tsol-gel.

Depending upon the gelling temperature and rheological behavior only five formulations

(P20, P18, PM15/3, MPG 3/2 and MPG 1.5/10) among the prepared thirty four

formulations were selected as drug delivery system. The selected formulations were clear

free flowing solution at room temperature (25oC) and converts to viscous gel below the

body temperature (37oC). All of the formulations showed perfect thermorevesible

behavior i.e. the solutions gels with increase in temperature and reverts to solution with

decrease in the temperature. The formed gels of the solutions (P18 and P20) containing

the pluronic as thermorevesible polymer were transparent, while the solutions prepared

by combination of pluronic and MC (PM15/3) form semitransparent gel. The gels of the

formulation (MPG 3/2 and MPG 1.5/10) containing MC and PG were turbid.

The studied preparations were Newtonian at room temperature and the formed gels were

thixotropic as indicated by the flow index and consistency index. It was also concluded in

the current study that sterilization by autoclaving have no significant effect on the Tsol-

gel and rheological properties.

193
CHAPTER 5 CONCLUSION

Addition of DS and insulin to the hydrogel solutions showed a positive effect on Tsol-gel

i.e. these drugs increase the transition temperature while TM decreases the Tsol-gel. The

polymer solutions of the drug were clear indicating complete solubility of the drugs.

For the determination of the drug content in the in-vitro and in-vivo samples of the DS

and TM HPLC-UV method was developed and validated as per ICH guidelines. The

analytes were separated within 7 minutes with total run time of 10 minutes. The LOD

determined for DS and timolol were 2.0 ng/ml and 1.0 ng/ml respectively, while the LOQ

for the drugs were 0.80 ng/ml and 0.25 ng/ml respectively.

Autoclaving have no significant effect on the Tsol-gel, viscosity, and clarity of

thermoreversible formulation of DS and TM, though it decreases the drug content of DS

formulations significantly. The filtration process carried out for sterilization of insulin

formulations showed no significant effect on the physical and chemical properties of

these formulations.

The in-vitro drug release data of the DS preparation reveals that the formulation DP 20

extended the drug release for 144 hours that was fully supported by the in-vivo data. The

drug release from the DP18 and DP 20 followed Higuchi model while DMPG 3/2,

DMPG 1.5/10 and DPM 15/3 followed Hixson-Crowell Model.

TPM 15/3 extended the drug release for 24 hours, while TP18 and TMPG 1.5/10 could

only retard the drug release for 8 hours. All formulations followed first order release

194
CHAPTER 5 CONCLUSION

kinetics. The linearity for Hixson –Crowell suggests the drug release through diffusion of

drug and erosion of polymer.

Among the formulations of insulin the formulation IPM 15/3 showed promising in-vitro

dissolution profile by extended the drug release for 264 hours (11 days). The drug release

form the formulation was governed by diffusion. The in-vivo studies indicate that the

formulation IPM15/3 is capable of maintaining the plasma insulin level for 10 days.

The study reveals that HPMC polymer didn’t gel below the body temperature (37oC) at

the studied concentration range (1-10%w/v).

The in-vivo data obtained correlates to the in-vitro dissolution profile. The

thermoreversible in-situ gel formulations of all three drugs showed greater and prolong

bioavailability compared with reference conventional formulations of these drugs. The

formulations of all the drugs studied were able to maintain the plasma and AH steady

state concentration over a longer period of time.

195
REFERENCES

REFERENCES

196
REFERENCES

6. References

1. Linda. F., E.L.W.W.P., ed. In Extended release and targeted drug delivery
systems. Adam, W.G. Remington The Science and Practice of Pharmacy. 2006; .
p 946.

2. Nicholas G. Lordi, L.L., ed. The Theory and Practice of Industrial Pharmacy. In
Sustained release dosage form. 2005, Ed.; Lea & Febiger: Philadelphia. pp
440,442.

3. Arnott, S., et al., The agarose double helix and its function in agarose gel
structure. Journal of molecular biology, 1974. 90(2): p. 269-272.

4. Rees, D.A. and E.J. Welsh, Secondary and tertiary structure of polysaccharides
in solutions and gels. Angewandte Chemie International Edition in English, 1977.
16(4): p. 214-224.

5. Rozier, A., et al., Gelrite®: A novel, ion-activated, in-situ gelling polymer for
ophthalmic vehicles. Effect on bioavailability of timolol. International Journal of
Pharmaceutics, 1989. 57(2): p. 163-168.

6. Franz, G., Polysaccharides in pharmacy. Pharmacy/ thermodynamics/


Elastomers/Telechelics, 1986: p. 1-30.

7. Peppas, N.A., Hydrogels in medicine and pharmacy. 1986: CRC Pr.

8. Bekturov, E. and L. Bimendina, Interpolymer complexes Speciality Polymers.


1981, Springer Berlin / Heidelberg. p. 99-147.

9. Jeong, B., Y.H. Bae, and S.W. Kim, Drug release from biodegradable injectable
thermosensitive hydrogel of PEG-PLGA-PEG triblock copolymers. Journal of
Controlled Release, 2000. 63(1-2): p. 155-163.

10. Gong, C., et al., In vitro drug release behavior from a novel thermosensitive
composite hydrogel based on Pluronic f127 and poly (ethylene glycol)-poly (ε-
caprolactone)-poly (ethylene glycol) copolymer. BMC biotechnology, 2009. 9(1):
p. 8.

11. Tyagi, P., et al., Sustained Intravesical Drug Delivery Using Thermosensitive
Hydrogel. Pharmaceutical Research, 2004. 21(5): p. 832-837.

197
REFERENCES

12. Mulik, R., V. Kulkarni, and R.S.R. Murthy, Chitosan-based thermosensitive


hydrogel containing liposomes for sustained delivery of cytarabine. Drug
Development and Industrial Pharmacy, 2009. 35(1): p. 49-56.

13. Kang, G.D., et al., Thermosensitive poly (organophosphazene) hydrogels for a


controlled drug delivery. European Journal of Pharmaceutics and
Biopharmaceutics, 2006. 63(3): p. 340-346.

14. Jeong, B., et al., Biodegradable block copolymers as injectable drug-delivery


systems. Nature, 1997. 388(6645): p. 860-862.

15. Suzuki, A. and T. Tanaka, Phase transition in polymer gels induced by visible
light. Nature, 1990. 346(6282): p. 345-347.

16. Miyata, T., N. Asami, and T. Uragami, A reversibly antigen-responsive hydrogel.


Nature, 1999. 399(6738): p. 766-769.

17. Nanjawade, B.K., F.V. Manvi, and A.S. Manjappa, In situ-forming hydrogels for
sustained ophthalmic drug delivery. Journal of Controlled Release, 2007. 122(2):
p. 119-134.

18. Zentner, G.M., et al., Biodegradable block copolymers for delivery of proteins
and water-insoluble drugs. Journal of Controlled Release, 2001. 72(1-3): p. 203-
215.

19. Jeong, B., S.W. Kim, and Y.H. Bae, Thermosensitive sol-gel reversible hydrogels.
Advanced Drug Delivery Reviews, 2002. 54(1): p. 37-51.

20. Ruel-Gariepy, E. and J.C. Leroux, In situ-forming hydrogels--review of


temperature-sensitive systems. European Journal of Pharmaceutics and
Biopharmaceutics, 2004. 58(2): p. 409-426.

21. Kissel, T., Y. Li, and F. Unger, ABA-triblock copolymers from biodegradable
polyester A-blocks and hydrophilic poly (ethylene oxide) B-blocks as a candidate
for in situ forming hydrogel delivery systems for proteins. Advanced Drug
Delivery Reviews, 2002. 54(1): p. 99-134.

22. Choi, S.W., et al., Thermoreversible gelation of poly (ethylene oxide)


biodegradable polyester block copolymers. II. Journal of Polymer Science Part A:
Polymer Chemistry, 1999. 37(13): p. 2207-2218.

198
REFERENCES

23. Cai Bing, L., et al., Thermoreversible gel-sol behavior of biodegradable PCL-
PEG-PCL triblock copolymer in aqueous solutions. Journal of Biomedical
Materials Research Part B: Applied Biomaterials, 2008. 84B(1): p. 165-175.

24. Li, J., et al., Synthesis and characterization of new biodegradable amphiphilic
poly (ethylene oxide)-b-poly [(R)-3-hydroxy butyrate]-b-poly (ethylene oxide)
triblock copolymers. Macromolecules, 2003. 36(8): p. 2661-2667.

25. Gong, C.Y., et al., A thermosensitive hydrogel based on biodegradable


amphiphilic poly (ethylene glycol)–polycaprolactone–poly (ethylene glycol)
block copolymers. Smart materials and structures, 2007. 16: p. 927.

26. Bromberg, L.E. and E.S. Ron, Temperature-responsive gels and thermogelling
polymer matrices for protein and peptide delivery. Advanced Drug Delivery
Reviews, 1998. 31(3): p. 197-221.

27. Gilbert, J.C., et al., The effect of solutes and polymers on the gelation properties
of pluronic F-127 solutions for controlled drug delivery. Journal of Controlled
Release, 1987. 5(2): p. 113-118.

28. Kumar, M.N.V.R., ed. Handbook of Particulate Drug Delivery. 2008, American
Scientific Publishers. .

29. Leong, K.W. and R. Langer, Polymeric controlled drug delivery. Advanced Drug
Delivery Reviews, 1988. 1(3): p. 199-233.

30. Remington, L.A., Clinical Anatomy and Physiology of the Visual System. 2012:
Elsevier/Butterworth-Heinemann.

31. Urtti, A., Challenges and obstacles of ocular pharmacokinetics and drug delivery.
Advanced Drug Delivery Reviews, 2006. 58(11): p. 1131-1135.

32. Hornof, M., E. Toropainen, and A. Urtti, Cell culture models of the ocular
barriers. European Journal of Pharmaceutics and Biopharmaceutics, 2005. 60(2):
p. 207-225.

33. Urtti, A., et al., Controlled drug delivery devices for experimental ocular studies
with timolol 2. Ocular and systemic absorption in rabbits. International Journal of
Pharmaceutics, 1990. 61(3): p. 241-249.

34. M.L.Sears, ed. Handbook of experimental pharmacology, . Ocular


pharmacokinetics,, ed. S.M. D.M. Maurice. Vol. 69. 1984, Springer: Verlag,
Berlin-Heidelberg. 16–119.

199
REFERENCES

35. A.L. Gomes dos Santos, A.B., A. Doyle, N. Tsapis, J.Siepmann, F. Siepmann, J.
Schmaler, M. Besnard, F. Behar-Cohen, E. Fattal, , Sustained release of
nanosized complexes of polyethylenimine and anti-TGF-beta 2 oligonucleotide
improves the outcome of glaucoma surgery, . J. Control. Release, 2006. 112: p.
369–381.

36. L. Pitkänen, V.P.R., H. Moilanen, A. Urtti, Permeability of retinal pigment


epithelium: effect of permeant molecular weight and lipophilicity, Investig.
Ophthalmol. Vis. Sci. , 2005. 46: p. 641–646.

37. M.R. Prausnitz, J.S.N., Permeability of cornea, sclera, and conjunctiva: a


literature analysis for drug delivery to the eye. J. Pharm. Sci., 1998. 87: p. 1479–
1488.

38. Divyesh H. Shastri*1, L.D.P., A Novel Alternative to Ocular Drug Delivery


System: Hydrogel. International Journal of Pharmaceutical Research, 2010.
Volume 2, Issue( 1): p. 1-13.

39. Arto, U. and S. Lotta, Minimizing systemic absorption of topically administered


ophthalmic drugs. Survey of Ophthalmology, 1993. 37(6): p. 435-456.

40. Urtti, A., L. Salminen, and O. Miinalainen, Systemic absorption of ocular


pilocarpine is modified by polymer matrices. International Journal of
Pharmaceutics, 1985. 23(2): p. 147-161.

41. Urtti, A., et al., Controlled Ocular Timolol Delivery: Systemic Absorption and
Intraocular Pressure Effects in Humans. Pharmaceutical Research, 1994. 11(9):
p. 1278-1282.

42. H.N. Bhargava, D.W.N., B.J. Oza, ed. Pharmaceutical dosage forms: Disperse
systems. ed. i.H.A.L. , R.M. Rieger, G.S. Banker (Eds.). Vol. 2. 1996, Marcel
Dekker, Inc. 183–241.

43. H.A. Lieberman, R.M.R., G.S. Banker (Eds.),, ed. Pharmaceutical dosage forms:
Disperse systems. Ophthalmic ointments and suspensions, ed. G.H. K.M. Bapatla.
Vol. 2. 1996, Marcel Dekker, Inc. 357-397.

44. Mitra, A.S., ed. Ophthalmic drug delivery systems. Mucoadhesive polymers in
ocular drug delivery,, ed. A.K.M. R. Krishnamoorthy. 1993, Marcel Dekker, Inc.
199–221.

45. Miller S, D.M., Effect of poloxamer 407 gel on the miotic activity of pilocarpine
nitrate in rabbits. Int. J. Pharm., 1982(12): p. 147-152.

200
REFERENCES

46. J.G. Hardman, L.E.L., & A.G. Gilman (Eds.), ed. Goodman & Gilman's the
pharmacological basis of therapeutics. Pharmacokinetics: The dynamics of drug
absorption, distribution, and elimination, ed. G.R. Wilkinson. 2001, : McGraw-
Hill: New York. 3-29.

47. Kretlow, J.D., L. Klouda, and A.G. Mikos, Injectable matrices and scaffolds for
drug delivery in tissue engineering. Advanced Drug Delivery Reviews, 2007.
59(4-5): p. 263-273.

48. Matschke, C., et al., Sustained-release injectables formed in situ and their
potential use for veterinary products. Journal of Controlled Release, 2002. 85(1-
3): p. 1-15.

49. Lachman L, L.H., Kanig J, , ed. In: The theory and practice of industrial
pharmacy,. ed. S.p. Avis Kenneth, Varghese publishing house,: Bombay, . 653-
654.

50. Marcel Dekker, ed. In: Novel drug delivery system, . second Ed, ed. Parenteral
drug delivery and delivery systems, , ed. Chien YW: New York,. 382-385.

51. Gabizon, A., et al., Development of liposomal anthracyclines: from basics to


clinical applications. Journal of Controlled Release, 1998. 53(1-3): p. 275-279.

52. Mantripragada, S., A lipid based depot (DepoFoam® technology) for sustained
release drug delivery. Progress in Lipid Research, 2002. 41(5): p. 392-406.

53. RC., W., Method for utilizing neutral lipids to modify in vivo release from
multivesicular liposomes. , U.S. Patent, Editor. 1999.: USA.

54. Okada, H., et al., Preparation of Three-Month Depot Injectable Microspheres of


Leuprorelin Acetate Using Biodegradable Polymers. Pharmaceutical Research,
1994. 11(8): p. 1143-1147.

55. R.S. Bezwada, S.C.A., Liquid absorbable copolymers for parenteral applications.
5 August 1997: USA.

56. P.C. Painter, M.M.C.E., Fundamentals of Polymer science, . 1994, , Lancaster,


PA, : Technomic. pp. 4–5.

57. Einmahl, S., et al., A viscous bioerodible poly(ortho ester) as a new biomaterial
for intraocular application. Journal of Biomedical Materials Research, 2000.
50(4): p. 566-573.

201
REFERENCES

58. In: Tannock I, H.R., editors. , ed. Basic science of oncology. . Hyperthermia and
photodynamic therapy., ed. W.B. Liu F. 1998. , McGraw-Hill; : New York: . p.
443-53.

59. Zhang, X., et al., Development of biodegradable polymeric paste formulations for
taxol: An in vitro and in vivo study. International Journal of Pharmaceutics, 1996.
137(2): p. 199-208.

60. Hatefi, A. and B. Amsden, Biodegradable injectable in situ forming drug delivery
systems. Journal of Controlled Release, 2002. 80(1-3): p. 9-28.

61. Soppimath, K.S., et al., Stimulus-Responsive "Smart" Hydrogels as Novel Drug


Delivery Systems*. Drug Development and Industrial Pharmacy, 2002. 28(8): p.
957-974.

62. Kim, S.J., S.J. Park, and S.I. Kim, Properties of smart hydrogels composed of
polyacrylic acid/poly (vinyl sulfonic acid) responsive to external stimuli. Smart
materials and structures, 2004. 13: p. 317.

63. Qiu, Y. and K. Park, Environment-sensitive hydrogels for drug delivery.


Advanced Drug Delivery Reviews, 2001. 53(3): p. 321-339.

64. Aikawa, K., et al., Drug release from pH-response polyvinylacetal


diethylaminoacetate hydrogel, and application to nasal delivery. International
Journal of Pharmaceutics, 1998. 168(2): p. 181-188.

65. Engström, S. and L. Engström, Phase behaviour of the lidocaine-monoolein-


water system. International Journal of Pharmaceutics, 1992. 79(1-3): p. 113-122.

66. mattiowitz, E., ed. Encyclopedia of Controlled Drug Delivery, . 1 ed. Fabrication
of controlled-delivery, ed. Z.Z. James P. English. Vol. 2. 1999, Wiley-
Interscience; .

67. Lin, C.-C. and A.T. Metters, Hydrogels in controlled release formulations:
Network design and mathematical modeling. Advanced Drug Delivery Reviews,
2006. 58(12-13): p. 1379-1408.

68. Chen, X., et al., Enzymatic and chemoenzymatic approaches to synthesis of sugar-
based polymer and hydrogels. Carbohydrate Polymers, 1995. 28(1): p. 15-21.

69. Kashyap, N., N. Kumar, and M.N.V.R. Kumar, Hydrogels for Pharmaceutical
and Biomedical Applications. 2005. 22(2): p. 107-150.

202
REFERENCES

70. Corkhill, P.H., C.J. Hamilton, and B.J. Tighe, Synthetic hydrogels VI. Hydrogel
composites as wound dressings and implant materials. Biomaterials, 1989. 10(1):
p. 3-10.

71. Lee, K.Y. and D.J. Mooney, Hydrogels for Tissue Engineering. Chemical
Reviews, 2001. 101(7): p. 1869-1880.

72. Plunkett, K.N. and J.S. Moore, Patterned Dual pH-Responsive Core−Shell
Hydrogels with Controllable Swelling Kinetics and Volumes. Langmuir, 2004.
20(16): p. 6535-6537.

73. Lutolf, M.P., et al., Cell-Responsive Synthetic Hydrogels. Advanced Materials,


2003. 15(11): p. 888-892.

74. Dušek, K., et al., Hydrogels as separation agents, in Responsive Gels: Volume
Transitions II. 1993, Springer Berlin / Heidelberg. p. 67-79.

75. Bennett, S.L., et al., Next-Generation HydroGel Films as Tissue Sealants and
Adhesion Barriers. Journal of Cardiac Surgery, 2003. 18(6): p. 494-499.

76. Colombo, P., Swelling-controlled release in hydrogel matrices for oral route.
Advanced Drug Delivery Reviews. 11(1-2): p. 37-57.

77. K.A. Davis, K.S.A., Controlled release from crosslinked degradable networks,
Crit. Rev. . Ther. Drug Carr. Syst. , 2002. 19: p. 385-423.

78. Van Tomme, S.R., G. Storm, and W.E. Hennink, In situ gelling hydrogels for
pharmaceutical and biomedical applications. International Journal of
Pharmaceutics, 2008. 355(1-2): p. 1-18.

79. Fogueri, L.R. and S. Singh, Smart polymers for controlled delivery of proteins
and peptides: a review of patents. Recent Patents on Drug Delivery &# 38;
Formulation, 2009. 3(1): p. 40-48.

80. Allen, L.V. and J.P.D.R. Ph, Secundum Artem. Current and Practical
Compounding Information for the Pharmacist. Pharmaceutical Compounding
Resources on the Internet, 2009. 10(1).

81. Morikawa, K., et al., Enhancement of therapeutic effects of recombinant


interleukin 2 on a transplantable rat fibrosarcoma by the use of a sustained
release vehicle, pluronic gel. Cancer research, 1987. 47(1): p. 37.

203
REFERENCES

82. Padilla, M., G.T. Clark, and R.L. Merrill, Topical medications for orofacial
neuropathic pain: a review. The Journal of the American Dental Association,
2000. 131(2): p. 184-195.

83. Jain, N.K., B.K. Shah, and L.N. Taneja, Nasal absorption of metoprolol tartrate.
Indian J. Pharm. Sci, 1991. 53: p. 16-19.

84. Barichello, J.M., et al., Enhanced rectal absorption of insulin-loaded Pluronic®


F-127 gels containing unsaturated fatty acids. International Journal of
Pharmaceutics, 1999. 183(2): p. 125-132.

85. Ryu, J.M., et al., Increased bioavailability of propranolol in rats by retaining


thermally gelling liquid suppositories in the rectum. Journal of Controlled
Release, 1999. 59(2): p. 163-172.

86. Miller, S.C. and M.D. Donovan, Effect of poloxamer 407 gel on the miotic activity
of pilocarpine nitrate in rabbits. International Journal of Pharmaceutics, 1982.
12(2-3): p. 147-152.

87. Saettone, M.F., et al., Solubilization of tropicamide by poloxamers:


physicochemical data and activity data in rabbits and humans. International
Journal of Pharmaceutics, 1988. 43(1-2): p. 67-76.

88. Desai, S.D. and J. Blanchard, Evaluation of Pluronic F127†based


sustained†release ocular delivery systems for pilocarpine using the albino
rabbit eye model. Journal of Pharmaceutical Sciences, 1998. 87(10): p. 1190-
1195.

89. Habib, F.S. and M.A. Attia, Comparative study of the ocular activity in rabbit
eyes of adrenaline bitartrate formulated carbopol amd poloxamers gels. Arch
Pharm. Chem. Sci, 1984. 12: p. 91-96.

90. Lin, H.R. and K.C. Sung, Carbopol/pluronic phase change solutions for
ophthalmic drug delivery. Journal of Controlled Release, 2000. 69(3): p. 379-388.

91. Edsman, K., J. Carlfors, and R. Petersson, Rheological evaluation of poloxamer


as an in situ gel for ophthalmic use. European journal of pharmaceutical sciences,
1998. 6(2): p. 105-112.

92. Koller, C. and P. Buri, Propriétés et intérêt pharmaceutique des gels


thermoréversibles à base de poloxamers et poloxamines. STP pharma, 1987.
3(2): p. 115-124.

204
REFERENCES

93. DiBiase, M., and Rhodes, C., Formulation and evaluation of Epidermal Growth
Factor in Pluronic-127 Gel, Drug Develop. Ind. Pharm., 1996. 22(8): p. 823-831.

94. Takats, Z., K. Vekey, and L. Hegedüs, Qualitative and quantitative


determination of poloxamer surfactants by mass spectrometry. Rapid
Communications in Mass Spectrometry, 2001. 15(10): p. 805-810.

95. Kabanov, A.V., E.V. Batrakova, and V.Y. Alakhov, Pluronic® block
copolymers as novel polymer therapeutics for drug and gene delivery. Journal of
Controlled Release, 2002. 82(2-3): p. 189-212.

96. Moghimi, S.M. and A.C. Hunter, Poloxamers and poloxamines in nanoparticle
engineering and experimental medicine. Trends in biotechnology, 2000. 18(10):
p. 412-420.

97. Dumortier, G., et al., Rheological study of a thermoreversible morphine gel. Drug
Development and Industrial Pharmacy, 1991. 17(9): p. 1255-1265.

98. Juhasz, J., et al., Diffusion of rat atrial natriuretic factor in thermoreversible
poloxamer gels. Biomaterials, 1989. 10(4): p. 265-268.

99. Dimitrova, E., et al., Development of model aqueous ophthalmic solution of


indomethacin. Drug Development and Industrial Pharmacy, 2000. 26(12): p.
1297-1301.

100. Barichello, J.M., et al., Absorption of insulin from Pluronic F-127 gels following
subcutaneous administration in rats. International Journal of Pharmaceutics,
1999. 184(2): p. 189-198.

101. Strickley, R.G., Solubilizing excipients in oral and injectable formulations.


Pharmaceutical Research, 2004. 21(2): p. 201-230.

102. Shin, S.C. and C.W. Cho, Physicochemical characterizations of piroxicam-


poloxamer solid dispersion. Pharmaceutical development and Technology, 1997.
2(4): p. 403-407.

103. Juhasz, J., et al., Diffusion of natirel factor in thermoreversible poloxamer gels.
Biomaterials, 1986. 2: p. 365-369.

104. Pec, E.A., Z.G. Wout, and T.P. Johnston, Biological activity of urease formulated
in poloxamer 407 after intraperitoneal injection in the rat. Journal of
Pharmaceutical Sciences, 1992. 81(7): p. 626-630.

205
REFERENCES

105. Wang, P. and T.P. Johnston, Enhanced stability of two model proteins in an
agitated solution environment using poloxamer 407. PDA Journal of
Pharmaceutical Science and Technology, 1993. 47(4): p. 183-189.

106. Bromberg, L.E., Interactions among proteins and hydrophobically modified


polyelectrolytes. Journal of pharmacy and pharmacology, 2001. 53(4): p. 541-547.

107. Katakam, M. and A.K. Banga, Use of poloxamer polymers to stabilize


recombinant human growth hormone against various processing stresses.
Pharmaceutical development and Technology, 1997. 2(2): p. 143-149.

108. Stratton, L.P., et al., Drug delivery matrix containing native protein precipitates
suspended in a poloxamer gel. Journal of Pharmaceutical Sciences, 1997. 86(9):
p. 1006-1010.

109. Olivieri, L., et al., Optimization of a thermally reversible W/O/W multiple


emulsion for shear-induced drug release. Journal of Controlled Release, 2003.
88(3): p. 401-412.

110. Yong, C.S., et al., Effect of sodium chloride on the gelation temperature, gel
strength and bioadhesive force of poloxamer gels containing diclofenac sodium.
International Journal of Pharmaceutics, 2001. 226(1-2): p. 195-205.

111. Choi, H.G., et al., Effect of additives on the physicochemical properties of liquid
suppository bases. International Journal of Pharmaceutics, 1999. 190(1): p. 13-19.

112. Dumortier, G., et al., Development of a thermogelling ophthalmic formulation of


cysteine. Drug Development and Industrial Pharmacy, 2006. 32(1): p. 63-72.

113. Chang, J.Y., et al., Rheological evaluation of thermosensitive and mucoadhesive


vaginal gels in physiological conditions. International Journal of Pharmaceutics,
2002. 241(1): p. 155-163.

114. Shawesh, A., et al., Influence of storage time and temperature on the stability of
indomethacin Pluronic® F-127 gels. Pharmazie, 2002. 57(10): p. 690-694.

115. Yong, C.S., et al., Effect of sodium chloride on the gelation temperature, gel
strength and bioadhesive force of poloxamer gels containing diclofenac sodium.
International Journal of Pharmaceutics, 2001. 226(1-2): p. 195-205.

116. Pisal, S.S., et al., Pluronic gels for nasal delivery of Vitamin B12. Part I:
Preformulation study. International Journal of Pharmaceutics, 2004. 270(1-2): p.
37-45.

206
REFERENCES

117. Wanka, G., H. Hoffmann, and W. Ulbricht, Phase diagrams and aggregation
behavior of poly (oxyethylene)-poly (oxypropylene)-poly (oxyethylene) triblock
copolymers in aqueous solutions. Macromolecules, 1994. 27(15): p. 4145-4159.

118. Prud'homme, R.K., G. Wu, and D.K. Schneider, Structure and rheology studies of
poly (oxyethylene-oxypropylene-oxyethylene) aqueous solution. Langmuir, 1996.
12(20): p. 4651-4659.

119. Alexandridis, P., J.F. Holzwarth, and T.A. Hatton, Micellization of Poly(ethylene
oxide)-Poly(propylene oxide)-Poly(ethylene oxide) Triblock Copolymers in
Aqueous Solutions: Thermodynamics of Copolymer Association. Macromolecules,
1994. 27(9): p. 2414-2425.

120. Cabana, A., A. Aït-Kadi, and J. Juhász, Study of the Gelation Process of
Polyethylene Oxidea-Polypropylene Oxideb-Polyethylene OxideaCopolymer
(Poloxamer 407) Aqueous Solutions. Journal of Colloid and Interface Science,
1997. 190(2): p. 307-312.

121. Vadnere, M., et al., Thermodynamic studies on the gel-sol transition of some
pluronic polyols. International Journal of Pharmaceutics, 1984. 22(2-3): p. 207-
218.

122. Anderson, B.C., N.K. Pandit, and S.K. Mallapragada, Understanding drug release
from poly(ethylene oxide)-b-poly(propylene oxide)-b-poly(ethylene oxide) gels.
Journal of Controlled Release, 2001. 70(1-2): p. 157-167.

123. Miyazaki, S., et al., Percutaneous absorption of indomethacin from pluronic F127
gels in rats. Journal of pharmacy and pharmacology, 1995. 47(6): p. 455-457.

124. Wang, Y.Y., et al., In vitro and in vivo evaluations of topically applied capsaicin
and nonivamide from hydrogels. International Journal of Pharmaceutics, 2001.
224(1-2): p. 89-104.

125. El-Kattan, A.F., et al., Effect of formulation variables on the percutaneous


permeation of ketoprofen from gel formulations. Drug Delivery, 2000. 7(3): p.
147-153.

126. Chi, S.C., H.K. Tan, and H.W. Chun, Antiinflammatory and analgesic
transdermal gel. 1996, Google Patents.

127. Fang, J.Y., et al., In vitro topical application and in vivo pharmacodynamic
evaluation of nonivamide hydrogels using Wistar rat as an animal model.
European journal of pharmaceutical sciences, 2002. 15(5): p. 417-423.

207
REFERENCES

128. Shin, S.C., C.W. Cho, and I.J. Oh, Effects of non-ionic surfactants as permeation
enhancers towards piroxicam from the poloxamer gel through rat skins.
International Journal of Pharmaceutics, 2001. 222(2): p. 199-203.

129. Liaw, J. and Y.C. Lin, Evaluation of poly (ethylene oxide)-poly (propylene oxide)-
poly (ethylene oxide)(PEO-PPO-PEO) gels as a release vehicle for percutaneous
fentanyl. Journal of Controlled Release, 2000. 68(2): p. 273-282.

130. Escobar-Chávez, J.J., D. Quintanar-Guerrero, and A. Ganem-Quintanar, In vivo


skin permeation of sodium naproxen formulated in pluronic F-127 gels: effect of
Azone® and Transcutol®. Drug Development and Industrial Pharmacy, 2005.
31(4-5): p. 447-454.

131. Erukova, V.Y., et al., Effect of ethylene oxide and propylene oxide block
copolymers on the permeability of bilayer lipid membranes to small solutes
including doxorubicin. Biochimica et Biophysica Acta (BBA)-Biomembranes,
2000. 1468(1-2): p. 73-86.

132. Pillai, O. and R. Panchagnula, Transdermal delivery of insulin from poloxamer


gel: ex vivo and in vivo skin permeation studies in rat using iontophoresis and
chemical enhancers. Journal of Controlled Release, 2003. 89(1): p. 127-140.

133. Nair, V. and R. Panchagnula, Poloxamer gel as vehicle for transdermal


iontophoretic delivery of arginine vasopressin: evaluation of in vivo performance
in rats. Pharmacological research, 2003. 47(6): p. 555-562.

134. Zhang, L., et al., Development and in-vitro evaluation of sustained release
Poloxamer 407 (P407) gel formulations of ceftiofur. Journal of Controlled
Release, 2002. 85(1-3): p. 73-81.

135. Fowler, E.B., et al., Evaluation of pluronic polyols as carriers for grafting
materials: study in rat calvaria defects. Journal of periodontology, 2002. 73(2): p.
191-197.

136. Shin, S.C. and J.Y. Kim, Enhanced permeation of triamcinolone acetonide
through the buccal mucosa. European Journal of Pharmaceutics and
Biopharmaceutics, 2000. 50(2): p. 217-220.

137. Morishita, M., et al., Pluronic® F-127 gels incorporating highly purified
unsaturated fatty acids for buccal delivery of insulin. International Journal of
Pharmaceutics, 2001. 212(2): p. 289-293.

208
REFERENCES

138. Bourre, L., et al., Potential efficacy of a delta 5-aminolevulinic acid thermosetting
gel formulation for use in photodynamic therapy of lesions of the gastrointestinal
tract. Pharmacological research, 2002. 45(2): p. 159-165.

139. Miyazaki, S., et al., Pluronic F-127 gels as a vehicle for topical administration of
anticancer agents. Chemical & pharmaceutical bulletin, 1984. 32(10): p. 4205.

140. Gilbert, J.C., et al., Drug release from Pluronic F-127 gels. International Journal
of Pharmaceutics, 1986. 32(2-3): p. 223-228.

141. Nalbandian, R.M., et al., Pluronic F-127 gel preparation as an artificial skin in
the treatment of third†degree burns in pigs. Journal of Biomedical Materials
Research, 1987. 21(9): p. 1135-1148.

142. Hokett, S.D., et al., Pluronic polyol effects on human gingival fibroblast
attachment and growth. Journal of periodontology, 2000. 71(5): p. 803-809.

143. Charrueau, C., et al., Poloxamer 407 as a thermogelling and adhesive polymer for
rectal administration of short-chain fatty acids. Drug Development and Industrial
Pharmacy, 2001. 27(4): p. 351-357.

144. Miyazaki, S., et al., Pluronic F-127 gels as a novel vehicle for rectal
administration of indomethacin. Chemical & pharmaceutical bulletin, 1986.
34(4): p. 1801.

145. Desai, S.D. and J. Blanchard, Pluronic® F127-based ocular delivery system
containing biodegradable polyisobutylcyanoacrylate nanocapsules of pilocarpine.
Drug Delivery, 2000. 7(4): p. 201-207.

146. El-Kamel, A.H., In vitro and in vivo evaluation of Pluronic F127-based ocular
delivery system for timolol maleate. International Journal of Pharmaceutics, 2002.
241(1): p. 47-55.

147. Shastri, D.H., S.T. Prajapati, and L.D. Patel, Design and development of
thermoreversible ophthalmic in situ hydrogel of moxifloxacin HCl. Current Drug
Delivery. 7(3): p. 238-243.

148. Mansour, M., et al., Ocular poloxamer-based ciprofloxacin hydrochloride in situ


forming gels. Drug Development and Industrial Pharmacy, 2008. 34(7): p. 744-
752.

149. Miyazaki, S., et al., Antitumor effect of PF-127 gel containing mytomicin C on
sarcoma-180 ascites tumor in mice. Chem Pharm. Bull, 1986. 34: p. 2224-2226.

209
REFERENCES

150. Paavola, A., et al., Controlled release injectable liposomal gel of ibuprofen for
epidural analgesia. International Journal of Pharmaceutics, 2000. 199(1): p. 85-
93.

151. Amiji, M.M., et al., Intratumoral administration of paclitaxel in an in situ gelling


poloxamer 407 formulation. Pharmaceutical development and Technology, 2002.
7(2): p. 195-202.

152. Paustian, P.W., et al., Intravenous Pluronic F-127 in early burn wound treatment
in rats. Burns, 1993. 19(3): p. 187-191.

153. Westerink, J., et al., ProJuvant (TM)(Pluronic F127®/chitosan) enhances the


immune response to intranasally administered tetanus toxoid. Vaccine, 2001.
20(5-6): p. 711-723.

154. Embuscado, M.E., Edible films and coatings for food applications. 2009: Springer
Verlag.

155. Guenet, J.M., Thermoreversible gelation of polymers and biopolymers. 1992:


Academic Pr.

156. Haque, A. and E.R. Morris, Thermogelation of methylcellulose. Part I: molecular


structures and processes. Carbohydrate Polymers, 1993. 22(3): p. 161-173.

157. Badiger, M.V., A. Lutz, and B.A. Wolf, Interrelation between the thermodynamic
and viscometric behaviour of aqueous solutions of hydrophobically modified ethyl
hydroxyethyl cellulose. Polymer, 2000. 41(4): p. 1377-1384.

158. Tomsic, B., et al., Biodegradability of cellulose fabric modified by


imidazolidinone. Carbohydrate Polymers, 2007. 69(3): p. 478-488.

159. Miyamoto, T., et al., Tissue biocompatibility of cellulose and its derivatives.
Journal of Biomedical Materials Research, 1989. 23(1): p. 125-133.

160. Muller, F.A., et al., Cellulose-based scaffold materials for cartilage tissue
engineering. Biomaterials, 2006. 27(21): p. 3955-3963.

161. Sarkar, N., Thermal gelation properties of methyl and hydroxypropyl


methylcellulose. Journal of Applied Polymer Science, 1979. 24(4): p. 1073-1087.

162. Nitis, S., Kinetics of thermal gelation of methylcellulose and


hydroxypropylmethylcellulose in aqueous solutions. Carbohydrate Polymers,
1995. 26(3): p. 195-203.

210
REFERENCES

163. Tate, M.C., et al., Biocompatibility of methylcellulose-based constructs designed


for intracerebral gelation following experimental traumatic brain injury.
Biomaterials, 2001. 22(10): p. 1113-1123.

164. Chen, C.H., et al., Novel living cell sheet harvest system composed of
thermoreversible methylcellulose hydrogels. Biomacromolecules, 2006. 7(3): p.
736-743.

165. Stabenfeldt, S.E., A.J. GarcÃ-a, and M.C. LaPlaca, Thermoreversible


laminin†functionalized hydrogel for neural tissue engineering. Journal of
Biomedical Materials Research Part A, 2006. 77(4): p. 718-725.

166. Upadrashta, S.M., P.R. Katikaneni, and N.O. Nuessle, Chitosan as a tablet
binder. Drug Development and Industrial Pharmacy, 1992. 18(15): p. 1701-1708.

167. Nystrom, C., J. Mazur, and J. Sjogren, Studies on direct compression of tablets II.
The influence of the particle size of a dry binder on the mechanical strength of
tablets. International Journal of Pharmaceutics, 1982. 10(3): p. 209-218.

168. Reading, S.J. and M.S. Spring, The effects of binder film characteristics on
granule and tablet properties. Journal of pharmacy and pharmacology, 1984.
36(7): p. 421-426.

169. Sheth, P.R. and J.L. Tossounian, Novel sustained release tablet formulations.
1979, Google Patents.

170. Casillan, A.B. and W.F. Frazier, Sucralfate suspension for use in treating ulcers.
1992, EP Patent 0,136,100.

171. Shah, N.B. and B.B. Sheth, Effect of polymers on dissolution from drug
suspensions. Journal of Pharmaceutical Sciences, 1976. 65(11): p. 1618-1623.

172. Al-Kassas, R.S. and M.M. El-Khatib, Ophthalmic controlled release in situ
gelling systems for ciprofloxacin based on polymeric carriers. Drug Delivery,
2009. 16(3): p. 145-152.

173. Sultana, Y., et al., Evaluation of carbopol-methyl cellulose based sustained-


release ocular delivery system for pefloxacin mesylate using rabbit eye model.
Pharmaceutical development and Technology, 2006. 11(3): p. 313-319.

174. Valenta, C., The use of mucoadhesive polymers in vaginal delivery. Advanced
Drug Delivery Reviews, 2005. 57(11): p. 1692-1712.

211
REFERENCES

175. Elhady, S.S.A., et al., Development of in situ gelling and mucoadhesive


mebeverine hydrochloride solution for rectal administration. Saudi
Pharmaceutical Journal, 2003. 11(4): p. 159-171.

176. El-Kamel, A. and M. El-Khatib, Thermally reversible in situ gelling


carbamazepine liquid suppository. Drug Delivery, 2006. 13(2): p. 143-148.

177. Choi, H.G., et al., Development of in situ-gelling and mucoadhesive


acetaminophen liquid suppository. International Journal of Pharmaceutics, 1998.
165(1): p. 33-44.

178. The United States Pharmacopeial Convention. USP31/NF26. 2007, Port City
Press: Baltimore. p.319–323.

179. Masae, T., S. Masako, and Y. Jun, Thermoreversible gelation and phase
separation in aqueous methyl cellulose solutions. Journal of Polymer Science Part
B: Polymer Physics, 2001. 39(1): p. 91-100.

180. Ford, J.L., Thermal analysis of hydroxypropylmethylcellulose and


methylcellulose: powders, gels and matrix tablets. International Journal of
Pharmaceutics, 1999. 179(2): p. 209-228.

181. Li, C.L., et al., The use of hypromellose in oral drug delivery. Journal of
pharmacy and pharmacology, 2005. 57(5): p. 533-546.

182. Liu, C.H., et al., In-vitro and In-vivo Studies of the Diclofenac Sodium Controlled
release Matrix Tablets. Journal of pharmacy and pharmacology, 1995. 47(5): p.
360-364.

183. Genc, L. and E. Jalvand, Preparation and in vitro evaluation of controlled release
hydrophilic matrix tablets of ketorolac tromethamine using factorial design. Drug
Development and Industrial Pharmacy, 2008. 34(8): p. 903-910.

184. Shanmugam, S., et al., Formulation and evaluation of sustained release matrix
tablets of Losartan potassium. International Journal of PharmTech Research. 3(1):
p. 526-534.

185. Kar, R.K., S. Mohapatra, and B.B. Barik, Design and characterization of
controlled release matrix tablets of Zidovudine. Asian Journal of Pharmaceutical
and Clinical Research, 2009. 2(2): p. 54-61.

212
REFERENCES

186. Hiremath, P.S. and R.N. Saha, Controlled release hydrophilic matrix tablet
formulations of isoniazid: Design and in vitro studies. Aaps Pharmscitech, 2008.
9(4): p. 1171-1178.

187. Iqbal, Z., et al., Preparation and In-Vitro Evaluation of Sustained Release Matrix
Diclofenac Sodium Tablets Using HPMC KM 100 and Gums. Archives of
Pharmacy Practice. 1.

188. Badshah, A., et al., Development of controlled-release matrix tablet of


risperidone: influence of methocel®-and ethocel®-based novel polymeric
blend on in vitro drug release and bioavailability. Aaps Pharmscitech: p. 1-9.

189. Badshah, A., F. Subhan, and K. Rauf, Controlled release matrix tablets of
olanzapine: influence of polymers on the in vitro release and bioavailability. Aaps
Pharmscitech: p. 1-8.

190. Chiwele, I., B.E. Jones, and F. Podczeck, The shell dissolution of various empty
hard capsules. Chemical and pharmaceutical bulletin -TOKYO-, 2000. 48(7): p.
951-956.

191. Ciper, M. and R. Bodmeier, Modified conventional hard gelatin capsules as fast
disintegrating dosage form in the oral cavity. European Journal of Pharmaceutics
and Biopharmaceutics, 2006. 62(2): p. 178-184.

192. Tochio, S., S. Nagata, and S. Yamashita, The influence of the composition of test
fluids on dissolution from HPMC capsules. AAPS PharmSci, 2002. 4(4): p.
W4340.

193. Jain, S.P., et al., In situ ophthalmic gel of ciprofloxacin hydrochloride for once a
day sustained delivery. Drug Development and Industrial Pharmacy, 2008. 34(4):
p. 445-452.

194. Liu, Z., et al., Study of an alginate/HPMC-based in situ gelling ophthalmic


delivery system for gatifloxacin. International Journal of Pharmaceutics, 2006.
315(1-2): p. 12-17.

195. Srividya, B., R.M. Cardoza, and P.D. Amin, Sustained ophthalmic delivery of
ofloxacin from a pH triggered in situ gelling system. Journal of Controlled
Release, 2001. 73(2-3): p. 205-211.

196. Catella-Lawson, F., et al., Cyclooxygenase inhibitors and the antiplatelet effects
of aspirin. New England Journal of Medicine, 2001. 345(25): p. 1809-1817.

213
REFERENCES

197. Reynolds, J.E.F. and K. Parfitt, eds. Martindale; Extra pharmacopoeia. 28 ed.
1989, The Pharmaceutical Press. 250.

198. Todd, P.A. and E.M. Sorkin, Diclofenac sodium. A reappraisal of its
pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy.
Drugs, 1988. 35(3): p. 244.

199. Brogden, R.N., et al., Diclofenac sodium: a review of its pharmacological


properties and therapeutic use in rheumatic diseases and pain of varying origin.
Drugs, 1980. 20(1): p. 24.

200. Sallmann, A.R., The history of diclofenac. The American Journal of Medicine,
1986. 80(4): p. 29-33.

201. Schubiger, B.I., S.N. Ciccolunghi, and K. Tanner, Once daily dose treatment with
a non-steroidal anti-rheumatic drug (diclofenac) in osteoarthrosis. The Journal of
international medical research, 1980. 8(2): p. 167.

202. Vetter, G., A comparison of naproxen and diclofenac sodium in the treatment of
osteoarthritis in elderly patients. The British journal of clinical practice, 1985.
39(7): p. 276.

203. Calabro, J.J., Efficacy of diclofenac in ankylosing spondylitis. The American


Journal of Medicine, 1986. 80(4): p. 58-63.

204. Sieper, J., et al., Ankylosing spondylitis: an overview. Annals of the Rheumatic
Diseases, 2002. 61(suppl 3): p. iii8.

205. Buller, H.R., et al., Antithrombotic therapy for venous thromboembolic disease.
Chest, 2004. 126(3 suppl): p. 401S-428S.

206. Haroutiunian, S., D.A. Drennan, and A.G. Lipman, Topical NSAID therapy for
musculoskeletal pain. Pain Medicine. 11(4): p. 535-549.

207. McKenna, F., Efficacy and gastroduodenal safety of a fixed combination of


diclofenac and misoprostol in the treatment of arthritis. Rheumatology, 1995.
34(suppl 1): p. 11-18.

208. Ciccolunghi, S.N., et al., Report on a long-term tolerability study of up to two


years with diclofenac sodium (Voltaren). Scandinavian Journal of Rheumatology,
1978. 7(S22): p. 86-96.

214
REFERENCES

209. Nsaid, N., Cyclooxygenase-2 Enzyme Inhibitors: Place in Therapy. Am Fam


Physician, 2000. 61(12): p. 3669-3676.

210. Riess, W., et al., Pharmacokinetics and metabolism of the anti-inflammatory


agent Voltaren. Scandinavian Journal of Rheumatology, 1978. 7(S22): p. 17-29.

211. John, V.A., The pharmacokinetics and metabolism of diclofenac sodium


(Voltarol) in animals and man. Rheumatology and rehabilitation, 1979: p. 22.

212. Willis, J.V., et al., The pharmacokinetics of diclofenac sodium following


intravenous and oral administration. European journal of clinical pharmacology,
1979. 16(6): p. 405-410.

213. Kendall, M.J., D.P. Thornhill, and J.V. Willis, Factors affecting the
pharmacokinetics of diclofenac sodium (Voltarol). Rheumatology and
rehabilitation, 1979: p. 38.

214. Fowler, P.D., et al., Plasma and synovial fluid concentrations of diclofenac
sodium and its hydroxylated metabolites during once-daily administration of a
100 mg slow-release formulation. European journal of clinical pharmacology,
1986. 31(4): p. 469-472.

215. Kienzler, J.L., M. Gold, and F. Nollevaux, Systemic bioavailability of topical


diclofenac sodium gel 1% versus oral diclofenac sodium in healthy volunteers.
The Journal of Clinical Pharmacology. 50(1): p. 50.

216. Gaucher, A., et al., Passage of diclofenac sodium in the liquid synovial. Therapie,
1983. 38(4): p. 431-434.

217. Benson, M.D., M. Aldo-Benson, and K.D. Brandt. Synovial fluid concentrations
of diclofenac in patients with rheumatoid arthritis or osteoarthritis. 1985.

218. Chan, K.K.H., K.H. Vyas, and K.D. Brandt, In vitro protein binding of diclofenac
sodium in plasma and synovial fluid. Journal of Pharmaceutical Sciences, 1987.
76(2): p. 105-108.

219. Fowler, P.D., et al., Plasma and synovial fluid concentrations of diclofenac
sodium and its major hydroxylated metabolites during long-term treatment of
rheumatoid arthritis. European journal of clinical pharmacology, 1983. 25(3): p.
389-394.

215
REFERENCES

220. Russeva, V.N. and Z.D. Zhivkova, Protein binding of some nonsteroidal anti-
inflammatory drugs studied by high-performance liquid affinity chromatography.
International Journal of Pharmaceutics, 1999. 180(1): p. 69-74.

221. Chamouard, J.M., et al., Diclofenac binding to albumin and lipoproteins in human
serum. Biochemical pharmacology, 1985. 34(10): p. 1695-1700.

222. Stierlin, H., et al., Biotransformation of diclofenac sodium (Voltaren) in animals


and in man. I. Isolation and identification of principal metabolites. Xenobiotica;
the fate of foreign compounds in biological systems, 1979. 9(10): p. 601.

223. Willis, J.V. and M.J. Kendall, Pharmacokinetic studies on diclofenac sodium in
young and old volunteers. Scandinavian Journal of Rheumatology, 1978. 7(S22):
p. 36-41.

224. Crook, P.R., et al., The pharmacokinetics of diclofenac sodium in patients with
active rheumatoid disease. European journal of clinical pharmacology, 1982.
21(4): p. 331-334.

225. Weinberger, M., Analgesic sensitivity in children with asthma. Pediatrics, 1978.
62(5s): p. 910.

226. VanArsdel Jr, P.P., Aspirin idiosyncracy and tolerance. The Journal of allergy
and clinical immunology, 1984. 73(4): p. 431.

227. Stevenson, D.D., Diagnosis, prevention, and treatment of adverse reactions to


aspirin and nonsteroidal anti-inflammatory drugs. Journal of Allergy and Clinical
Immunology, 1984. 74(4): p. 617-622.

228. Bendix, T., et al., Diclofenac (Voltaren) and ketoprofen (Orudis), in rheumatoid
arthritis: a randomized double-blind multicentre trial. Curr Ther Res, 1983. 33: p.
192-9.

229. Aylward, M., et al., Mefenamic acid and diclofenac sodium in osteoarthritis of the
weight bearing joints: a double blind comparison. The British journal of clinical
practice, 1985. 39(4): p. 135.

230. Machtey, I. Diclofenac in the treatment of painful joints and traumatic tendinitis
(including strains and sprains): a brief review. 1985.

231. Menasse, R., et al., Pharmacological properties of diclofenac sodium and its
metabolites. Scandinavian Journal of Rheumatology, 1978. 7(S22): p. 5-16.

216
REFERENCES

232. Willis, J.V., M.J. Kendall, and D.B. Jack, A study of the effect of aspirin on the
pharmacokinetics of oral and intravenous diclofenac sodium. European journal of
clinical pharmacology, 1980. 18(5): p. 415-418.

233. Muler, F.O., H.K. Hundt, and D.G. Muller, Pharmacokinetic and
pharmacodynamic implications of long-term administration of non-steroidal anti-
inflammatory agents. International journal of clinical pharmacology and
biopharmacy, 1977. 15(9): p. 397.

234. Bird, H.A., et al., A study to determine the clinical relevance of the
pharmacokinetic interaction between aspirin and diclofenac. Inflammation
Research, 1986. 18(3): p. 447-449.

235. Jenkins, J.K. and P.J. Seligman, Analysis and recommendations for Agency action
regarding non-steroidal anti-inflammatory drugs and cardiovascular risk. FDA
memorandum, Center for Drug Evaluation and Research, Office of New Drugs,
2005.

236. Craig Brater, D., Drug-drug and drug-disease interactions with nonsteroidal anti-
inflammatory drugs. The American Journal of Medicine, 1986. 80(1): p. 62-77.

237. Reimann, I.W. and J.C. Frölich, Effects of diclofenac on lithium kinetics.
Clinical Pharmacology & Therapeutics, 1981. 30(3): p. 348-352.

238. Zachariae, H., Methotrexate and non†steroidal anti†inflammatory drugs.
British Journal of Dermatology, 1992. 126(1): p. 95-95.

239. Segev, S., M. Rehavi, and E. Rubinstein, Quinolones, theophylline, and


diclofenac interactions with the gamma-aminobutyric acid receptor.
Antimicrobial agents and chemotherapy, 1988. 32(11): p. 1624.

240. Davies, B.I. and F.P.V. Maesen, Drug interactions with quinolones. Review of
Infectious Diseases, 1989. 11(Supplement 5): p. S1083.

241. Iqbal, Z., et al., Pharmacokinetic interaction of ciprofloxacin with diclofenac: a


single-dose, two-period crossover study in healthy adult volunteers. Clinical drug
investigation, 2009. 29(4): p. 275-281.

242. Budavari, S., M.J. O'Neil, and P.E. Heckelman, The merck index an encyclopedia
of chemical, drugs, and biologicals. 1989.

243. Reynolds'Martindale, J.E.F., The Extra Pharmacopoeia'1989, The pharmaceutical


Press, LONDON.

217
REFERENCES

244. Fourtillan, J.B., et al., Pharmacokinetics of oral timolol studied by mass


fragmentography. European journal of clinical pharmacology, 1981. 19(3): p.
193-196.

245. Nieminen, T., et al., Ophthalmic timolol: plasma concentration and systemic
cardiopulmonary effects. Scandinavian journal of clinical & laboratory
investigation, 2007. 67(2): p. 237-245.

246. Tocco, D.J., et al., Physiological disposition and metabolism of timolol in man
and laboratory animals. Drug Metabolism and Disposition, 1975. 3(5): p. 361-
370.

247. Zimmerman, T.J. and H.E. Kaufman, Timolol: Dose response and duration of
action. Archives of Ophthalmology, 1977. 95(4): p. 605.

248. Wilson, T.W., et al., Timolol and propranolol: bioavailability, plasma


concentrations, and beta blockade. Clinical Pharmacology & Therapeutics, 1982.
32(6): p. 676-685.

249. Vermeij, P., M. Sherbini†Schepers, and P.A. Zwieten, The disposition of
timolol in man. Journal of pharmacy and pharmacology, 1978. 30(1): p. 53-55.

250. Vlasses, P.H., et al., Initial Evaluation of Transdermal Timolol: Serum


Concentrations and [beta]-Blockade. Journal of cardiovascular pharmacology,
1985. 7(2): p. 245.

251. Lustgarten, J.S. and S.M. Podos, Topical timolol and the nursing mother.
Archives of Ophthalmology, 1983. 101(9): p. 1381.

252. Zimmerman, T.J., J.D. Baumann, and J. Hetherington Jr, Side effects of timolol.
Survey of Ophthalmology, 1983. 28: p. 243-249.

253. Zimmerman, T.J., B.J. Leader, and D.S. Golob, Potential side effects of timolol
therapy in the treatment of glaucoma. Annals of ophthalmology, 1981. 13(6): p.
683.

254. Pickup, J.C.a.G.W., Eds., Textbook of Diabetes. 2nd ed. Vol. 1. 1997, Malden:
MA: Blackwell Science Ltd.

255. L.L. Brunton, E., The Pharmacological Basis of Therapeutics. 11 ed. Insulin, Oral
Hypoglycemic Agents, and the Pharmacology of the Endocrine Pancreas, ed. S.N.
Davis. 2006: McGraw-Hill.

218
REFERENCES

256. Volund, A., Conversion of insulin units to SI units. The American Journal of
Clinical Nutrition, 1993. 58(5): p. 714-715.

257. B.G. Katzung, E., Basic and Clinical Pharmacology. 10 ed. Pancreatic Hormones
& Anitdiabetic Drugs, ed. M.S. Nolte, Karam, John H. 2007, New York:
McGraw-Hill.

258. Dean, L. and J. McEntyre, The genetic landscape of diabetes. 2004.

259. Arbit, E., The physiological rationale for oral insulin administration. Diabetes
Technology and Therapeutics, 2004. 6(4): p. 510-517.

260. Duckworth, W.C., R.G. Bennett, and F.G. Hamel, Insulin degradation: progress
and potential. Endocrinology Review, 1998. 19(5): p. 608-624.

261. Schmolka, I.R., Artificial skin I. Preparation and properties of pluronic F†127
gels for treatment of burns. Journal of Biomedical Materials Research, 1972. 6(6):
p. 571-582.

262. Tung, I., Rheological behavior of poloxamer 407 aqueous solutions during sol-gel
and dehydration processes. International Journal of Pharmaceutics, 1994. 107(2):
p. 85-90.

263. Copetti, G., et al., Synergistic gelation of xanthan gum with locust bean gum: a
rheological investigation. Glycoconjugate journal, 1997. 14(8): p. 951-961.

264. Owen, D.H., J.J. Peters, and D.F. Katz, Rheological properties of contraceptive
gels. Contraception, 2000. 62(6): p. 321-326.

265. Wei, G., et al., Thermosetting gels with modulated gelation temperature for
ophthalmic use: the rheological and gamma scintigraphic studies. Journal of
Controlled Release, 2002. 83(1): p. 65-74.

266. Nasir, F., et al., Simultaneous determination of timolol maleate, rosuvastatin


calcium and diclofenac sodium in pharmaceuticals and physiological fluids using
HPLC-UV. Journal of chromatography. B, 2011. 879(30): p. 3434-3443.

267. T. Moore, S.C., S. Mallapragada, and N. Pandit., Experimental investigation and


mathematical modeling of Pluronic F127 gel dissolution, drug release in stirred
systems. . J. Control.Release 2000. 67(2-3): p. 191-202.

219
REFERENCES

268. Desai, S.D. and J. Blanchard, In vitro evaluation of pluronic F127-based


controlled-release ocular delivery systems for pilocarpine. Journal of
Pharmaceutical Sciences, 1998. 87(2): p. 226-230.

269. Chandrashekhar G, U.N., Biodegradable injectable implant system for long term
drug delivery using poly (lactic-co-glycolic) acid copolymers. J Pharm
Pharmacol, 1998. 48: p. 669-674.

270. Ravi, S., et al., Development and validation of an HPLC-UV method for the
determination of insulin in rat plasma: Application to pharmacokinetic study.
Chromatographia, 2007. 66(9): p. 805-809.

271. Yu, A. and L. Shargel, Applied Biopharmaceutics & Pharmacokinetics. 1999,


McGraw-Hill New York:.

272. Épshtein, N.A., Validation of HPLC Techniques for Pharmaceutical Analysis.


Pharmaceutical Chemistry Journal, 2004. 38(4): p. 212-228.

273. Rassing, J. and D. Attwood, Ultrasonic velocity and light-scattering studies on


the polyoxyethylene--polyoxypropylene copolymer Pluronic F127 in aqueous
solution. International Journal of Pharmaceutics, 1982. 13(1): p. 47-55.

274. Dumortier, G., et al., A Review of Poloxamer 407 Pharmaceutical and


Pharmacological Characteristics. Pharmaceutical Research, 2006. 23(12): p.
2709-2728.

275. Veyries, M.L., et al., Controlled release of vancomycin from poloxamer 407 gels.
International Journal of Pharmaceutics, 1999. 192(2): p. 183-193.

276. Roy, J., et al., Diclofenac sodium injection sterilized by autoclave and the
occurrence of cyclic reaction producing a small amount of impurity. Journal of
Pharmaceutical Sciences, 2001. 90(5): p. 541-544.

277. Gong, C.Y., et al., Synthesis and characterization of PEG-PCL-PEG


thermosensitive hydrogel. International Journal of Pharmaceutics, 2009. 365(1-2):
p. 89-99.

278. El Gendy, A.M., H.W. Jun, and A.A. Kassem, In vitro release studies of
flurbiprofen from different topical formulations. Drug Development and Industrial
Pharmacy, 2002. 28(7): p. 823-831.

220
REFERENCES

279. Anderson, B.C., N.K. Pandit, and S.K. Mallapragada, Understanding drug release
from poly (ethylene oxide)-b-poly (propylene oxide)-b-poly (ethylene oxide) gels.
Journal of Controlled Release, 2001. 70(1-2): p. 157-167.

280. Alexandridis, P. and T. Alan Hatton, Poly (ethylene oxide)---poly (propylene


oxide)---poly (ethylene oxide) block copolymer surfactants in aqueous solutions
and at interfaces: thermodynamics, structure, dynamics, and modeling. Colloids
and Surfaces A: Physicochemical and Engineering Aspects, 1995. 96(1-2): p. 1-
46.

281. Bhardwaj, R. and J. Blanchard, Controlled release delivery system for the MSH
analog Melanotan†I using poloxamer 407. Journal of Pharmaceutical
Sciences, 1996. 85(9): p. 915-919.

282. Marcel Dekker, ed. Encyclopedia of Pharmaceutical Technology. Gels and jellies,
ed. J. Swarbrick, Boylan, J.C. Vol. 6. 1992. : New York. 418–419.

283. Paavola, A., J. Yliruusi, and P. Rosenberg, Controlled release and dura mater
permeability of lidocaine and ibuprofen from injectable poloxamer-based gels.
Journal of Controlled Release, 1998. 52(1-2): p. 169-178.

284. Pearlman, R., Stability and characterization of protein and peptide drugs: case
histories. Vol. 5. 1993: Springer.

285. Guzman, M., et al., Polyoxyethylene-polyoxypropylene block copolymer gels as


sustained release vehicles for subcutaneous drug administration. International
Journal of Pharmaceutics, 1992. 80(1): p. 119-127.

286. Chen-Chow, P.C. and S.G. Frank, In vitro release of lidocaine from Pluronic F-
127 gels. International Journal of Pharmaceutics, 1981. 8(2): p. 89-99.

287. Lu, G. and H.W. Jun, Diffusion studies of methotrexate in Carbopol and
Poloxamer gels. International Journal of Pharmaceutics, 1998. 160(1): p. 1-9.

288. Nishihata, T., et al., Clinical investigation of sodium diclofenac sustained-release


suppositories. International Journal of Pharmaceutics, 1988. 42(1-3): p. 251-256.

289. Durrani, A.M., S.J. Farr, and I.W. Kellaway, Influence of molecular weight and
formulation pH on the precorneal clearance rate of hyaluronic acid in the rabbit
eye. International Journal of Pharmaceutics, 1995. 118(2): p. 243-250.

221
REFERENCES

290. Wei, G., et al., Pharmacokinetics of timolol in aqueous humor sampled by


microdialysis after topical administration of thermosetting gels. Biomedical
Chromatography, 2006. 20(1): p. 67-71.

291. Zaki, N.M., et al., Enhanced bioavailability of metoclopramide HCl by intranasal


administration of a mucoadhesive in situ gel with modulated rheological and
mucociliary transport properties. European journal of pharmaceutical sciences,
2007. 32(4-5): p. 296-307.

292. Hoffman, A. and E. Ziv, Pharmacokinetic considerations of new insulin


formulations and routes of administration. Clinical pharmacokinetics, 1997.
33(4): p. 285.

222
ANNEXURE

PUBLICATION FROM THESIS


ANNEXURE

1. Nasir F, Iqbal Z, Khan A, Ahmad L, Shah Y, Khan AZ, Khan JA, Khan S.
Simultaneous determination of timolol maleate, rosuvastatin calcium and
diclofenac sodium in pharmaceuticals and physiological fluids using HPLC-UV.
Journal of Chromatography B. 2011 Nov 15;879(30):3434-43.

Você também pode gostar