You are on page 1of 359

SELECTION OF THE BEST VESICULAR CARRIER SYSTEM

FOR ENHANCED TRANSDERMAL DRUG DELIVERY: A


COMPARATIVE STUDY

A Thesis submitted to the


Jawaharlal Nehru Technological University Anantapur
For the award of

DOCTOR OF PHILOSOPHY
in
PHARMACEUTICAL SCIENCES

By
K. SRIKANTH
[Reg. No. 0900PH1309]

Under the guidance of

Dr. V. RAMA MOHAN GUPTA & Dr. DEVANNA NAYAKANTI

RESEARCH AND DEVELOPMENT


JAWAHARLAL NEHRU TECHNOLOGICAL UNIVERSITY ANANTAPUR
ANANTHAPURAMU – 515002, A.P., INDIA
JULY-2014
DECLARATION

I declare that the thesis entitled “SELECTION OF THE BEST VESICULAR

CARRIER SYSTEM FOR ENHANCED TRANSDERMAL DRUG DELIVERY: A

COMPARATIVE STUDY” has been prepared by me under the guidance of Dr. V.

RAMA MOHAN GUPTA, Professor & Principal, Pulla Reddy Institute of

Pharmacy, Medak, Hyderabad and Dr. N. Devanna, Director, JNTUA-OTRI,

Ananthapuramu. No part of this thesis has formed the basis for the award of end

degree or fellowship previously.

K.SRIKANTH

Dept of Pharmaceutical sciences,

Jawaharlal Nehru Technological University,Anantapur,

Ananthapuramu.

Dt:
CERTIFICATE

We certify that K. Srikanth has preparaed his thesis entitled “SELECTION

OF THE BEST VESICULAR CARRIER SYSTEM FOR ENHANCED TRANSDERMAL

DRUG DELIVERY: A COMPARATIVE STUDY” for the award of Ph.D degree of the

Jawaharlal Nehru Technological University Anatapur, Ananthapuramu under our

guidance. He has carried out the work at the Dept of Pharmaceutics, Pulla Reddy

Institute of Pharmacy, Medak, Hyderabad, A.P, (INDIA.).


Signature of the co-guide Signature of the co-guide

Dr. N. Devanna Dr. V. Rama Mohan Gupta

Director, Professor & Principal

JNTUA-OTRI, Pulla Reddy Institute ofPharmacy,

Ananthapuramu Medak, Hyderabad


ACKNOWLEDGEMENT
It would not have been possible to do research work and to write my

doctoral thesis without the help and support of my god Saibaba. I don’t have words

to express my deep sense of gratitude towards my God Sai Baba for his blessings.

I have only option to make him happy is that following his words. I want to be a

dust particle under his feet forever.

At the outset, I would like to thank my supervisor Dr. V. Rama Mohan

Gupta, Principal, Pulla Reddy Institute of Pharmacy for his quick evaluation and

decision on my PhD application. In fact, it was a big surprise to receive his positive

reply within a few minutes after my request for the PhD position, grateful for his

confidence and trust on me. It is my privilege to have his encouragement and

galvanizing my spirit in accomplishing this project successfully. His valuable and

precious suggestions, criticisms have been guiding force throughout the process of

my work. The sincerity, righteousness and virtuousness which he has inculcated in

me will take me a long way in my life. During this period, I had a great freedom to

plan and execute my ideas in research without any pressure. This made me to

identify my own strength and drawbacks and particularly boosted my self-

confidence. It is a great pleasure working under his supervision and I would ever be

indebted to him for the unstinted support exleished to me during my Ph.D.

I will forever be thankful to my co-guide Dr. N. Devanna, Director, OTRI,

Anathapur for his timely help and sincere support during my research period. He

was and remains my best role model as a scientist, mentor, and teacher. His helping

hand is a source in crossing the barriers associated in completion of synopsis,

thesis, etc.
I take this opportunity to sincerely acknowledge Mr. CH. Sathi Reddy,

Chairman, Pulla Reddy Institute of Pharmacy, Medak and Mr. Aditya, Director,

Pulla Reddy Institute of Pharmacy, Medak for providing facilities to carry out

my research work at Pulla Reddy Institute of Pharmacy which buttressed me to

perform my research work comfortably.

I oblige my deep sense of gratitude to Mr. V.S. Prakash Rao,

Administrative Officer, Pulla Reddy Institute of Pharmacy, Medak for his

supportive suggestions and motivations from the commencement of my project

work. He shared his experiences which he had faced in his long tenure and offered

valuable suggestions to become ideal person.

I express my sincere gratitude to my colleagues Mr. Ashwaq Hussain, Mr.

Subhas Sahoo, Mrs. Srivani Voviliveni, Ms. Nikunja B.Pati, Mrs. Parimala devi,

and others who have been behind me and for their timely help during my Ph.D and

bench works.

I wish to thank Kremoint Pharma Pvt. Ltd, Thane, Maharastra and Lipoid

AG, Switzerland for providing gift samples of drugs and polymers.

I extend my sincere thanks to all the supporting staff members of my

college Mr. Srinivas, Mr. Omkar Rao, Mr. Siddaiah, Mrs. Sangeetha for their

involvement and friendly attitude during my work.

I would like to pay high regards to my Mother, Father and Brother for

their sincere encouragement and inspiration throughout my research work and

lifting me uphill this phase of life. I owe everything to them. Words fail me to

express my appreciation to my mother and father for their support, generous care.

A simple thanks doesn’t seem sufficient but it is said with appreciation and respect

to my brother who has been instrumental for the good shape of my thesis because

of his help and suggestions in formatting the entire thesis. During the inevitable
ups and downs of conducting my research he often reminded me life’s true

priorities by what could be the influence of a loving God Sai baba. A journey is

easier when you travel together. My wife always stood beside me during the happy

and hard moments to push me and motivate me. I am much indebted to my wife for

her love, care, support and creating a pleasant atmosphere at home during my

entire research period.

Besides this, my sincere thanks also goes to several people who have

knowingly and unknowingly helped me in the successful completion of this project.

K. SRIKANTH
ABSTRACT

The present research work aimed to select the best vesicular carrier

system among niosomes, ethosomes and transferosomes (primary objective) for

administration of drugs across the skin to produce systemic effect. Nystatin, a

polyene antifungal agent, was used as model drug in the present study. The

secondary objective of the study is enhancement of bioavailability of the

nystatin and to decrease the adverse effects associated with higher doses of the

same by administering through skin.

Niosomal and transferosomal suspensions of nystatin prepared by using

thin film hydration Technique whereas ethosomal suspensions of nystatin was

prepared method reported by Touitou et al. Totally 24 vesicular suspensions

(eight formulations from each type of vesicular systems) were prepared.

Niosomal suspensions were prepared using two different non-ionic

surfactants i.e span-80 and span-60. Each surfactant used at four different

concentrations (eight niosomal suspensions) was prepared. Similarly, eight

transferosomal and eight ethosomal suspensions were prepared.

Transferosomal suspensions were prepared using two different edge activators

i.e. span-60 and span-80 at four different concentrations whereas in

preparation of ethosomal suspensions ethanol and lecithin concentrations has

been changed for preparing the same (each at four different concentrations).

The prepared niosomal, ethosomal and transferosomal suspensions (24

formulations) evaluated for different parameters like vesicles morphology (size,

shape and surface texture), entrapment efficiency and in vitro drug release. The
drug release data fitted in different mathematical models such as Zero order,

First order, Higuchi, Hixon-crowel, Korsmeyer-peppas to find out the order and

mechanism of drug release from all formulations.

Based on the size, entrapment efficiency and invitro drug release results,

one optimized vesicular suspension from each type of vesicular formulation

was selected.

Elasticity and surface charge (Zeta potential) of the vesicles present in

three optimized vesicular suspensions was studied.

After performing the zetapotential and elasticity studies, using the

optimized niosomal, ethosomal and transferosomal suspensions niosomal,

ethosomal and transfersosmal gels having concentration of 0.01%w/w was

prepared respectively. Along with three vesicular nystatin gels, conventional

nystatin gel having same concentration was also prepared with pure nystatin

sample.

The prepared gels were characterized for in vitro and ex vivo drug release,

skin retention, stability studies. Finally in vivo studies also carried out to find

out the systemic availability of nystatin from individual gels in rabbits.

In vitro, ex vivo drug release data showed a good correlation between

each other. The study revealed that ethosomes are effective for permeation of

nystatin across the dialysis membrane and skin than other two vesicles.

Transdermal flux, permeation coefficient and enhancement ratio was

calculated from ex vivo drug release data of all three vesicular carrier gels. The

data showed higher efficiency to the ethosomes than other two vesicular gels.
Skin retention study was performed to find out the quantity of nystatin that

reached systemic circulation and retained in and on the skin after ex vivo drug

release study. The skin retention study also showed that ethosomes are

efficient than other two vesicular carriers i.e. transferosomes and niosomes.

In vivo study was also carried out in rabbits to find out the efficiency of

all vesicular gels and conventional gel in production of systemic effect. Highest

Cmax and AUC was exhibited by the ethosomes than other two vesicular and

conventional gels which stating that ethosomes are effective carriers to produce

systemic effect than other two carriers.

Our findings contribute to the evidence base for enhancing the

permeability of nystatin across the skin and to produce the systemic anti

fungal activity. (Secondary objective)

Key Words: transdermal drug delivery, systemic effect, localized effect,

niosomes, ethosomes, transferosomes, gels.


PREFACE

This Ph.D. work I have started by the grace of my god Sai baba and is

desire of my father. This thesis contains my five years research work which I

have carried out at Pulla Reddy Institute of Pharmacy, Medak. It was my

brother who made me a real person. He is the person who ignited passion in

me towards research. Without this foundation, I probably would not have dared to

study an emergent technology which, at the point I started this work, still had a very

unclear future. I would have never reached the point of finishing my dissertation

without the help and support of others.

These five years have been a challenging trip, with both ups and downs.

Fortunately, I was not alone on this road, but accompanied by an extended

team of experts, always willing to coach, sponsor, help, and motivate me. For

this, I would like to kindly thank them.

This thesis is divided into eight chapters. Chapter one focuses on the

general aspects of Drug Delivery and the need for various approaches to Novel

Drug Delivery Systems. It also provides an overview of transdermal drug

delivery that envisages briefly about vesicular carriers in drug delivery, through

skin. Chapter two includes a careful and systematic presentation of published

literature that has been reviewed to develop and evaluate the system designed

in the present investigation. Chapter three outlines the aim & objective of the

present investigation, preformulation studies of drug and various polymers

with their characterization. It deals with brief profiles of the drug, polymers,

Materials and methods adopted in the preparation and characterization of the


microspheres in the present work. Chapter four outlines the results of all

investigations obtained in an orderly manner. Chapter Five deals with

discussions of all results obtained in the investigation with their probable

implications. Chapter Six deals with summary and conclusion drawn from the

present work. Chapter seven gives list of Bibliography/References. Chapter

eight gives list of Publications & Conferences.


TABLE OF CONTENTS

Declaration

Certificate

Acknowledgements

Abstract

Preface

List of Tables

List of Figures

List of Abbreviations

List of Appendices

Chapter No. Name of the Chapter Page No.

1. Introduction

2. Literature Survey

3. Theoretical Analysis

4. Experimental Investigations

5. Experimental Results

6. Discussion of Results

7. Summary, Conclusion and Recommendations

8. References/Bibliography

Index
LIST OF TABLES
Table No Title of the table Pg.no
Chapter -1
1.1 Transdermal drugs approved by the US FDA
Chapter -3
3.5.1 List of chemicals
3.5.2 List of equipments
Chapter -5
5.1 Solubility of nystatin in Phosphate buffer saline solution pH 7.4
Standard curve of nystatin in Phosphate buffer
5.2
saline solution pH 7.4
5.3 Composition of niosomal formulations
5.4 Composition of ethosomal formulations
5.5 Composition of transferosomal formulations
Vesicles size & Entrapment efficiency of
5.6
all niosomal formulations
Vesicles size & Entrapment efficiency of
5.7
all ethosomal formulations
Vesicles size & Entrapment efficiency of all
5.8
transferosomal formulations
Summary of in vitro drug release from all niosomal
5.9
formulations across the dialysis membrane
5.10 In vitro drug release from niosomal formulation FN1
5.11 In vitro drug release from niosomal formulation FN2
5.12 In vitro drug release from niosomal formulation FN3
5.13 In vitro drug release from niosomal formulation FN4
5.14 In vitro drug release from niosomal formulation FN5
5.15 In vitro drug release from niosomal formulation FN6
5.16 In vitro drug release from niosomal formulation FN7
5.17 In vitro drug release from niosomal formulation FN8
Summary of release kinetics data of all nystatin
5.18
Niosomal Formulations
Summary of in vitro nystatin release from all ethosomal
5.19
formulations across the dialysis membrane
5.20 In vitro drug release from ethosomal formulation FE1
5.21 In vitro drug release from ethosomal formulation FE2
5.22 In vitro drug release from ethosomal formulation FE3
5.23 In vitro drug release from ethosomal formulation FE4
5.24 In vitro drug release from ethosomal formulation FE5
5.25 In vitro drug release from ethosomal formulation FE6
5.26 In vitro drug release from ethosomal formulation FE7
5.27 In vitro drug release from ethosomal formulation FE8
Summary of Release kinetics data of all
5.28
nystatin ethosomal suspension
Summary of in vitro nystatin release from all transferosomal
5.29
formulations across the dialysis membrane
5.30 In vitro drug release from transferosomal formulation FT1
5.31 In vitro drug release from transferosomal formulation FT2
5.32 In vitro drug release from transferosomal formulation FT3
5.33 In vitro drug release from transferosomal formulation FT4
5.34 In vitro drug release from transferosomal formulation FT5
5.35 In vitro drug release from transferosomal formulation FT6
5.36 In vitro drug release from transferosomal formulation FT7
5.37 In vitro drug release from transferosomal formulation FT8
Summary of Release kinetics data of all nystatin
5.38
transferosomal suspension
Elasticity measurements of three optimized vesicular
5.39
formulations
5.40 Zeta potential of three optimized vesicular formulations
Composition of conventional, niosomal, ethosomal
5.41
&transferosomal gels
In vitro drug release from conventional gel across
5.42
the dialysis membrane
In vitro drug release from niosomal gel across
5.43
the dialysis membrane
In vitro drug release from ethosomal gel across
5.44
the dialysis membrane
In vitro drug release from transferosomal gel across
5.45
the dialysis membrane
5.46 Ex vivo drug release from conventional gel across the rat skin
5.47 Ex vivo drug release from niosomal gel across the rat skin
5.48 Ex vivo drug release from ethosomal gel across the rat skin
5.49 Ex vivo drug release from transferosomal gel across the rat skin
Nystatin retention from all gels during ex vivo drug
5.50
release study in and on skin
5.51 Stability studies of optimized niosomal formulation gel (FN3)
5.52 Stability studies of optimized ethosomal formulation gel (FE6)
Stability studies of optimized transferosomal formulation gel
5.53
(FT7)
Analysis of ex vivo drug release study of three
5.54
optimized vesicular formulations
5.55 Calibration curve of the nystatin by HPLC
5.56 In vivo study of the conventional gel in rabbits
5.57 In vivo study of the niosomal gel in rabbits
5.58 In vivo study of the ethosomal gel in rabbits
5.59 In vivo study of the transferosomal gel in rabbits
Summary of the nystatin concentrations in blood plasma at
5.60
different time intervals (In vivo study)
5.61 Bioavailability studies of nystatin from all gels

LIST OF FIGURES
Figure
Title of the figure Pg.no
No
Chapter -1
1.1 Different routes of drug administration
1.2 Market segmentation the global drug delivery market in 2007
1.3 Topical and transdermal drug delivery systems
1.4 Number of Transdermal drugs approved in each year.
1.5 Structure of the skin
1.6 Pathways of drug absorption through the skin
1.7 Chemical structure of typical chemical penetration enhancers
1.8 Basic design of electroporation drug delivery device
1.9 Basic principle of iontophoresis.
1.10 Basic principle of phonophoresis
1.11 Basic design of microneedle delivery devices
1.12 Enhancement of transdermal permeation by pressure wave
1.13 various pharmaceutical carriers
1.14 Structure of typical vesicle
1.15 structure of conventional liposome
1.16 Chemical structure of Phosphatidylcholine (PC)
1.17 Structure of emulsomes
Proposed mechanism for penetration of molecule from
1.18 ethosomal system across the lipid domain of stratum
corneum
1.19 structure of sphingolipids
1.20 Structure of transferosomes
1.21 Mechanism of transferosomes permeation across the skin
1.22 Non-ionic surfactant vesicle / Niosome
Chapter -5
5.1 Absorption spectrum of Nystatin between 200-400nm
5.2 FTIR graph of Nystatin
5.3 Standard curve of nystatin in PBS pH 7.4
5.4 SEM images of the nystatin loaded niosomes
In vitro drug release from niosomal preparations
5.5
prepared using span-60
In vitro drug release from niosomal preparations
5.6
prepared using span-80
5.7 Release kinetics of niosomal suspension FN1
5.8 Release kinetics of niosomal suspension FN2
5.9 Release kinetics of niosomal suspension FN3
5.10 Release kinetics of niosomal suspension FN4
5.11 Release kinetics of niosomal suspension FN5
5.12 Release kinetics of niosomal suspension FN6
5.13 Release kinetics of niosomal suspension FN7
5.14 Release kinetics of niosomal suspension FN8
5.15 SEM Photographs of ethosomes
Effect of ethanol concentration on drug release from
5.16
ethosomes
Effect of lecithin concentration on drug release from
5.17
ethosomes.
5.18 Effect of cholesterol on drug release from ethosomes
5.19 Release kinetics of ethosomal formulation FE1
5.20 Release kinetics of ethosomal formulation FE2
5.21 Release kinetics of ethosomal formulation FE3
5.22 Release kinetics of ethosomal formulation FE4
5.23 Release kinetics of ethosomal formulation FE5
5.24 Release kinetics of ethosomal formulation FE6
5.25 Release kinetics of ethosomal formulation FE7
5.26 Release kinetics of ethosomal formulation FE8
5.27 SEM photographs of transferosomes
In vitro drug release from transferosomal formulations
5.28
prepared using span - 60
n vitro drug release from transferosomal formulations
5.29
prepared using span - 80
5.30 Release kinetics of transferosomal formulation FT1
5.31 Release kinetics of transferosomal formulation FT2
5.32 Release kinetics of transferosomal formulation FT3
5.33 Release kinetics of transferosomal formulation FT4
5.34 Release kinetics of transferosomal formulation FT5
5.35 Release kinetics of transferosomal formulation FT6
5.36 Release kinetics of transferosomal formulation FT7
5.37 Release kinetics of transferosomal formulation FT8
Zeta potential measurement of nystatin loaded niosomes
5.38
of an optimized formulation (FN3)
Zeta potential measurement of nystatin loaded ethosomes
5.39
of an optimized formulation (FE6)
Zeta potential measurement of nystatin loaded
5.40 transferosomes
of an optimized formulation (FT7)
In vitro drug release from all gels across the dialysis
5.41
membrane
5.42 Ex vivo drug release from all gels across the rat skin
calculation of flux at steady state and other parameters from
5.43
ex vivo drug release data
5.44 Calibration curve of nystatin by HPLC
5.45 In vivo drug release from all gels
5.46 HPLC chromatogram of nystatin (300ng/ml)
5.47 HPLC chromatogram of nystatin (600ng/ml)
5.48 HPLC chromatogram of nystatin (900ng/ml)
5.49 HPLC chromatogram of nystatin (1200ng/ml)
5.50 HPLC chromatogram of nystatin (1500ng/ml)
5.51 HPLC chromatogram of nystatin (1800ng/ml)
5.52 HPLC chromatogram of nystatin (2100ng/ml)
Conventional gel HPLC chromatograms of 0th hour blood
5.53
sample in Rabbit -1
Conventional gel HPLC chromatograms of 0th hour blood
5.54
sample in Rabbit -2
Conventional gel HPLC chromatograms of 1st hour blood
5.55
sample in Rabbit -1
Conventional gel HPLC chromatograms of1st hour blood
5.56
sample in Rabbit -2
Conventional gel HPLC chromatograms of 3rd hour blood
5.57
sample in Rabbit -1
Conventional gel HPLC chromatograms of 3rd hour blood
5.58
sample in Rabbit -2
Conventional gel HPLC chromatograms of 5th hour blood
5.59
sample in Rabbit -1
Conventional gel HPLC chromatograms of 5th hour blood
5.60
sample in Rabbit -2
Conventional gel HPLC chromatograms of 7th hour blood
5.61
sample in Rabbit -1
Conventional gel HPLC chromatograms of 7th hour blood
5.62
sample in Rabbit -2
Conventional gel HPLC chromatograms of 9th hour blood
5.63
sample in Rabbit -1
Conventional gel HPLC chromatograms of 9th hour blood
5.64
sample in Rabbit -2
Conventional gel HPLC chromatograms of 11th hour blood
5.65
sample in Rabbit -1
Conventional gel HPLC chromatograms of 11th hour blood
5.66
sample in Rabbit -2
Conventional gel HPLC chromatograms of 24th hour blood
5.67
sample in Rabbit -1
Conventional gel HPLC chromatograms of 24th hour blood
5.68
sample in Rabbit -2
Niosomal gel HPLC chromatograms of 0th hour blood sample
5.69
in Rabbit -1
Niosomal gel HPLC chromatograms of 0th hour blood sample
5.70
in Rabbit -2
Niosomal gel HPLC chromatograms of 1st hour blood sample
5.71
in Rabbit -1
Niosomalgel HPLC chromatograms of1st hour blood sample in
5.72
Rabbit -2
Niosomal gel HPLC chromatograms of 3rd hour blood sample
5.73
in Rabbit -1
Niosomalgel HPLC chromatograms of 3rd hour blood sample
5.74
in Rabbit -2
Niosomalgel HPLC chromatograms of 5th hour blood sample in
5.75
Rabbit -1
Niosomal gel HPLC chromatograms of 5th hour blood sample
5.76
in Rabbit -2
Niosomal gel HPLC chromatograms of 7th hour blood sample
5.77
in Rabbit -1
Niosomal gel HPLC chromatograms of 7th hour blood sample
5.78
in Rabbit -2
Niosomal gel HPLC chromatograms of 9th hour blood sample
5.79
in Rabbit -1
Niosomal gel HPLC chromatograms of 9th hour blood sample
5.80
in Rabbit -2
Niosomal gel HPLC chromatograms of 11th hour blood sample
5.81
in Rabbit -1
Niosomalgel HPLC chromatograms of 11th hour blood sample
5.82
in Rabbit -2
Niosomalgel HPLC chromatograms of 24th hour blood sample
5.83
in Rabbit -1
Niosomal gel HPLC chromatograms of 24th hour blood sample
5.84
in Rabbit -2
Ethosomal gel HPLC chromatograms of 0th hour blood sample
5.85
in Rabbit -1
Ethosomall gel HPLC chromatograms of 0th hour blood sample
5.86
in Rabbit -2
Ethosomal gel HPLC chromatograms of 1st hour blood sample
5.87
in Rabbit -1
Ethosomal gel HPLC chromatograms of1st hour blood sample
5.88
in Rabbit -2
Ethosomal gel HPLC chromatograms of 3rd hour blood
5.89
sample in Rabbit -1
Ethosomal gel HPLC chromatograms of 3rd hour blood
5.90
sample in Rabbit -2
Ethosomal gel HPLC chromatograms of 5th hour blood sample
5.91
in Rabbit -1
Ethosomal gel HPLC chromatograms of 5th hour blood sample
5.92
in Rabbit -2
Ethosomal gel HPLC chromatograms of 7th hour blood sample
5.93
in Rabbit -1
Ethosomal gel HPLC chromatograms of 7th hour blood sample
5.94
in Rabbit -2
Ethosomal gel HPLC chromatograms of 9th hour blood sample
5.95
in Rabbit -1
Ethosomalgel HPLC chromatograms of 9th hour blood sample
5.96
in Rabbit -2
Ethosomal gel HPLC chromatograms of 11th hour blood
5.97
sample in Rabbit -1
Ethosomal gel HPLC chromatograms of 11th hour blood
5.98
sample in Rabbit -2
Ethosomal gel HPLC chromatograms of 24th hour blood
5.99
sample in Rabbit -1
Ethosomal gel HPLC chromatograms of 24th hour blood
5.100
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 0th hour blood
5.101
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of 0th hour blood
5.102
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 1st hour blood
5.103
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of1st hour blood
5.104
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 3rd hour blood
5.105
sample in Rabbit -1
Transferosomal HPLC chromatograms of 3rd hour blood
5.106
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 5th hour blood
5.107
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of 5th hour blood
5.108
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 7th hour blood
5.109
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of 7th hour blood
5.110
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 9th hour blood
5.111
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of 9th hour blood
5.112
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 11th hour blood
5.113
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of 11th hour blood
5.114
sample in Rabbit -2
Transferosomal gel HPLC chromatograms of 24th hour blood
5.115
sample in Rabbit -1
Transferosomal gel HPLC chromatograms of 24th hour blood
5.116
sample in Rabbit -2

LIST OF ABBREVIATIONS

µg microgram

µm micrometer

nm nanometer

ng nanogram

0
C degree centigrade.

hrs hours
min minutes

ml milliliter

v/v volume by volume

w/w weight by weight

w/v weight by volume

v/w olume by weight

% percentage

FN1-FN8 Nystatin niosomal suspensions

FE1-FE8 Nystatin ethosomal suspensions

FT1-FT8 Nystatin transferosomal suspensions

UV ultra violet

LUVs large unilamellar vesicles

SUVs small unilamellar vesicles.

MLVs multi lamellar vesicles

SEM Scanning electron microscope

HPLC High pressure liquid chromatography

n Release exponent
CPCSEA Committee for the purpose of supervision of

experiments on animals

ICH International conference on harmonization

SD Standard deviation

LIST OF APPENDICES

Appendix - 1 Publication - 1

Appendix - 2 Publication - 2

Appendix - 3 Publication - 3
Appendix - 4 Conference - 1

Appendix - 5 Conference - 2

Appendix - 6 Approval Letter from CPCSEA


Chapter - 1

INTRODUCTION

Chapter - 1: Introduction

Table of contents

S.No Name of the subtitle Page No


1.1 Advantages of Transdermal drug delivery system  
1.2 Disadvantages of Transdermal drug delivery system  
1.3 Physiology of the skin  
1.4 Pathways of drug absorption through the skin  
General method of drug absorption from skin
1.5 to produce systemic effect  
1.6 Vesicular Carrier Systems  

1. INTRODUCTION

Route of administration is also one of the important influential factors, to

be considered, to produce desired therapeutic effect and patient compliance.

Different routes are available to administer the drugs which includes IM, IV,

SC, Nasal, Oral, transdermal, topical, rectal, etc.


Figure 1.1: Different routes of drug administration

Though different routes are available, the Oral route is an ancient, highly

successful, comfortable and economical route for administration of drugs. Its

success rate is limited by different reasons includes first pas metabolism,

gastric irritation, nausea, vomiting, unpleasant taste and odour of some drugs,

etc. Third highly successful route of administration is transdermal route from

ancient days. Even in current days, the analysis of the global market scenario

of the drug delivery market revealing the same1.


Figure 1.2: Market segmentation the global drug delivery market in 2007

Transdermal drug delivery systems were defined as self–contained,

discrete dosage forms which when applied to the intact skin, deliver the drug

(s) through the skin at controlled rate to the systemic circulation 2.

There is much difference between the transdermal and topical drug

delivery systems. In transdermal drug delivery systems the drug reaches to the

deeper layers of the skin and from there into the systemic circulation and

produces the systemic effect. The concentration of the drug in the plasma is

significant.

Eg. NitroDur transdermal patch, Fentanyl transdermal patch.

Topical drug delivery can be defined as the application of a drug

containing formulation to the skin to directly treat cutaneous disorders with

the intent of containing the pharmacological or other effect of the drug to the

surface of skin or within3.


In topical drug delivery systems, the drug will not reach the deeper layers

of the skin but remains upper layers of the skin and produces the localized

effect. Concentration of the drug in plasma is insignificant.

Eg. Lidocain patch.

Figure 1.3: Topical and transdermal drug delivery systems

The process of administration of drugs to the skin is using over the

millennia. They used to administer the drugs to produce localized effect and

the medication is used to administer in the form of ointments, gels, creams,

pates and poultices, etc. these dosage formsare incapable to produce systemic

effect. Administration of drugs through skin to produce systemic effect is a

relatively new phenomenon and is introduced to overcome the drawbacks

associated with oral administration and IV routes 4.

The first adhesive transdermal delivery system (TDDS) patch was

approved by the Food and Drug Administration in 1979 (scopolamine patch for

motion sickness). Nitroglycerine patches were approved in 1981. This method


of delivery became widely recognized when nicotine patches for smoking

cessation were introduced in 1991. Transdermal drug delivery systems growth

between the 2003-2007 was more than tripled. Today, there are 19

transdermal delivery systems available in the market and the drug is

administering by different techniques such as usage of the permeation

enhancers, iontophoresis, etc5.

Figure 1.4: Number of Transdermal drugs approved in each year.


Table 1.1: Transdermal drugs approved by the US FDA.

Approval
Drug Indication Product Name Marketing company
year

Novartis Consumer
Transderm-
1979 Scopolamine Motion sickness Health (Parsippany,
Scop
NJ)

Transderm- Novartis (East


1981 Nitroglycerin Angina pectoris
Nitro Hannover, NJ)

Boehringer Ingelheim
1984 Clonidine Hypertension Catapres-TTS
(Ridgefield, CT)

Menopausal Novartis (East


1986 Estradiol Estraderm
symptoms Hannover, NJ)

Janssen

1990 Fentanyl Chronic pain Duragesic Pharmaceutica

(Titusville, NJ)

GlaxoSmithKline

Nicoderm, (Philadelphia, PA),

1991 Nicotine Smoking cessation Habitrol, Novartis Consumer

ProStep Health (Parsippany,

NJ) Elan (Gainesville)

Testosterone Alza, Mountain View,


1993 Testosterone Testoderm
deficiency CA
Approval
Drug Indication Product Name Marketing company
year

Lidocaine/
Local dermal Iomed (Salt Lake
1995 epinephrine Iontocaine
analgesia City, UT)
(iontophoresis)

Estradiol/ Menopausal Novartis (East


1998 Combipatch
norethidrone symptoms Hannover, NJ)

Endo
Post-herpetic
1999 Lidocaine Lidoderm Pharmaceuticals
neuralgia pain
(Chadds Ford, PA)

Ortho-McNeil
Ethinylestradiol/
2001 Contraception Ortho Evra Pharmaceutical
norelgestromin
(Raritan, NJ)

Bayer Healthcare
Estradiol/ Menopausal
2003 Climara Pro Pharmaceuticals
levonorgestrel symptoms
(Wayne, NJ)

2003 Oxybutynin Overactive bladder Oxytrol Watson Pharma

Lidocaine Local dermal Echo Therapeutics


2004 SonoPrep
(ultrasound) anesthesia (Franklin, MA)

Endo
Lidocaine/ Local dermal
2005 Synera Pharmaceuticals
tetracaine analgesia
(Chadds Ford, PA)
Approval
Drug Indication Product Name Marketing company
year

Fentanyl HCl Acute postoperative Alza, Mountain View,


2006 Ionsys
(iontophoresis) pain CA

Attention deficit

2006 Methylphenidate hyperactivity Daytrana Shire (Wayne, PA)

disorder

Major depressive Bristol-Myers Squibb


2006 Selegiline Emsam
disorder (Princeton, NJ)

Schwarz Pharma
2007 Rotigotine Parkinson’s disease Neupro
(Mequon, WI)

Novartis (East
2007 Rivastigmine Dementia Exelon
Hannover, NJ)

1.1. Advantages of Transdermal drug delivery system

 Avoidance of first pass metabolism of drugs.


 Transdermal medication delivers a steady infusion of a drug over a prolonged

period of time. Adverse effects or therapeutic failures frequently associated with

intermittent dosing can also be avoided.

 The simplified medication regimen leads to improved patient compliance and

reduced the side effects, inter and intra-patient variability.

 No interference with gastric and intestinal fluids.

 Maintains stable or constant and controlled blood levels for longer period of

time.

 Comparable characteristics with interavenous infusion.

 It increases the therapeutic value of many drugs via avoiding specific problems

associated with the drug like GI irritation, lower absorption, decomposition due

to ‘hepatic first pass’ effect.

 This route is suitable for the administration of drugs having very short half life,

narrow therapeutic window and poor oral availability.

 Improved patient compliance and comfort via non-invasive, painless and simple

application.

 Flexibility of terminating the drug administration by simply removing the patch

from the skin.

 Self administration is possible in these systems.

1.2. Disadvantages of Transdermal drug delivery system


 The possibility of local irritation may develop at the site of application. Many

problems like Erythema, itching, and local edema can be caused by the drug,

the adhesive, or other excipients in the patch formulation.

 Drugs having large molecular size make absorption difficult. So drug molecule

should ideally be below 800-1000 daltons.

 Many drugs with a hydrophilic structure having a low penetration through the

skin and slowly to be of therapeutic benefit. Drugs with a lipophillic character,

however, are better suited for transdermal delivery.

 The barrier function of the skin changes from one site to another on the same

person, from person to person and with age.

 Transdermal drug delivery system cannot achieve high drug levels in

blood/plasma.

1.3. Physiology of the skin

The skin6 also knows as the cutaneous membrane covers the external

surface of the body and is the largest organ of the body in the both surface

area and weight. In adults, the skin covers an area of about 2 square meters

(22 square feet) and weight 4.5-5 kg (10-11 lb), about 7% of total body weight.

It ranges in thickness from 0.5 mm on the eyelids to 4.0 mm on the heels. Over

most of the body it is 1-2 mm thick. Anatomically, the skin has many

histological layers but in general, it is described in terms of three major tissue

layers. Figure shows the structure of the skin.

The three layers of the skin are as follows.

1. The epidermis
2. The dermis and

3. The hypodermis.

The superficial, thinner portion, which is composed of epithelial tissue, is

the epidermis. The deeper, thicker connective tissue portion is the dermis.

While the epidermis is a vascular, the dermis is vascular. For this reason, if

you cut the epidermis there is no bleeding, but if you cut the dermis there is

bleeding.

Deep to the dermis, but not part of the skin, is the subcutaneous layer.

Also called the hypodermis, this layer consists of areolar and adipose tissues,

Fibers that extend from the dermis anchor the skin to underlying fascia, the

connective tissue around muscles and bones. The subcutaneous layer serves

as a storage depot for fat and contains large blood vessels that supply the skin.

This region (and sometimes the dermis) also contains nerve endings called

pacinian (lamellated) corpuscles that are sensitive to pressure.

1.3.1. Epidermis

The epidermis is composed of keratinized stratified squamous epithelium.

It contains four principal types of cells. They are

1. Keratinocytes

2. Melanocytes

3. Langerhans cells and

4. Merkel cells.

About 90% of epidermal cells are Keratinocytes, which are arranged in

four or five layers and produce the protein keratin. Recall from chapter four
that keratin is a tough, fibrous protein that helps protect the skin and

underlying tissues from abrasions, heat, microbes, and chemicals.

Keratinocytes also produce lamellar granules, which release a water-repellent

sealant that decreases water entry and loss and inhibits the entry of foreign

materials.

About 8% of the epidermal cells are melanocytes, which develop from the

ectoderm of a developing embryo and produce the pigment melanin. Their long,

slender projections extend between the keratinocytes and transfer

melaningranules to them. Melanin is a yellow-red or brown-black pigment that

contributes to skin color and absorbs damaging ultraviolet (UV) light. Once

inside keratinocytes, the melanin granules cluster to from a protective veil over

the nucleus, on the side towards the skin surface. In this way they shield the

nuclear DNA from damage by UV light. Although their melanin granules

effectively protect keratinocytes, melanocytes themselves are particularly

susceptible to damage by UV light.

Langerhans cells also called epidermal dendritic cells, arise from red

bone marrow and migrate to the epidermis ,where they constitute a small

fraction of the epidermal cells.they participate in immune responses mounted

against microbes that invade the skin ,and are easily damaged by UV light.

Their role in the immune response is to help other cells of the immune system

recognize an invading microbe and destroy it.

Merkel cells are the least numerous are the epidermal cells. They are

located in the deepest layer of the epidermis, where they contact the flattened
process of a sensory neuron, a structure called a merkel (tactile) disc .Merkel

cells and their associated merkel discs detect touch sensations.

Several distinct layers of keratinocytes in various stages of development form

the epidermis. In most regions of the body the epidermis has four strata or

layers –stratum basale, stratum spinosum, stratum granulosum and a thin

stratum corneum. This is called thin skin. Where exposure to friction is

greatest, such as in the fingertrips, palms and soles, the epidermis has five

layers - stratum basale, stratum spinosum, stratum granulosum, stratum

lucidem, and a thick stratum corneum. This is called thin skin.

1.3.1.1. Stratum basale

The deepest layer of the epidermis is the stratum basle composed of a

single row of cuboidal or columnar keratinocytes.Some cells in this layer are

stem cells that undergo cell division to continually produce new keratinocytes.

The nuclei of keratinocytes in the stratum basale are large, and their cytoplasm

contains many ribosomes,a small Golgi complex, a few mitochondria , and

some rough endo plasmic reticulum.the cytoskeleton within keratinocytes of

the stratum basale includes scatterd intermeadiare filaments, called keratin

intermediate filaments (tonofilaments) . The keratin intermediate filament

forms the tough protein keratin and its more superficial epidermal layers.

Keratin protects the deeper layers from injury. Keratin intermediate filaments

attach to desmosomes, which bind cells of the stratum basale to rach other

and to the cell of the adjacent stratum spinosum, and to hemidesmosomes,

which bind the keratinocytes to the basement membrane posisioned between


the epidermis and the dermis. Melanocytes and merkel cells with their

associated merkel discs are scatterd among the keratinocytes of the basal layer

.The stratum basale is also known as the stratum germinativum to indicate its

role in forming new cells.

1.3.1.2. Stratum spinosum

Superficial to the stratum basale there is stratum spinosum. This

stratum mainly consists of numerous keratinocytes arranged in 8-10 layers.

Cells in the more superficial layers become somewhat flattened.The

keratinocytes in the stratum spinosum, which are produced by the stem cells

in the basale layer, have the same organells as cells of the stratum basale and

some retained their ability to devide. The keratinocytes of these layers produce

coarser bundles of keratin in intermediate filaments than those of the basal

layer. All though they are rounded and larger in living tissue ,cells of the

stratum spinosum shrink and pull apart when prepared for microscopic

examination so that they appeare to be coverd with thornlike spins. At each

spinelike projection, bundles of keratin intermediate filaments insert in to

desmosomes, which tightly join the cells to one another. This arrangement

provides both strength and flexibility to the skin. Langerhans cells and

projections of melonocytes are also presents in the stratum spinosum.

1.3.1.3. Stratum granulosum


At about the middle of the epidermis, the stratum granulosum consists

of three to five layers of flattened keratinocytes that are undergoing apoptosis.

The nuclei and other organelles of these cells begin to degenerate as they move

further from their source of nutrition. Even though keratin intermediate

filaments or no longer being produced by these cells, they become more

apparent because the organelles in the cells are regressing. A distinctive future

of cells in this layer is the presence of darkly staining granules of a protein

called keratohyalin, which assembles keratin intermediate filaments into

keratin. Also present in the keratinocytes are membrane-enclosed lamellar

granules, which fuse with the plasma membrane and release a lipid-rich

secretion. This secretion is deposited in the spaces between cells of the stratum

granulosum, stratum lucidum, and stratum corneum. The lipid-rich secretion

act as a water –repellent sealant,retardingloss and entry of water and entry of

foreign materials. As their nuclei break down during apoptosis, the

keratinocytes of the stratum granulosum can do longer carry on vi-tal

metabolic reactions, and they die. Thus, the stratum granulosum marks the

transition between the deeper, metabolically active strata and the dead cells of

the more superficial strata.

1.3.1.4. Stratum Lucidum

The stratum lucidum is present only in the thick skin of areas such as

the fingertips, palms, and soles. It consists of four to six layers of flattened

clear, dead keratinocytes that contain large amount of keratin and thickened
plasma membranes. This probably provides an additional level of toughness in

this resign of thick skin.

1.3.1.5. Stratum Corneum

The stratum corneum consists on average of 25 to 30 layers of flsttened

dead keratinocytes, but can range in thickness from a few cells in thin skin to

50 or more cell layers in thick skin. The cells are extremely thin, flat, plasma

membrane –enclosed packages of keratin that no longer contain a nucleus or

any internal organelles. They are the final product of the differentiation process

of the keratinocytes. The cells within each layer overlap one another like the

scales on the skin of a snake. Neighboring layers of cells also form strong

connections with one another. The plasma membranes of adjacent cells are

arranged in complex, wavy folds that fit together like pieces of a jigsaw puzzle

to hold the layers together. In this outer stratum of the epidermis, cells are

continuously shed and replaced by cells from the deeper strata. Its multiple

layers from injury and microbial invasion. Constant exposure of skin to friction

stimulates increased cell production that results in the formation of a callus,

an abnormal thickening of the stratum corneum.

1.3.2. Dermis

The second, deeper part of the skin, the dermis, is composed of dense

irregular connective tissue containing collagen and elastic fibers. This woven

network of fibers has great tensile strength. The dermis also has the ability to

stretch and recoil easily. It is much thicker than the epidermis, and this

thickness varies from region to region in the body, reaching is greatest


thickness on the palms and soles. Leather, which we use for belts, shoes,

baseball gloves, and basketballs, is the dried and treated dermis of other

animals. The few cells present in the dermis include predominantly fibroblasts,

with some macrophages, and a few adipocytes near its boundary with the

subcutaneous layer. Blood vessels, nerves, glands, and hair follicles are

embedded in the dermal layer. The dermis is essential to the survival of the

epidermis, and these adjacent layers form many important structural and

functional relations. Based on its tissue structure, the dermis can be divided

into a thin superficial papillary region and a thick deeper reticular region.

The papillary region makes up one-fifth of the thickness of the total

layer. It consists of thin collagen and fine elastic fibers. Its surface area is

greatly increased by dermal papillae, small, nipple-shaped structure that

project into the undersurface of the epidermis. All dermal papillae contain

capillary loops. Some also contain tactile receptors called meissner corpuscles (

MIS-ner) or corpuscles of touch , nerve endings that are sensitive to touch. Still

other dermal papillae also contain free nerve endings, dendrites that lack any

apparent structural specialization. Different free nerve endings initiate signals

that give rise to sensations of warmth, coolness, pain, tickling, and itching.

The reticular region, which is attached to the subcutaneous layer,

consists of bundles of thick collagen fibers, scattered fibroblasts, and various

wandering cells. Some adipose cells can be present in the deepest part of the

layer, along with some coarse elastic fibers. The collagen fibers in the reticular

region are arranged in a netlike manner and have a more regular arrangement
than those in the papillary region. The more regular orientation of the large

collagen fibers helps the skin resist stretching. Blood vessels, nerves, hair

follicles, sebaceous (oil) glands, and sudoriferous (sweat) glands occupy the

spaces between fibers.

The combination of collagen and elastic fibers in the reticular region

provides the skin with strength, extensibility, the ability to stretch, and

elasticity, the ability to return to original shape after stretching. The

extensibility of skin can be readily seen around joints and in pregnancy and

obesity.

The surfaces of the palms, fingers, soles, and toes have a series of ridges

and grooves. They appear either as straight lines or as a pattern of loops and

whorls, as on the tips of the digits. These epidermal ridges are produced during

the third month of fetal development as downward projections of the epidermis

into the dermis between the dermal papillae of the papillary region. The

epidermal ridges create a strong bond between the epidermis and dermis in a

region of high mechanical stress. The epidermis ridges also increase the

surface area of the epidermis and thus increase the grip of the hand or foot by

increasing friction. Finally, the epidermal ridges greatly increase surface area,

which increase the number of meissner corpuscles and thus increases tactile

sensitivity. Because the ducts of sweat glands open on the tops of the

epidermal ridges as sweat pores, the sweat and ridges form fingerprints on

touching a smooth object. The epidermal ridges pattern is in part genetically

determined and is unique for each individual. Even identical twins have
different patterns. Normally, the ridge pattern does not change during life,

except to enlarge, and thus can serve as the basis for identification. The study

of the pattern of epidermal ridges is called dermatoglyphics.

In addition to forming epidermal ridges, the complex papillary surface of

the dermis has other functional properties. The dermal papillae greatly

increase the surface contact between the dermis and epidermis. This increased

dermal contact surface, with its extensive network of small blood vessels,

serves as an important source of nutrition for the overlying epidermis.

Molecules diffuse from the small blood capillaries in the dermal papillae to the

cells of the stratum basale, allowing the basal epithelial stem cells to divide and

the keratinocytes to grow and develop. As keratinocytes push toward the

surface and away from the dermal blood source, they are no longer able to

obtain the nutrition they require, leading to the eventual breakdown of their

organelles.

The dermal papillae fit together with the complementary epidermal ridge

to form an extremely strong junction between the two layers. This jigsaw

puzzle-like connection strengthens the skin against shearing forces that

attempt to separate the epidermis from the dermis.


Figure 1.5: Structure of the skin

1.4. Pathways of drug absorption through the skin

Several investigations were carried out to determine the penetration

pathways of the topically applied active principles into/through the skin to

produce both localized and/or systemic effects. The investigations proposed

different pathways through which drugs can be absorbed into and/or across

the skin depending on the physicochemical properties of the drug. Both

hydrophilic and lipophilic drugs are absorbed from different routes of the skin.

The barrier properties of the stratum corneum will not permeate drugs through
the skin but due to the presence of various absorption routes facilitates the

permeation of drugs and allows the drugs to reach the systemic circulation.

The different absorption routes proposed by the researchers are as follows7:

1. Transfollicular route

2. Transcellular route

3. Intercellular route

1.4.1. Transfollicular route

This route is the shortest pathway for drugs to reach the systemic

circulation that provides a large area for diffusion of drugs. Various oil glands,

hair follicles, sweat glands and pores openings present on the outer surface of

the skin via their ducts. These ducts acts continuous channels for

transportation of the drugs across the stratum corneum but extent of drug

transportation across the skin will be influenced by various factors like

secretions from the glands, amount of secretion and content etc. However

transappendageal route occupies only 0.1% of total skin surface and therefore

contributes minimum.

1.4.2. Transcellular route

In transcellular route drug transportation will takes place from the

corneocytes which consists of highly hydrated keratin. This corneocytes will

create hydrophilic pathways for transportaion of drugs. Around the corneocytes

there will be lipids which connect these cells. Therefore drugs require

partitioning and diffusion steps for permeation into the systemic circulation.

This is the widely using route for transportation of various drugs. In this route
the drugs will pass through the matrix (cytoplasm) of the cells and is suitable

route for hydrophilic drugs. The highly hydrated keratin provide aqueous

pathway to the hydrophilic drugs. A number of partitioning and diffusion steps

are require to pass the drug through the cell matrix.

1.4.3. Intercellular route

In intercellular route, the drugs diffuse through the lipid matrix present

between the cells. The barrier property of this route is due to the tortuous

structure formed by corneocytes and the drug has to pass through the

alternating lipid and aqueous domain by partitioning into the lipid bilayer and

diffusing to the inner side. It has been found that water has to travel 50 times

more by this route so; it is suitable mainly for uncharged lipophilic drugs.

Figure 1.6: Pathways of drug absorption through the skin


1.5. General method of drug absorption from skin to produce systemic

effect

To produce desired therapeutic effect, the drug concentration in the

plasma should lie in the therapeutic window. i.e. between minimum effective

concentration (MEC) and maximum safe concentration (MSC). All drugs of

desired concentration will not reach the therapeutic window when applied on

the skin by means of conventional dosage forms such as ointments, creams,

gels, etc because of various reasons like barrier properties of the skin, high

molecular weight, high melting point, less lipophilicity of the drug etc. To

overcome these difficulties and to achieve of desired concentration in the

plasma different researchers introduced different technologies. The different

technologies introduced by the researchers for administration of drugs across

the skin to produce systemic effect are as follows.

 Chemical potential adjustment

 Ion pairs and complex coacervates

 Eutectic systems

 Hydration

 Chemical penetration enhancers

 Electrophoresis

 Iontophoresis

 Ultrasound ( Phonophoresis, Sonophoresis)

 Laser radiation and photochemical waves

 Radio frequency
 Magnetophoresis

 Temperature (“thermophoresis”)

 Microneedle based devices

 Skin puncture and perforation

 Needleless injection

 Suction ablation

 Application of Pressure

 Skin stretching

 Skin abrasion

 Pharmaceutical carrier systems, etc

1.5.1. Chemical potential adjustment

The maximum skin penetration rate is obtained when a drug is at its

highest thermodynamic activity as is the case in a supersaturated. The

diffusion of paraben from saturated solutions in eleven different solvents

through a silicone membrane was determined. Due to the different solubility of

the parabens in the various solvents, the concentration varied over two orders

of magnitude. However, paraben flux was the same from all solvents, as the

thermodynamic activity remained constant because saturated conditions were

maintained throughout the experiment. Supersaturated solutions can occur

due to evaporation of solvent or by mixing of cosolvents. Clinically, the most

common mechanism is evaporation of solvent from the warm skin surface,

which probably occurs, in many topically applied formulations. In addition, if


water is imbibed from the skin into the vehicle and acts as an antisolvent, the

thermodynamic activity of the permeant would increase .


8-11

1.5.2. Ion pairs and complex coacervates:

Charged drug molecules do not readily partition into or permeate

through human skin. Formation of lipophilic ionpairs has been investigated to

increase stratum corneum penetration of charged species. This strategy

involves adding an oppositely charged species to the charged drug, forming an

ion-pair in which the charges are neutralised so that the complex can partition

into and permeate through the stratum corneum. The ion-pair then dissociates

in the aqueous viable epidermis releasing the parent charged drug, which can

diffuse within the epidermal and dermal tissues12.

1.5.3. Eutectic systems

The melting points of a drug influences solubility and hence skin

penetration. According to regular solution theory, the lower the melting point,

the greater the solubility of a material in a given solvent, including skin lipids.

The melting point of a drug delivery system can be lowered by formation of a

eutectic mixture: a mixture of two components which, at a certain ratio, inhibit

the crystalline process of each other, such that the melting point of the two

components in the mixture is less than that of each component alone. A

number of eutectic systems containing a penetration enhancer as the second

components have been reported, for example: Ibuprofen with terpenes 13, and

methyl nicotinate14, propranolol with fatty acids15 and lignocaine with

menthol16.
1.5.4. Hydration:

Water is the most widely used and safest method to increase skin

penetration of both hydrophilic17 and lipophilic permeants18. The water content

of the stratum corneum is around 15 to 20% of the dry weight but can vary

according to humidity of the external environment. Additional water within the

stratum corneum could alter permeant solubility and thereby modify

partitioning from the vehicle into the membrane. In addition, increased skin

hydration may swell and open the structure of the stratum corneum leading to

an increase in penetration, although this has yet to be demonstrated

experimentally. For example, Scheuplein and Blank showed that the diffusion

coefficients of alcohols in hydrated skin were ten times that observed in dry

skin19. Hydration can be increased by occlusion with plastic films; paraffins,

oils, waxes as components of ointments and water-in-oil emulsions that

prevent transepidermal water loss; and oil-in-water emulsions that donate

water. Of these, occlusive films of plastic or oily vehicle have the most profound

effect on hydration and penetration rate20. A commercial example of this is the

use of an occlusive dressing to enhance skin penetration of lignocaine and

prilocane from EMLA cream in order to provide sufficient local anaesthesia

within about 1 hour. Also drug delivery from many transdermal patches

benefits from occlusion.

1.5.5. Chemical penetration enhancers (CPEs)

The use of CPEs over the other techniques has certain advantages,

including design flexibility of the patch and ease of patch application over a
large area (>10 cm2)21. An ideal penetration enhancer should reversibly reduce

the barrier resistance of the SC without damaging the skin cells. According to

Finnin et al.22 : ideal penetration enhancers should possess the following

properties:

 Pharmacologically inert

 Nontoxic, nonirritating, and non-allergenic

 Rapid onset of action; predictable and suitable duration of action for the

drug used

 Reversible effect of the CPE on the barrier property of SC

 Chemically and physically compatible with the delivery system

 Readily incorporated into the delivery system

 Inexpensive and cosmetically acceptable

Because the skin provides such a formidable barrier to the delivery of most

drugs, a broad range of different chemical additives have been tested to

enhance transdermal penetration during the last two decades. Much of the

cited literature is found in patents 23 as well as pharmaceutical science

literature24. Even though many chemical entities have been identified, only a

few were introduced in the market due to several limitations, which include

their economic feasibility and the toxic effects on skin, which make them

undesirable for developing transdermal patches. Penetration enhancers may

act by one or more of three main mechanisms 25:

 Disruption of the highly ordered structure of stratum corneum lipid.

 Interaction with intercellular protein.


 Improved partition of the drug, coenhancer or solvent into the stratum

corneum.

1.5.5.1. Sulphoxides and similar chemicals

Dimethyl sulphoxides (DMSO) is one of the earliest and most widely

studied penetration enhancers. It is a powerful aportic solvent which hydrogen

bonds with itself rather than with water. It is colourless, odourless and is

hydroscopic and is often used in many areas of pharmaceutical sciences as a

“universal solvent”. DMSO alone has been applied topically to treat systemic

inflammation. DMSO works rapidly as a penetration enhancer - spillage of the

material onto the skin can be tasted in the mouth within a second. Although

DMSO is an excellent accelerant, it does create problems. The effect of the

enhancer is concentration-dependent and generally cosolvents containing >

60% DMSO are needed for optimum enhancement efficacy. However, at these

relative high concentrations, DMSO can cause erythema and wheal of the

stratum corneum. Denaturing of some skin proteins results in erythema,

scaling, contact uticaria, stinging and burning sensation 26. Since DMSO is

problematic for use as a penetration enhancer, researchers have investigated a

similar chemically-related material as a accelerant. Dimethylacetamide (DMAC)

and dimethylformamide (DMF) are similarly powerful aportic solvents.

However, Southwell and Barry, showing a 12-fold increase in the flux of

caffeine permeating across a DMF-treated human skin, concluded that the

enhancer caused irreversible membrane damage27. DMF irreversibly damages

human skin membranes but has been found in vivo to promote the
bioavailability of betamethasone-17-benzoate as measured by vasoconstrictor

assay28, 29
. DMSO may also extract lipids, making the horny layer more

permeable by forming aqueous channels.

It has been postulated that DMSO denatures the intercellular structural

proteins of the stratum corneum, or promotes lipid fluidity by disruption of the

ordered structure of the lipid chains. In addition, DMSO may alter the physical

structure of the skin by elution of lipid, lipoprotein and nucleoprotein

structures of the stratum corneum. Decylmethylsulfoxide (DCMS) is thought to

promote permeation enhancement as a result of protein-DCMS interaction

creating aqueous channels, in addition to lipid interactions.

1.5.5.2. Alkanes

Long chain alkanes (C7-C16) have been shown to enhance skin

permeability by non-destructive alteration of the stratum corneum barrier 30.

These findings were confirmed in studies in which nonane was investigated as

an enhancer31, although there must be some destructive solubilisation and

biochemical extraction caused by these lipophilic solvents.

1.5.5.3. Azone

Azone (1-dodecylazacycloheptan-2-one or laurocapran) was the first

molecule specifically designed as a skin penetration enhancer. Azone is a

colourless, odourless liquid with a melting point of -7 ºC and it possesses a

smooth, oily but yet non-greasy feel. Azone is a highly lipophilic material with a

log p octanol / water of around 6.2 and it is soluble in and compatible with

most organic solvents including alcohol and propylene glycol. Azone enhances
the skin transport of a wide variety of drugs including steroids, antibiotics and

antiviral agents. Azone is most effective at low concentrations, being employed

typically between 0.1- 5% but more often between 1- 3% 32. Azone partitions

into a bilayer lipid to disrupt their packing arrangement but integration into

the lipid is unlikely to be homogeneous. Azone provokes dynamic structural

disorder of the intercellular lamellar lipid structure throughout the stratum

corneum and the creation of fluid domains involving the intercellular lipids,

which was suggested by 2H NMR assay. Another mechanism was also

proposed based on the alteration of the lateral bonding within stratum corneum

lipid lamellae33. Azone/PG increase penetration through the stratum corneum

by affecting both the hydrophilic and lipophilic routes of penetration. Azone

increases the fluidity of the lipid layer, while PG increases the water content of

the proteinaceous region and helps azone partition into the aqueous region. A

combination of these two helps the penetration of hydrophilic drugs greatly 34.

1.5.5.4. Pyrrolidones

Pyrrolidones have been used as permeation enhancers for numerous

molecules including hydrophilic (e.g. mannitol and 5-flurouracil) and lipophilic

(progesterone and hydrocortisone) permeants. N-methyl-2- pyrolidone was

employed with limited success as a penetration enhancer for captopril when

formulated in a matrix-type transdermal patch 35. The pyrrolidones partition

well into human stratum corneum within the tissue and they may act by

altering the solvent nature of the membrane. Pyrrolidones have been used to

generate reservoirs within the skin membrane. Such a reservoir effect offers a
potential for sustained release of a permeant from the stratum corneum over

extended time periods36.

1.5.5.5. Urea

Urea promotes transdermal permeation by facilitating hydration of the

stratum corneum and by the formation of hydrophilic diffusion channels within

the barrier. As urea itself possesses only marginal penetration enhancing

activity, attempts have been made to synthesis analogues containing more

potent enhancing moieties. Thus Wong and co-workers synthesised cyclic urea

analogues and found them to be as potent as Azone for promoting

indomethacin across shed snake skin and hairless mouse skin Cyclic urea

permeation enhancers are biodegradable and non-toxic molecules consisting of

a polar parent moiety and a long chain alkyl ester group. As a result,

enhancement mechanism may be a consequence of both hydrophilic activity

and lipid disruption mechanism37.

1.5.5.6. Fatty acids and Esters

Percutaneous drug absorption has been increased by a wide variety of

fatty acids and their esters, the most popular of which is oleic acid. A general

trend has been seen that unsaturated fatty acids are more effective in

enhancing percutaneous absorption of drugs than their saturated

counterparts. It is of interest to note that many penetration enhancers such as

azone contain saturated or unsaturated hydrocarbon chains and some

structure - activity relationships have been drawn from the extensive studies of

Aungst who employed a range of fatty acids, acids, alcohols, sulphoxides,


surfactants and amides as enhancers for naloxone 38, 39
. Shin et al40 studied

various penetration enhancers like glycols (diethylene glycol and tetraethylene

glycol), fatty acids (lauric acid, myristic acid and capric acid) and nonic

surfactant (polyoxyethylene-2-oleyl ether, polyoxy ethylene-2-stearly ether) on

the release of triprolidone. Lauric acid in Propylene glycol enhanced the

delivery of highly lipophilic antiestrogen 41. Oleic acid greatly increased the flux

of many drugs such as increasing the flux of salicylic acid 28-fold and 5-

flurouracil flux 56-fold through human skin membrane in vitro42. The enhancer

interacts with and modifies the lipid domains of the stratum corneum as would

be expected for a long chain fatty acid with cis- configuration.

1.5.5.7. Alcohols, fatty alcohols and glycols

Alcohols may influence transdermal penetration by a number of

mechanisms. The alkyl chain length of the alkanols (fatty alcohols) is an

important parameter in the promotion of permeation enhancement.

Augmentation appears to increase as the number of carbon units increases, up

to a limiting value43. In addition, lower molecular weight alkanols are thought

to act as solvents, enhancing the solubility of drugs in the matrix of the

stratum corneum. Disruption of the stratum corneum integrity through

extraction of biochemicals by the more hydrophobic alcohols almost certainly

also contributes to enhanced mass transfer through this tissue 44. Ethanol is

the most commonly used alcohol as a transdermal penetration enhancer.

Ethanol acts as a penetration enhancer by extracting large amounts of stratum

corneum lipids. It also increases the number of free sulphydryl groups of


keratin in the stratum corneum proteins. Usually, pretreatment of skin with

ethanol increases the permeation of hydrophilic compounds, while it decreases

that of hydrophobic ones45. The molecular complexity of different glycol

molecules is a determinant of their efficacy as permeation enhancers. Solubility

of the drug in the delivery vehicle is markedly influenced by the number of

ethylene oxide functional groups on the enhancer molecule; this solubility

modification may either enhance or retard transdermal flux depending on the

specific drug and delivery environment. The activity of propylene glycol (PG) is

thought to result from solvation of α keratin within the stratum corneum; the

occupation of proteinaceous hydrogen bonding sites reducing drug-tissue

binding and thus promoting permeation46. PG is widely used as a vehicle for

penetration enhancers and shows synergistic action when used with, for

example, oleic acid.

1.5.5.8. Surfactants

Many surfactants are capable of interacting with the stratum corneum to

increase the absorption of drugs and other active compounds from products

applied to the skin. Skin penetration measurements are valuable in quantifying

these effects and observing the influence of surfactant chemistry and

concentration. A surfactant interacts with skin by depositing onto the stratum

corneum, thereby disorganizing its structure. Then surfactant can solubilise or

remove lipids or water-soluble constituents in or on the surface of the stratum

corneum. Finally it can be transported into and through the stratum corneum.

This last effect is related to the surfactant and stratum corneum protein
interaction and epidermal keratin denaturation 47. In general, anionic

surfactants are more effective than cationic and nonionic surfactants in

enhancing skin penetration of target molecules. Some anionic surfactants

interact strongly with both keratin and lipids, whereas the cationic surfactants

interact with the keratin fibrils of the cornified cells and result in a disrupted

cell-lipid matrix. Nonionic surfactants enhance absorption by inducing

fluidization of the stratum corneum lipids.Scheuplein and Ross reported that

the capacity of the statum corneum to retain significant quantities of

membrane-bound water is reduced in the presence of sodium dodecanoate and

sodium dodecyl sulfate48. This effect is readily reversible upon removal of the

agents. These investigations proposed that anionic surfactants alter the

permeability of the skin by acting on the helical filaments of the stratum

corneum, thereby resulting in the uncoiling and extension of _-keratin

filaments to produce _- keratin. Then they cause an expansion of the

membrane, which increases permeability. However, more recent findings

suggest that impairment of the skin’s barrier properties is unlikely to result

from changes in protein conformation alone. Based on differential scanning

calorimetry results, sodium lauryl sulfate (SLS) disrupted both the lipid and

the protein components. The amount of surfactant that penetrates the skin

after the disruption of the skin barrier depends on the monomer activity and

the critical micelle concentration (CMC). Above the CMC, the added surfactant

exists as micelles in the solution and micelles are too large to penetrate the

skin. The extent of barrier disruption and penetration enhancement of a


surfactant is also strongly dependent on surfactant structure, especially alkyl

chain length. In general, studies have shown that surfactants having 12

carbons in their alkyl chain cause more disruption to the skin barrier and

allow drugs to penetrate more readily than those that have more or less than

12 carbons. The explanation for this optimum of 12 carbons is not known yet.

1.5.5.9. Essential oil, terpenes and terpenoids

Terpenes are found in essential oils, and are compounds comprising of

only carbon, hydrogen and oxygen atoms, but which are not aromatic.

Numerous terpenes have long been used as medicines as well as flavoring and

fragrance agents. The essential oils of eucalyptus, chenopodium and ylang-

ylang have been found to be effective penetration enhancers for 5-flouorouracil

transversing human skin in vivo49. Cornwell et al50 investigated the effect of 12

sesquiterpenes on the permeation of 5-flurouracil in human skin. Pretreatment

of epidermal membranes with sesquiterpene oil or using solid sesquiterpenes

saturated in dimethyl isosorbide increased the absorption of 5- flurouracil. L-

menthol has been used to facilitate in vitro permeation of morphine

hydrochloride through hairless rat skin as well as diffusion of imipramine

hydrochloride across rat skin and hydrocortisone through hairless mouse

skin51. One mechanism by which this agent operates is to modify the solvent

nature of the stratum corneum, thus improving drug partitioning into the

tissue. Many terpenes permeate human skin well and large amounts of terpene

have been found in the epidermis after application from a matrix-type patch.

Terpenes may also modify drug diffusivity through the membrane. During
steady state permeation experiments using terpenes as penetration enhancers,

the lag time for permeation was usually reduced, indicating some increase in

drug diffusivity through the membrane following terpene treatment.

1.5.5.10. Cyclodextrins

Cyclodextrins are biocompatible substances that can form inclusion

complexes with lipophilic drugs with a resultant increase in their solubility,

particularly in aqueous solutions. However, cyclodextrins alone were

determined be to less effective as penetration enhancers than when combined

with fatty acids and propylene glycol52.

1.5.5.11. Oxazolidinones

Oxazolidinones are a new class of chemical agents which have the

potential for use in many cosmetic and personal care product formulations.

This is due to their ability to localize co-administered drug in skin layers,

resulting in low systemic permeation. The structural features of these

permeation enhancers are closely related to sphingosine and ceramide lipids

which are naturally found in the upper skin layers. Oxazolidinones such as 4-

decyloxazolidin-2-one has been reported to localize the delivery of many active

ingredients such as retinoic acid and diclofenac sodium in skin layers. This

compound has a higher molecular weight and lipophilicity than other solvent-

type enhancers, physical characteristics that may be beneficial in terms of a

reduction in local toxicity because of the lack of effective absorption of these

enhancers into the lower skin layers where irritation is likely to be occur.
Figure 1.7: Chemical structure of typical chemical penetration enhancers

1.5.6. Electroporation

The use of electropermeabilization, as a method of enhancing diffusion

across biological barriers, dates back as far as 100 years 53. Electroporation

involves the application of high voltage pulses to induce skin perturbation.

High voltages (≥100 V) and short treatment durations (milliseconds) are most

frequently employed. Other electrical parameters that affect delivery include

pulse properties such as waveform, rate and number. The increase in skin

permeability is suggested to be caused by the generation of transient pores

during electroporation. The technology has been successfully used to enhance

the skin permeability of molecules with differing lipophilicity and size (i.e. small

molecules, proteins, peptides and oligonucleotides) including

biopharmaceuticals with a molecular weight greater that 7kDA, the current

limit for iontophoresis. Genetronics Inc (San Diego, California) have developed
a prototype electroporation transdermal device, which has been tested with

various compounds with a view to achieving gene delivery, improving drug

delivery and aiding the application of cosmetics. Other transdermal devices

based on electroporation have been proposed by various groups however, more

clinical information on the safety and efficacy of the technique is required to

assess the future commercial prospects.

Figure 1.8: Basic design of electroporation drug delivery device

1.5.7. Iontophoresis

This method involves enhancing the permeation of a topically applied

therapeutic agent by the application of a low level electric current either

directly to the skin or indirectly via the dosage form 54. Increase in drug

permeation as a result of this methodology can be attributed to either one or a

combination of the following mechanisms; electrorepulsion (for charged

solutes), electro osmosis (for uncharged solutes) and electropertubation (for

both charged and uncharged). Parameters that affect design of an iontophoretic


skin delivery system include; electrode type, current intensity, pH of the

system, competitive ion effect and permeant type. The launch of

commercialised systems of this technology has either occurred or is currently

under investigation by various companies. Extensive literature exists on the

many types of drugs investigated using iontophoretic delivery and the reader is

referred to the following extensive reviews. The PhoresorTM device (Iomed Inc.),

was the first iontophoretic system to be approved by the FDA in the late 70’s as

a physical medicine therapeutic device. In order to enhance patient compliance

the use of patient- friendly, portable and efficient iontophoretic systems have

been under intense development over the years. Such improved systems

include the Vyteris and E-TRANS iontophoretic devices. Previous work has also

reported that the combined use of iontophoresis and electroporation is much

more effective than either technique used alone in the delivery of molecules

across the skin. The limitations of ionotophoretic systems include the

regulatory limits on the amount of current that can be used in humans

(currently set at 0.5 mA cm-2) and the irreversible damage such currents could

do to the barrier properties of the skin. In addition, iontophoresis has failed to

significantly improve the transdermal delivery of macromolecules of >7000

Da55.
Figure 1.9: Basic principle of iontophoresis. A current passed between the
active electrode and the indifferent electrode repelling drug away from
the active electrode and into the skin.

1.5.8. Ultrasound (sonophoresis and phonophoresis)

Ultrasound involves the use of ultrasonic energy to enhance the

transdermal delivery of solutes either simultaneously or via pre-treatment and

is frequently referred to as sonophoresis or phonophoresis. The proposed

mechanism behind the increase in skin permeability is attributed to the

formation of gaseous cavities within the intercellular lipids on exposure to

ultrasound resulting in disruption of the SC 56. Ultrasound parameters such as

treatment duration, intensity and frequency are all known to affect

percutaneous absorption, with the latter being the most important. Although

frequencies between 20 kHz-16 MHz have been reported to enhance skin

permeation, frequencies at the lower end of this range (< 100 kHz) are believed

to have a more significant effect on transdermal drug delivery with the delivery

of macromolecules of molecular weight up to 48 kDa being reported. The


SonoPrep ® device (Sontra Medical Corporation) uses low frequency ultrasound

(55 kHz) for an average duration of 15 s to enhance skin permeability. This

battery operated hand held device consists of a control unit, ultrasonic horn

with control panel a disposable coupling medium cartridge, and a return

electrode. The ability of the SonoPrep® device to reduce the time of onset of

action associated with the dermal delivery of local anaesthetic from EMLA

cream was recently reported. In the study by Kost et al.(37), skin treatment by

ultrasound for an average time of 9 s resulted in the attainment of dermal

anaesthesia within 5 min, which was comparable to the 60 min required in for

non-treated skin. The use of other small, lightweight novel ultrasound

transducers to enhance the in vitro skin transport of insulin has also been

reported by a range of workers57.

Figure 1.10: Basic principle of phonophoresis. Ultrasound pulses are


passed through the probe into the skin fluidizing the lipid bilayer by the
formation of bubbles caused by cavitation.
1.5.9. Laser radiation and photomechanical waves

Lasers have been used in the clinical therapies for decades, therefore

their effects on biological membranes are well documented. Lasers are

frequently used for the treatment of dermatological conditions such as acne

and to confer ‘facial rejuvenation’ where the laser radiation destroys the target

cells over a short frame of time (~300 ns). Such direct and controlled exposure

of the skin to laser radiation results in ablation of the SC without significant

damage to the underlying epidermis. Removal of the SC via this method has

been shown to enhance the delivery of lipophilic and hydrophilic drugs. The

extent of barrier disruption by laser radiation is known to be controlled by

parameters such wavelength, pulse length, pulse energy, pulse number and

pulse repetition rate. A hand-held portable laser device has been developed by

Norwood Abbey Ltd (Victoria, Australia). In a study involving human

volunteers, the Norwood Abbey laser device was found to reduce the onset of

action of lidocaine to 3-5 min, whilst 60 min was required to attain a similar

effect in the control group. The Norwood Abbey system has been approved by

the US and Australian regulatory bodies for the administration of a topically

applied anaesthetic. Pressure waves (PW), which can be generated by intense

laser radiation, without incurring direct ablative effects on the skin have also

been recently found to increase the permeability of the skin. It is thought that

PW form a continuous or hydrophilic pathway across the skin due to expansion

of the lacunae domains in the SC. Important parameters affecting delivery such

as peak pressure, rise time and duration has been demonstrated. The use of
PW may also serve as a means of avoiding problems associated with direct

laser radiation. Permeants that have been successfully delivered in vivo include

insulin58, 40 kDa dextran and 20 nm latex particles. A design concept for a

transdermal drug delivery patch based on the use of PW has been proposed by

Doukas & Kollias.

1.5.10. Radio-frequency

Radio-frequency involves the exposure of skin to high frequency

alternating current (~ 100 kHz) resulting in the formation of heat-induced

microchannels in the membrane similar to when laser radiation is employed.

The rate of drug delivery is controlled by the number and depth of the

microchannels formed by the device, which is dependent on the properties of

the microelectrodes used in the device. The Viaderm device (Transpharma Ltd)

is a hand held electronic device consisting of a microprojection array (100

microelectrodes/cm2) and a drug patch. The microneedle array is attached to

the electronic device and placed in contact with the skin to facilitate the

formation of the microchannels. Treatment duration takes less than a second,

with a feedback mechanism incorporated within the electronic control

providing a signal when the microchannels have been created, so as to ensure

reproducibility of action. The drug patch is then placed on the treated area.

Experiments in rats have shown the device to enhance the delivery of

granisetron HCL, with blood plasma levels recorded after 12 h rising to 30

times higher levels than that recorded for untreated skin after 24 h. A similar

enhancement in diclofenac skin permeation was also observed in the same


study. The device is reported not to cause any damage to skin with the radio-

frequency-induced microchannels remaining open for less than 24 h. The skin

delivery of drugs such as testosterone and human growth hormone by this

device is also currently in progress.

1.5.11. Magnetophoresis

This method involves the application of a magnetic field which acts as an

external driving force to enhance the diffusion of a diamagnetic solute across

the skin. Skin exposure to a magnetic field might also induce structural

alterations that could contribute to an increase in permeability. In vitro studies

by Murthy showed a magnetically induced enhancement in benzoic acid flux,

which was observed to increase with the strength of the applied magnetic field.

Other in vitro studies using a magnet attached to transdermal patches

containing terbutaline sulphate (TS), demonstrated an enhancement in

permeant flux which was comparable to that attained when 4% isopropyl

myristate was used as a chemical enhancer. In the same paper the effect of

magnetophoresis on the permeation of TS was investigated in vivo using guinea

pigs. The preconvulsive time (PCT) of guinea pigs for those subjected to

magnetophoretic treatment was found to last for 36 h which was similar to that

observed after application of a patch containing 4% IPM. This was in contrast

to the response elicited by the control (patch without enhancer), when the

increase in PCT was observed for only 12 h. In human subjects, the level of TS

in the blood was higher but, not significantly different to that observed with the

patch containing 4% IPM. The fact that this technique can only be used with
diamagnetic materials will serve as a limiting factor in its applicability and

probably explains the relative lack of interest in the method.

1.5.12. Temperature (“thermophoresis”)

The skin surface temperature is usually maintained at 32ºC in humans

by a range of homeostatic controls. The effect of elevated temperature (non-

physiological) on percutaneous absorption was initially reported by Blank et

al., (54). Recently, there has been a surge in the interest of using

thermoregulation as means of improving the delivery profile of topical

medicaments. Previous in vitro studies59 have demonstrated a 2-3 fold increase

in flux for every 7-8°C rise in skin surface temperature. The increased

permeation following heat treatment has been attributed to an increase in drug

diffusivity in the vehicle and an increase in drug diffusivity in the skin due to

increased lipid fluidity. Vasodilation of the subcutaneous blood vessels as a

homeostatic response to a rise in skin temperature also plays an important role

in enhancing the transdermal delivery of topically applied compounds. The in

vivo delivery of nitroglycerin, testosterone, lidocaine, tetracaine and fentanyl

from transdermal patches with attached heating devices was shown to increase

as a result of the elevated temperature at the site of delivery. However, the

effect of temperature on the delivery of penetrants over 500 Da has not been

reported. The controlled heat-aided drug delivery patch (CHADD) (Zars Inc, Salt

Lake City, Utah) consists of a patch containing a series of holes at the top

surface which regulate the flow of oxygen in to the patch. The patch generates

heat chemically in a powder filled pouch by an oxidative process regulated by


the rate of flow of oxygen through the holes in to the patch. The CHADD

technology was used in the delivery of a local anaesthetic system (lidocaine and

tetracaine) from a patch (S-Caine®) and found to enhance the depth and

duration of the anaesthetic action in human volunteers when the results

obtained in active and placebo groups were compared. Zars Inc together with

Johnson and Johnson, recently submitted an investigational new drug (IND)

application to the FDA for Titragesia™ (a combination of CHADD disks and

Duragesic Patches, the latter contains fentanyl for treatment of acute pain).

Kuleza & Dvoretzky also describe a heat delivery patch or exothermic pad for

promoting the delivery of substances into the skin, subcutaneous tissues,

joints, muscles and blood stream, which may be of use in the application of

drug and cosmetic treatments. All the studies described above employed an

upper limit skin surface temperature of 40 - 42 oC, which can be tolerated for

a long period (> 1 h). In heat-patch systems where patient exposure to heat is ≤

24 h, such an upper limit may be necessary for regulatory compliance. In

addition, the issue of drug stability may also need to be addressed when

elevated temperatures are used. Thermopertubation refers to the use of

extreme temperatures to reduce the skin barrier. Such perturbation has been

reported in response to using high temperatures over a short duration (30ms),

with little or no discomfort, using a novel patch system 60. These investigators

developed a polydimethylsiloxane (PDMS) patch for non intrusive transdermal

glucose sensing via thermal micro-ablation. Ablation was achieved by

microheaters incorporated within the patch. The heat pulse is regulated by


means of a resistive heater, which ensures that the ablation is limited within

the superficial of dead layers of the skin. Average temperatures of 130 oC are

required for ablation to occur within 33 ms after which SC evaporation results.

Other heat assisted transdermal delivery devices under development include

the PassPort ® patch (Althea therapeutics) which ablates the SC via a similar

manner as the above described PDMS patch. The exposure of skin to low

(freezing) temperatures has been reported to decrease its barrier function but

has however not been exploited as means of enhancing skin absorption. The

final group of active enhancement methods entails the use of a physical or

mechanical means to breach or bypass the SC barrier.

1.5.13. Microneedle based devices

One of the first patents ever filed for a drug delivery device for the

percutaneous administration of drugs was based on this method 61. The device

as described in the patent consists of a drug reservoir and a plurality of

projections extending from the reservoir. These microneedles of length 50-110

μm will penetrate the SC and epidermis to deliver the drug from the reservoir.

The reservoir may contain drug, solution of drug, gel or solid particulates and

the various embodiments of the invention include the use of a membrane to

separate the drug from the skin and control release of the drug from its

reservoir. As a result of the current advancement in microfabrication

technology in the past ten years, cost effective means of developing devices in

this area are now becoming increasingly common. A recent commercialisation

of microneedle technology is the Macroflux® microprojection array developed


by ALZA Corporation. The macroflux® patch can either be used in combination

with a drug reservoir or by dry coating the drug on the microprojection array;

the latter being better for intracutaneous immunization. The lengths of the

microneedles have been estimated to be around 50-200 μm and therefore are

not believed to reach the nerve endings in the dermo-epidermal junction. The

microprojections/ microneedles (either solid or hollow) create channels in the

skin, hence allowing the unhindered movement of any topically applied drug.

Clinical evaluations report minimal associated discomfort and skin irritation

and erythema ratings associated with such systems are reportedly low. This

technology serves as an important and exciting advance in transdermal

technology due to the ability of the technique to deliver medicaments with

extremes of physicochemical properties (including vaccines, small molecular

weight drugs and large hydrophilic biopharmaceuticals) Yuzhakov et al.,

describes, the production of an intracutaneous microneedle array and provides

an account of its use (microfabrication technology). Various embodiments of

this invention can include a microneedle array as part of a closed loop system

‘smart patch’ to control drug delivery based on feedback information from

analysis of body fluids. Dual purpose hollow microneedle systems for

transdermal delivery and extraction which can be coupled with electrotransport

methods are also described by Trautman et al., Down et al., Allen et al.,62 These

mechanical microdevices which interface with electronics in order to achieve a

programmed or controlled drug release are referred to as

microelectromechanical systems (MEMS) devices.


Figure 1.11: Basic design of microneedle delivery devices. Needles of
approximately with or without centre hollow channels are placed onto the
skin surface so that they penetrate the stratum corneum and epidermis
without reaching the nerve endings present in the upper dermis.

1.5.14. Skin puncture and perforation

These devices are similar to the microneedle devices produced by

microfabrication technology. They include the use of needle-like structures or

blades, which disrupt the skin barrier by creating holes and cuts as a result of

a defined movement when in contact with the skin. Godshall and Anderson

(81), described a method and apparatus for disruption of the epidermis in a

reproducible manner. The apparatus consists of a plurality of microprotrusions

of a length insufficient for penetration beyond the epidermis. The

microprotrusions cut into the outer layers of the skin by movement of the

device in a direction parallel to the skin surface. After disruption of the skin,

passive (solution, patch, gel, ointment etc) or active (iontophoresis,

electroporation etc) delivery methods can then be utilised. Descriptions of other


devices based on a similar mode of action have been described by Godshall 63,

Kamen64, Jang65and Lin et al66.

1.5.15. Needleless injection

Needleless injection is reported to involve a pain free method of

administering drugs to the skin. This method therefore avoids the issues of

safety, pain and fear associated with the use of hypodermic needles.

Transdermal delivery is achieved by firing the liquid or solid particles at

supersonic speeds through the outer layers of the skin using a suitable energy

source. Over the years there have been numerous examples of both liquid (Ped-

O-Jet®, Iject® Biojector2000® Medi-jector® and Intraject®) and powder

(PMEDTM device formerly known as powderject® injector) systems. The latter

device has been reported to deliver successfully testosterone, lidocaine

hydrochloride and macromolecules such as calcitonin and insulin 66,67.

Problems facing needless injection systems include the high developmental cost

of both the device and dosage form and the inability, unlike some of the other

techniques described previously, to programme or control drug delivery in

order to compensate for inter-subject differences in skin permeability. In

addition, the long-term effect of bombarding the skin with drug particles at

high speed is not known thus, such systems may not be suitable for the

regular administration of drugs. It may however be very useful in the

administration of medicaments which do not require frequent dosing e.g.

vaccines.
1.5.16.Suction ablation

Formation of a suction blister, involves the application of a vacuum or

negative pressure to remove the epidermis, whilst leaving the basal membrane

intact. The cellpatch® (Epiport Pain Relief, Sweden) is a commercially available

product based on this mechanism. It comprises a suction cup, epidermatome

(to form a blister) and device (which contains morphine solution) to be attached

to the skin this method which avoids dermal invasivity there by avoiding pain

and bleeding is also referred to as skin erosion. Such devices have also been

shown to induce hyperaemia in the underlying dermis in in vivo studies which

was detected via laser Doppler flowmetry and confirmed via microscopy, and is

thought to further contribute to the enhancement of dextran and morphine

seen with this method. The disadvantages associated with the suction method

include the prolonged length of time required to achieve a blister (2.5 h),

although this can be reduced to 15-70 min by warming the skin to 38 oC. In

addition, whilst there is no risk of systemic infection compared to the use of

intravenous catheters, the potential for epidermal infections associated with

the suction method cannot be ignored even though the effects might be less

serious.

1.5.17.Application of Pressure

The application of modest pressures (i.e. 25 kPa) has been shown to

provide a potentially noninvasive and simple method of enhancing skin

permeability of molecules such as caffeine 68. These workers attributed the

increase in transcutaneous flux to either an improved transapendageal route or


an increased partition of the compound into the SC when pressure was

applied. This method may also work due to the increased solubility of caffeine

in the stratum corneum caused by the increase in pressure.

Figure1.12: Enhancement of transdermal permeation by pressure wave

1.5.18. Skin stretching

These devices hold the skin under tension in either a unidirectional or

multidirectional manner69,70. The authors claim that a tension of about 0.01 to

10 mP results in the reversible formation of micropathways. The efficiency of

the stretching process was demonstrated by monitoring the delivery of a

decapeptide (1 kDa) across the skin of hairless guinea pigs using a

microprotrusion array. The results of the study showed that the bi-directional

stretching of skin after microprotrusion piercing, allowed the pathways to stay

open (i.e. delayed closure) hence facilitating drug permeation to a greater extent

(27.9 ± 3.3 μg/cm2 h) than in the control group (9.8 ± 0.8 μg/cm2 h), where

the skin was not placed under tension after microneedle treatment. However,

increased skin permeation in the absence of microneedle pre-treatment was

found not to occur. Other methods involving the use of skin stretching with
subsequent use of delivery devices based on electrotransport, pressure,

osmotic and passive mechanisms have also been suggested but the value of

skin stretching alone without the benefit of a secondary active delivery device

remains to be seen.

1.5.19.Skin abrasion

These techniques, many of which are based on techniques employed by

dermatologists in the treatment of acne and skin blemishes (e.g.

microdermabrasion), involve the direct removal or disruption of the upper

layers of the skin to enhance the permeation of topically applied compounds.

The delivery potential of skin abrasion techniques are not restricted by the

physicochemical properties of the drug and previous work has illustrated that

such methods enhance and control the delivery of a hydrophilic permeant,

vitamin C vaccines and biopharmaceuticals. One current method is performed

using a stream of aluminium oxide crystals and motor driven fraises Sage &

Bock also describe a method of pre-treating the skin prior to transdermal drug

delivery which consists of a plurality of microabraders of length 50-200 μm.

The device is rubbed against the area of interest, to abrade the site, in order to

enhance delivery or extraction. The microabraders/microprotrusions terminate

as blunt tips and therefore do not penetrate the SC. The device functions by

removing a portion of the SC without substantially piercing the remaining

layer. Some of these methods are claimed to offer advantages such as minimal

patient discomfort, increased patient compliance, ease of use and less risk of

infection compared to their more “invasive” predecessors such as ablation and


the use of hypodermic needles/cannulas to deliver medicaments across the

skin.

1.5.20.Pharmaceutical carrier systems

Different types of pharmaceutical carriers are present. They are –

particulate, polymeric, macromolecular, and cellular carrier. Particulate type

carrier also known as a colloidal carrier system, includes lipid particles (low

and high density lipoprotein-LDL and HDL, respectively), microspheres,

nanoparticles, polymeric micelles and vesicular like liposomes, niosomes

pharmacosomes, virosomes, etc. 48- 51


these carriers were introduced for

different purposes and all will not have same qualities.

Figure1.13: various pharmaceutical carriers

1.6. Vesicular Carrier Systems

Since two decades the interest on the vesicles has been increased

tremendously because of the potential advantages of vesicles. Huge research

work is going on the vesicles to achieve different objectives. They become


vehicle of choice for drug delivery to researchers and are using these vesicles in

the areas like immunology, membrane biology, diagnostic techniques, genetic

engineering, etc and are using in the transport and targeting of active

pharmaceutical ingredients.

The vesicular systems are highly ordered assemblies of one or several

concentric lipid bilayers formed, when certain amphiphilic building blocks are

confronted with water. Vesicles can be formed from a diverse range of

amphiphilic building blocks. The terms such as synthetic bilayers allude to

the non-biological origin of such vesiculogenes. Biologic origin of these

vesicles was first reported in 1965 by Bingham71, and was given the name

Bingham bodies.

Figure1.14: Structure of typical vesicle

The different advantages of the vesicular carrier systems are as

follows.

1. Lipid vesicles have evolved successfully, as vehicles for controlled delivery.


2. Vesicles can be administered from different routs such as oral, IM, nasal,

transdermal, vaginal, rectal, ocular, etc. and possible to produce both the

localized and/or systemic effects.

3. Enahnaces the bioavailability of the drugs by increasing the mean residence

time in biological environment and/or by enhancing the solubility.

4. Reduces the adverse effects associated with the high doses of the drugs by

increasing the bioavalability of the active principles.

5. Enhances the stability of the drugs by avoiding the exposure of the drugs to the

susceptible environments.

6. Drug targeting can be possible by means of these vesicles.

7. Both hydrophilic and lipophilic drugs can incorporate in these vesicles.

As these vesicles have potential applications, the interest on these

vesicles have been increased and developed different vesicles for different

purposes. The different vesicles include liposomes, niosomes, ethosomes,

transferosomes, pharmacosomes, etc.

1.6.1. LIPOSOMES

Liposomes are simple microscopic lipid vesicles ranging from 20

nanometers to several micrometers in size and in these vesicles the lipid bilayer

encloses the aqueous environment. Both hydrophilic and lipophilic drugs can

be successfully entrapped72 in these vesicles. Hydrophilic and lipophilic drugs

entrapped in the central aqueous environment and within the lipid bilayers

respectively[6]. Liposomes can be used for delivery of drugs through oral,

topical, ocular, vaginal, etc routes.


Figure 1.15: structure of conventional liposome

Phospholipid and cholesterol are the main ingredients for manufacturing

of liposomes. Phosphatidylcholine (PC), a natural phospholipid, is most

commonly used for preparation of liposomes. One can also use other

phospholipids such as phosphatidyl ethanolamine (PE), phosphatidyl serine

(PS), phosphatidyl inositol (PI) and phosphatidyl glycerol (PG) for

manufacturing of liposomes. Cholesterol is used for different purposes such as

fluidity buffer and also provides rigidity to the lipid bilayer.

Figure 1.16: Chemical structure of Phosphatidylcholine (PC)

Liposomes, as particulate carriers, naturally can target cells of the


mononuclear phagocytic system(MPS), particularly macrophages to treat a

number of diseases such as infectious diseases, cancer and atherosclerosis

and inflammatory diseases. To achieve targeting of liposomes to monocytes,

macrophages and dendritic cells, the physicochemical properties of liposomes

has been modified by addition of surface ligands such as proteins, peptides,

antibodies, polysaccharides, glycolipids, glycoproteins and lectins. Doijad

Rajindra C et al prepared zidovudine, loaded liposomes for targeting to liver

followed by lungs, kidney and spleen against human immunodeficiency virus

(HIV)73. Though this liposomes have wide advantages, its success rate

decreased due to its bigger size, impermeable to deeper layers of the skin,

fusion, leakage, etc.

The proposed mechanisms for drug delivery across the skin through

liposomes are intact vesicular skin permeation, the penetration enhancing

effect, the adsorption effect and the penetration of liposomes through the

transappendageal route.

1.6.2. EMULSOMES

Emulsomes are lipid vesicles designed for poorly soluble drugs to

administer them through the parenteral route 74. The internal core of

emulsomes is prepared with fats and triglycerides and is stabilized in form of

o/w emulsion by adding high concentration of lecithin. These will posses both

liposomes and emulsions characteristics. Entrapment efficiency of lipophilic

drugs can be enhanced by the process of solidification or semisolidification of

internal oily core which also controls the release of drugs from vesicles. As
liposomes, in these vesicles also one can encapsulate hydrophilic drugs in

aqueous compartments of surrounding phospholipid layers. The solvent-

free and surfactant-free emulsome technologies were developed to increase the

encapsulation capacity of water insoluble antifungal and anticancer drugs

which showed improved preclinical efficacy for oral route. Vyas et al developed

zidovudine emulsomes for sustained and targeted drug delivery to liver for the

treatment of life-threatening viral infections like hepatitis, HIV and Epstein-

Barr virus infection.

Figure 1.17: Structure of emulsomes

1.6.3. ETHOSOMES

These lipid vesicles developed to deliver the drugs having poor

penetration through the skin. They are soft in nature having size range from

tens of nano meters to microns. These vesicles contains more concentration of

ethanol (20-50%) which brings soft malleable nature to the vesicles and also

solubilizes the inter cellular lipid and enhances the penetration of the drugs
through the skin75. Ethanol induces surface negative net charge to the vesicles

which decreases the size of the vesicles. Concentration of ethanol in vesicles is

inversely proportional to the size of the vesicles. Ethosomes has been used as

carrier for delivery of many drugs through the skin successfully to produce

both localized and/or systemic effects. The drugs includes antifungal agents

(fluconazole) antiviral agents (lamivudine, acyclovir, stavudine, and zidovudine),

NSAIDS (aceclofenac, diclofenac), antibiotics (cannabidol, erythromycin), etc. in

all the studies ethosomes has shown enhanced therapeutic effect when

compared with conventional methods.

Exact mechanism of drug permeation across the skin through ethosomes

is not proposed. It is assumed that a combination of two processes contribute

to the enhancing effect. The two processes are solubilisation of stratum

corneum lipid and on vesicles fluidity as well as dynamic interaction between

ethosomes and the stratum corneum.


Figure 1.18: Proposed mechanism for penetration of molecule from
ethosomal system across the lipid domain of stratum corneum

1.6.4. ENZYMOSOMES

These are developed to deliver the therapeutic proteins like enzymes and

are developed by complexing the enzymes with the lipids. Superoxide

Dismutase, a therapeutic agent for oxidative stress related diseases like

rheumatoid arthritis and ischaemia/reperfusion situations, loaded

enzymosomes have been developed for long circulation time in the blood, in

order to accumulate at inflammed target sites, while maintaining enzymatic

activity in its intact form.

1.6.5. SPHINGOSOME

These are introduced to overcome the stability problems associated with

liposomes and are called as sphingosomes due to the presence of the


sphigolipids in place of phospholipids in liposomes. Phospholipids readily

undergo chemical degradation such as oxidation, hydrolysis due to the

presence of ester linkage. In sphingolipids either an ether or amide linkage will

present which are resistant to chemical degradation and brings stability to the

sphingolipids than the phospholipids. Sphingosomes are efficient carriers

because of high stability, for targeting of the drug to the site of action and are

being biodegradable, innocuous nature and also identical to biological

membrane.

Sphingosomes offer selective passive targeting to tumour tissues and

flexibly bind with site-specific ligands to achieve active targeting. These are

much more stable to acid hydrolysis and have better drug retention

characteristics. Sphingosomal products e.g., Marqibo(TM) (vincristine loaded

sphingosomes) are loaded with active, cell cycle-specific anticancer agents that

are benefited from increased targeting and long duration of drug exposure at

the tumor site. Vinorelbine and topotecan are also selected for sphingosomal

formulation specifically for their ability to benefit from this novel

encapsulation.

Figure 1.19: structure of sphingolipids


1.6.6. TRANSFEROSOMES

Transferosome was introduced by GregorCevc in the year 1991. The

word “Transferosome” means “carrying body” and is derived from Latin word

“transferre” means “to carry across” and the Greek word “soma” means

“body”. The unique nature of these vesicles is that they have deformable

nature. Due to its deformable nature these vesicles penetrate through the

pores having less size than the vesicles and deliver the drugs across the skin

without making puncture of the skin and with less loss.

Figure 1.20: Structure of transferosomes

These are used as carriers for many drugs such as hoprmones,

proteins, peptides and various drug molecules. These vesicles are also used

successfully to deliver the active principles, such as insulin, interferons,

which are highly unstable in GIT, through the skin. Large molecular weight

substances such as proteins are difficult to deliver through skin due to their

large m.wt which can also delivered successfully through the skin by these

vesicles. In a study it was shown that these vesicles delivered diclofenac


sodium 10times more than the commercial hydrogel across the skin and

shown longer effect76. Anti-HIV agents like zidovudine, indinavir, etc also

enclosed in transferosomes and characterized for delivery of drugs across the

skin which shown higher concentration of drug in the systemic circulation.

Two mechanisms have been proposed to explain the drug permeation

across the skin by means of transferosomes. First, vesicles can act as drug

carrier systems, where by intact vesicles enter the saturation stratum

corneum carrying vesicle bound drug molecules into the skin. Second vesicles

can acts as penetration enhancers, whereby vesicle bilayers enter the stratum

corneum and subsequently modify the inter cellular lipid lamellae.

Figure1.21: Mechanism of transferosomes permeation across the

skin

1.6.7. PHARMACOSOMES
Pharmacosomes are novel vesicular drug delivery systems having unique

advantages over other drug delivery systems 77. Pharmacosomes are amphiphilic

lipoidal colloidal dispersions of drugs, covalently bound to lipids with potential

to improve bioavailability of poorly water soluble as well as poorly lipophilic

drugs. Any drug possessing a free carboxyl group or an active hydrogen atom(-

OH, -NH2) can be esterified (with or without a spacer group) to the hydroxyl

group of a lipid molecule, thus generating an amphiphilic prodrug. An

amphiphilic prodrug is then converted to pharmacosomes upon dilution with

water. The prodrug conjoins hydrophilic and lipophilic properties (thereby

acquiring amphiphilic characteristics), reduces interfacial tension, and at

higher concentration exhibits mesomorphic behavior. Because of decrease in

interfacial tension, the contact area increases, therefore bioavailability also

increases. As the drug is covalently conjugated with lipids, loss due to leakage

of drug does not occur. Hence, provides maximum entrapment efficiency. The

three main components for the preparation of pharmacosomes are drug,

solvent and lipid. Drug should contain active hydrogen atom (-COOH, OH,

NH2), can be esterified with lipid and form amphiphilic complexes, which

facilitate membrane transfer. The solvent should have high purity, volatility

and intermediate polarity (between the polarity of phospholipid and drug) for

the preparation of pharmacosomes. The most commonly used lipid for the

preparation of pharmacosomesis phosphatidylcholine. The pharmacosomes can

be prepared by hand-shaking and ether-injection methods. These have been

prepared for various non-steroidal anti-inflammatory drugs, proteins,


cardiovascular and antineoplastic drugs. Drug release from pharmacosomes is

by hydrolysis (including enzymatic) unlike liposomes the release of drug is by

diffusion through bilayer, desorption from the surface or degradation of

liposomes.

1.6.8. VIROSOMES

Virosomes are reconstituted viral envelopes that are composed of a lipid

bilayer in which inserted viral glycoproteins can be derived from different

enveloped viruses. Virosomes are described as liposomes with influenza virus

hemagglutinin (HA) and neuraminidase (NA) spikes on their surface.

Virosomes closely mimic the intact virus except that they do not contain virus

replication machineries. They retain the cell entry and membrane fusion

characteristics of the virus derived from. The two pathways by which

reconstituted vesicles are able to enter the cells and deliver their contents into

the cytoplasm are plasma membrane fusion(Sendai virus) and acid-induced

fusion from within endosomes (Influenza virus). As a result, foreign

substances encapsulated within the lumen of virosomes are effectively

delivered to the cytosol of target cells. Virosomes can be used in vaccination

for the efficient induction of antibody responses against the virus they are

derived from78.

1.6.9. NON-IONIC SURFACTANT VESICLES/NIOSOMES

First, in 1979, Handjanivila et al reported the niosomes will form upon

hydration of a mixture of cholesterol and single alkyl chain. These are also said

to be non-ionic surfactant vesicles. Since 1979, different non-ionic surfactants


such as polyglycerol alkyl ethers, glucosyldialkyl ethers, crown ethers,

polyoxyethylene alkyl ethers, ester linked surfactants, steroid linked

surfactant, brij and a series of spans, tweens, etc have to used to manufacture

the niosomes79. These have microscopic lamellar structure of size range 10-

1000nm consisting of spherical, uni or multilamellar and polyhedral vesicles in

aqueous media. cationic, anionic and ampholytic surfactants also used to

prepare niosomes but as they causes skin irritation, non ionic surfactants are

preferred which causes no irritation. These niosomes have many advantages

over lipid vesicles with respect to stability, manufacturing and storage. These

will not require special condition for manufacturing and storage because the

non ionic surfactants are less prone to oxidation comparatively lipids which are

used in preparation of liposomes and other lipid vesicles. These have shown

better patient compliance and better therapeutic effect in comparison to oily

formulations. Niosomes are osmotically active and stable and whose shape,

size, composition and fluidity of niosomes can be controlled as and when

required.
Figure 1.22: Non-ionic surfactant vesicle / Niosome

1.6.10. BILOSOMES

Bilosomes are the novel innovative drug delivery carriers consist of

deoxycholic acid incorporated into the membrane of niosomes. As

conventional vesicles (liposomes and niosomes) can cause dissolution and

undergo enzymatic degradation in gastro intestinal tract but incorporation of

bile salts (commonly used penetration enhancers) in niosomal formulation

could stabilize the membrane against the detrimental effects of bile acids in GI

tract. These bile salt stabilized vesicles are known as bilosomes. These are

highly biocompatible and have been found to improve the therapeutic efficacy

of drugs due to their stability in gastro intestinal tract. Bilosomes have been

found to increase the bioavailability of drugs as they can readily absorbed

through small intestine to the portal circulation (hepatocirculation). Through

this circulation they approach to liver and release the drug, so found to be an

effective tool in drug targeting to liver. Shukla et al showed that HBsAg loaded

bilosomes produced both systemic as well as mucosal antibody responses

upon oral administration. For extended humoral, cell-mediated and mucosal

immune responses, additional coating carrier system provided better

protection against disease for longer period of time. Optimum mannan coating

was found to stabilize the vesicles in gastrointestinal environment and also act

as a targeting ligand for mannose receptors expressed on macrophages and

dendritic cells80.
1.6.11. AQUASOMES

Aquasomes firstly developed by Kossovsky, are one of the most recently

developed delivery system for bioactive molecules 81. Aquasomes are three

layered structures (i.e. core, coating and drug) that are self-assembled

through non covalent bonds, ionic bonds and vander waals forces. They

consist of tin oxide, nanocrystalline carbon ceramic (diamonds) or brushite

(calcium phosphate dihydrate) core coated with oligomeric film to which

biochemically active molecules are adsorbed by copolymerization, diffusion or

adsorption with or without modification. The solid core provides the structural

stability, while the carbohydrate coating protects against dehydration and

stabilizes the biochemically active molecules. Aquasomes are spherical 60-

300nm size particles called Âbodies of waterÊ. Their water like properties

protects and preserves fragile biological molecules. Mechanism of action of

aquasomes is controlled by their surface chemistry, which deliver contents

through combination of specific targeting, molecular shielding and slow and

sustained release process. Due to their size and structural stability, these

avoid clearance by reticuloendothelial system and degradation by other

environmental changes. Aquasomes can be used as red blood cell substitutes

for the release of oxygen by haemoglobin. Aquasomes can be used as vaccines

for delivery of viral antigen, for targeted intracellular gene therapy, for delivery

of insulin and enzymes like DNAase and pigments/dyes.


Chapter - 2

LITERATURE REVIEW
Chapter - 2: Literature review

Table of contents

S.No Name of the subtitle Page No


2.1 Literature on niosomes  
2.2 Literature on ethosomes  
2.3 Literature on transferosomes  
2. LITERATURE REVIEW

Baillie et al82 (1985), have studied niosomes prepared by liposomal technology

and has concluded that niosomes are promising vehicles for drug delivery to

target organs, it was also concluded that these are less toxic than the vesicles

prepared from ionic surfactants.

Rogerson et al83 (1988), have studied the distribution of doxorubicin in mice

following the administration of niosomes and concluded that half life of the

drug were prolonged compared with the free drug solution profile and also

concluded that metabolism and maintenance of anti tumour effects are altered

when administered with the niosomes.

Chandra prakash et al84 (1990), have studied the pharmacokinetics of

surfactant vesicles entrapped methotrexate in tumour bearing mice and has

concluded that entrapment efficiency was increased with increase in the

lipophilicity of the surfactant and also concluded that its pharmacokinetics in

mice was markedly different in comparison to unentrapped drug.

Raja Naresh et al85 (1993), have studied the anti-inflammatory action of

niosomes encapsulated diclofenac sodium in rats and has concluded that

niosomal encapsulated diclofenac sodium shows higher anti-inflammatory

action than the diclofenac sodium when administered intra-peritoneally and

transdermally.

Uduppa et al86 (1993), have formulated and evaluated the methotrexate

niosomes and has concluded niosomes produce sustained activity and

niosomes are useful carrier for delivery of the methotrexate.


Malhotra et al87 (1994), have studied the properties and preparation of

niosomes for the delivery of bioactive agents and various drugs.

Partha Sarathi et al88 (1994), have studied the anti-cancer activity of vincristine

sulphate encapsulated niosomes in mice and has concluded that anti-cancer

activity increased with reduced toxicity.

Partha Sarathi et al89(1994), have formulated and evaluated the vincristine

encapsulated niosomes and have concluded that transmembrane pH gradient

drug uptake process was most satisfactory drug release from niosomes slowed

down and also stability of the system was enhanced.

Naresh et al90 (1996), have studied the antitumor activity of niosomes

encapsulated bleomycin and have concluded that these shows better

antitumor activity due to better delivery of bleomycin to the tumor site.

Raja naresh et al91 (1996), have studied the kinetics and tissue distribution of

niosomal bleomycin in tumour bearing mice and have concluded plasma

concentration of bleomycin significantly increased and removal of it from the

plasma decreased much when it administered niosomally than the free drug.

Arun et al92 (1998), have studied the effect of niosomes on the kinetics of

ketorlac tromethamine and have concluded that niosomes increases the

plasma level and biological half life of the drug significantly than the plain

drug.

Erdogan et al93 (1998), have studied the stability of niosomes and liposomes

encapsulated with iopromide and have concluded that niosomes have greater

stability than the liposomes.


Sheena et al94 (1998), have studied the antitumour efficiency of niosomal

entrapped withaferin-A and plain withaferin-A in mice and have concluded

that antitumor efficiency of niosomal encapsulated formulation is superior

than plain formulation.

Namdeo et al95 (1999), have prepared and evaluated the niosomes encapsulated

indomethacin and have concluded that niosomal encapsulated indomethacin

shown superior anti-inflammatory activity than the plain indomethacin

formulation.

Devi et al96 (2000), have studied the niosomal entrapped sumatriptan

succinate for nasal administration.

Ruckmani et al97 (2000), have prepared the niosomes encapsulated cytarabin

hydrochloride by film hydration method and evaluated for encapsulation,

storage and In vitro drug release and have concluded that drug release profile

was increased significantly and stability profile was good over long period.

Fang et al (2001)98, have studied the In vitro skin permeation from various

proniosomal gel formulations across the excited rat skin and have concluded

that the permeation of estradiol was increased after entrapping it in niosomes.

Ravichandran et al99 (2001), have prepared and evaluated the In vitro release

of diclofenac sodium from niosomes and have concluded that niosomal

formulations are stable and the drug release was extended for long period.

Ruckmani et al100 (2001), have studied the cytarabin (1-beta-D-

arabiofuranosylcytosine) entrapped niosomes and concluded that the


antitumor activity of cytarabin in mice was enhanced after encapsulation in

niosomes.

Alia sagar Shashiwala et al101 (2002), have studied the topical niosomal

encapsulated nimesulide gel and concluded that drug release was prolonged,

retention of the drug is increased, and permeation across the skin and anti

inflammatory activity have increased after entrapping in niosomes when

compared with plain nimesulide gel.

Balasubramaniam et al102 (2002), have prepared and evaluated niosomal

encapsulated daunorubicin hydrochloride formulation and concluded that

niosomal formulation shows greater /superior anti-tumor activity than the free

drug treatment.

Dutta Shivani et al103 (2002), have studied the niosomal delivery of plumbagin

ester and have concluded that it shows better anti fertility activity also reduces

toxicity of plumbagin ester.

Joseph et al104 (2002), have studied antitumor activity of etoposide

encapsulated niosomes against daltons lymphoma cells in Swiss albino mice

and have concluded that niosomes have the potential to produce significant

antitumor effect than the free drug and also increases the percentage of tumor

cell inhibition.

Manconi et al105 (2002), have prepared and evaluated the properties of trenitoin

loaded niosomes and have concluded that entrapment efficiency depends upon

the structure of the vesicle and In vitro drug release from niosomal formulation

is faster than from trenitoin solution.


Roux et al106 (2002), have studied the pH sensitive niosomal and liposomal

formulations bearing alkylated N-isopropylacrylamide copolymers with respect

to vesicle-polymer interaction, pH-responsiveness and stability in human

serum and have concluded that both formulations were released their contents

under mild acidic conditions, the niosomes lost their pH-sensitivity after

incubation in serum whereas liposomes maintained their ability to respond to

pH only when complexed with copolymer containing a high proportion of

hydrophobic anchor and also concluded that the interactions that take place

between the polymer and the vesicles strongly depends on the vesicle nature.

Satturwar et al107 (2002), was formulated and evaluated the ketoconazole

niosomes and have concluded that niosomes have the potential to reduce the

therapeutic dose of ketoconazole improving the performance.

Varshasoz et al108 (2003), have prepared insulin entrapped niosomes by film

hydration method and found that vesicles containing span 60 showed the

highest protection of insulin against proteolytic enzymes and good stability in

the presence of sodium desoxycholate & storage temperatures.

Aggarwal et al 109
(2004), have studied the topical niosomal preparation of

acetazolamide by ether injection method, lipid hydration method and reverse

phase evaporation methods and have concluded that drug entrapment

efficiency and percent of permeation vary with the charged and neutral

niosomes and also shown that topical niosomal formulation shows greater

physiological activity than the oral preparation of acetazolamide.


Dufes et al110 (2004), have studied the glucose targeted niosomes deliver

vasoactive intestinal peptide to the brain and have concluded that glucose

bearing vesicles are a novel tool to deliver drug across the blood brain barrier.

Mullaicharam et al111 (2004), have studied the organ distribution pattern of

niosomes encapsulated rifampicin compared with that of free rifampicin

solution in albino rats to find out the potential delivery system for the site

specific deliver of rifampicin and also to find out the most effective route of

administration in intravenous and intra tracheal routes and have concluded

that best route of administration is intra-tracheal administration and there is

significant difference in the distribution of rifampicin administered in the form

of solution and administered after entrapping in niosomes.

Aggarwal et al112 (2005), have prepared mucoadhesive niosomal ophthalmic

drug delivery system of timolol maleate and compared with the timolol maleate

solution in terms of in-vitro drug release and intra-occular pressure lowering

pharmacodynamic effect, finally concluded that niosomal formulation shown

significantly better results than the timolol maleate solution.

Alsarra et al113 (2005), have studied the uptake of atenolol from GIT tract

using the niosomes as drug carriers / permeation enhancers and have

concluded that atenolol entrapped in niosomes shows greater absorption. This

is evidenced by showing the greater permeation through an intestinal sac.

Guinedi et al114 (2005), have studied the bioavailability and corneal penetration

of the acetazolamide by loading the drug in niosomes and have concluded that
niosomes decreases the intra-ocular pressure significantly than the free drug

solution.

Huang et al115 (2005), have prepared cationic niosomes of sorbiton

monostearates by film hydration method and evaluated for their effect on

delivery of antisense oligonucleotides and the formulation results showed

positive results on cellular uptake of antisense oligonucleotides.

Jain et al116 (2005), have studied niosomes as carriers adjuvant system for

topical immunization and have concluded that niosomal immunization is more

effective when compared other topical immunizations.

Nasseri et al117 (2005), have studied the effect of cholesterol and temperature

on the elastic properties of niosomal membranes and have concluded that

interaction between the cholesterol and sorbitan monosterates should increase

the rigidity of the membranes.

Ning et al118 (2005), have prepared and evaluated the vaginal administering

niosomal clotrimazole gel and have concluded that it shows greater antifungal

activity than the plain clotrimazole gel.

Jain et al119 (2006), have studied the niosomal system for deliver of

rifampicin to lymphatic and have concluded that rifampicin encapsulated in

niosomes can successfully be used for the treatment of tuberculosis along

lymphatic system.

Manconi et al120 (2006), have studied the influence of thermodynamic activity

and niosome composition, size, lamellarity and charge on the transdermal

delivery of trenitoin and have concluded that small, negatively charged


niosomal formulations, which are saturated with trenitoin have shown to give

higher cutaneous drug retention than liposomes and marketed formulation.

Manivannan Rangasamy et al121 (2008) developed Acyclovir entrapped

niosomes by hand shaking and ether injection process with different ratios of

(1:1, 1;2 and 1:3) cholesterol (CHOL) and Span-80 (Non-ionic surfactant). They

found that the size range of niosomes prepared using ether injection method is

less than the niosomes prepared using hand shaking method. They also found

that the order of encapsulation efficiency increases when span-80

concentration was increased and also found In-vitro release of acyclovir more

from formulation prepared with CHOL: Span-80 (1:1).

Naresh Ahuja et al 122


(2008) developed Lansoprazole entrapped niosomes by

reverse phase evaporation method using a homogenizer and characterized for

its size rang, entrapment efficiency and in-vitro release of drug. They concluded

that they will reduce the systemic toxicity of drugs.

V. Sankar et al123 (2009) developed carboplatin niosomes for enhanced delivery

to cancer cells by thin film hydration technique using (cholesterol: surfactant)

in the micromolar ratio of 30:100 and with/without the addition of charge

inducing agents and Pluronic F 68. The In vitro cytotoxicity results of the

formulations reveal that the Tween 80 formulation with Pluronic shows the

highest level of cytotoxicity (90%) when compared with drug in solution. Also,

the stability studies of the formulations reveal that the formulations were

stable in refrigerated condition.


Gupta Naveen et al124 (2010) developed and evaluated Niosomes, a synthetic

microscopic vesicles consisting of an aqueous concentration enclosed in a

bilayer consisting of cholesterol and one or more nonionic surfactants to

improve the low corneal penetration and bioavailability characteristic shown by

conventional ophthalmic vehicles. They concluded that Niosomes formed from

span 60 and cholesterol in the ratio 200:100 (in mol) is a promising approach

to improve the bioavailability of ofloxacin even for an extended period of time

which showed good physicochemical properties, good stability and controlled

drug release pattern, thereby improving the bioavailability of the drug.

Y. Anand Kumar et al125 (2010) developed and optimized niosomal formulation

of aceclofenac in order to improve its bioavailability. They studied the effect of

the varying composition of non ionic surfactant and cholesterol on the

properties such as encapsulation efficiency, particle size and drug release.

They concluded that the bioavailability of aceclofenac can increase by this

technology.

Kandasamy Ruckmani et al126 (2010) developed Zidovudine entrapped

niosomes and characterized process-related variables like hydration and

sonication time, rotation speed of evaporation flask, and the effects of charge-

inducing agent and centrifugation on zidovudine entrapment and release from

niosomes ans stated that niosomes can be formulated by proper adjustment of

process parameters to enhance zidovudine entrapment and sustainability of

release. These improvements in zidovudine formulation may be useful in

developing a more effective AIDS therapy.


N. Paval rani et al127 (2010) prepared niosomes of rifampicin and gatifloxacin by

lipid hydration technique using rotary flash evaporator and evaluated for size,

entrppment efficiency and bactericidal activity. The prepared rifampicin and

gatifloxacin niosomes showed a vesicle size in the range of 100-300nm, the

entrapment efficiency were 73% and 70% respectively. The invitro release study

showed that 98.98% and 97.74% of release of rifampicin and gatifloxacin

niosomes respectively. The bactericidal activities of the niosomal formulation

were studied by BACTEC radiometric method using the resistant strains (RF

8554) and sensitive strains (H37Rv) of Mycobacterium tuberculosis which

showed greater inhibition and reduced growth index.

Saggam Srinivas et al128 (2010) developed aceclofenac niosomes and evaluated

for the effect of varying composition of non ionic surfactant and cholesterol on

the properties such as entrapment efficiency, particle size and drug release.

Release of the drug was modified and extended over a period of 72 h in all

formulations. The mechanism of dug release was found to be non fickian

(anomalous) diffusion governed by Peppas’ model.

D. Akhilesh et al129 (2011) possibility of manufacturing proniosomes and using

proniosome-based niosomes as drug carriers by preparing the Gliclazide

Loaded Maltodextrin Based Proniosomes. The study revealed that Proniosomes

are very promising as drug carriers.

Chawda Himmat Singh et al130 (2011) prepared niosome-encapsulated drug

delivery for anti-inflammatory drugs like nimesulide using different Non-ionic

surfactants like span 20, 40, 60 and cholesterol was used in different molar
ratios by lipid film hydration method and ether injection technique and

evaluated the vesicle size, encapsulation efficiency, In vitro release and physical

stability of the niosomes. Finding of all this investigation conclusively

demonstrate prolongation of drug release at a constant and controlled rate

after niosomal encapsulation of nimesulide.

P.U.Mohamed Firthouse et al131 (2011) formulated miconazole entrapped

Niosomes by varying the cholesterol and surfactant ratios as 1:0.5, 1:1,1:1.5 by

Thin Film Hydration Technique and each formulation was evaluated for

percentage of drug entrapment and for their cumulative drug release. They

found that formulation with 1:1 CHOL: SA ratio has shown higher entrapment

efficiency and drug release was found up to 24 hours.

M. Abraham Lingan et al132 (2011) formulated topical gel containing clobetasol

propionate niosomes to prolong the duration of action and prevent its side

effects and evaluated the same. The niosomes were prepared using by varying

the concentration ratios between various non-ionic surfactants (Span 40, 60,

80) and cholesterol by three methods such as thin film hydration method,

ether injection method and hand shaking methods. The results suggested that

the niosomal delivery of clobetasol propionate in carbopol gel base acts as a

suitable topical drug delivery system to prolong the duration of action.

Vyas Jigar et al133 (2011) developed erythromycin entrapped niosomes by thin

film hydration technique and various process parameters were optimized by

partial factorial design and concluded demonstrates prolongation of drug

release, an increase in amount of drug retention into skin and improved


permeation across the skin after encapsulation of Erythromycin into niosomal

topical gel.

V. satyavathi et al134 (2012) developed niosomal in situ gel ocular delivery

system of brimonidine tartrate to enhance bioavailability and evaluated the

same and concluded that the niosomal preparations are one of the tool to

enhance the bioavailability.

Pardakhty A et al135 (2012) developed different positively charged niosomal

formulations containing sorbitan esters, cholesterol and cetyl trimethyl

ammonium bromide by film hydration method for the entrapment of autoclaved

Leishmania major (ALM). Size distribution pattern and stability of niosomes

were investigated by laser light scattering method and ALM encapsulation per

cent was measured by the bicinchoninic acid method. Finally, the selected

formulation was used for the induction of the immune response against

cutaneous leishmaniasis in BALB/c mice. The percentage of ALM entrapped in

all formulations varied between 14.88% and 36.65%. In contrast to ALM, In

vivo studies showed that the niosomes containing ALM have a moderate effect

in the prevention of cutaneous leishmaniasis in BALB/c mice.

Disha Mehtani et al136 (2012) have employed the concept of mixed solvency, to

investigate the various solubilizers, to increase the solubility of surfactants and

the intended drug in diethyl ether, formulate niosomal gel for transdermal use

and perform its evaluation and concluded that this is one of the useful

technology for development of particulate carriers.


Rajesh Z. Mujoriya et al137 (2012) developed ketoprofen niosomes using various

concentrations of Surfactant (Span 60) Cholesterol and Dicetyl phosphate

(DCP) by thin film hydration method and evaluated all formulations. They

concluded based on the results that the optimized niosomal formulation

showed a sustained action of about 16 hrs was subjected to In vivo studies

(anti-inflammatory activity). This formulation was found to be more effective in

reducing edema after 2 hrs as compared to the free drug.

Guiling et al (2012)138 have prepared tracolimus loaded ethosomes to

characterize a novel ethosomal carrier for tacrolimus, an immunosuppressant

treating atopic dermatitis (AD), and to investigate inhibition action upon

allergic reactions of mice aiming at improving pharmacological effect for

tacrolimus in that commercial tacrolimus ointment with poor penetration

capability exhibited weak impact on AD compared with common glucocorticoid.

The results shown that the ethosomes showed lower vesicle size and higher

encapsulation efficiency (EE) as compared with traditional liposomes with

cholesterol. In addition, thequantity of tacrolimus remaining in the epidermis

at the end of the 24-h experiment was statistically significantly greater from

the ethosomal delivery system than from commercial ointment which indicates

the greater penetration capability of the ethosomes into the deep strata of the

skin. Finally concluded the ethosomal tacrolimus delivery systems may be a

promising candidate for topical delivery of tacrolimus in treatment of AD.

Manish K. Chourasia et al (2011) 139 developed ketoprofen loaded ethosomes

with an aim to study the potential applications of ethosomes as carrier for


improved transdermal drug delivery and evaluated the same. In the study they

revealed that there is improved drug permeation through the skin from

ethosomes than the hydroethonolic solution. The study also revealed that the

drug concentration in plasma increased with ethosomes rather than the

hydroethonilic solution.

Limsuwan et al140 (2012) developed ethosomes containing mycophenolic acid

with an aim to enhance the permeation of drug across the skin upon topical

administration and characterized for the same and theu substiated the same.

Godin et al141 (2004) have developed bacitricin loaded ethosomes with an aim to

investigate the dermal and intracellular delivery of bacitracin, a model

polypeptide antibiotic, from ethosomes and characterized. The evaluation

substantiated that the antibiotic peptide was delivered into deep skin layers

through intercorneocyte lipid domain of stratum corneum (SC). Occlusion had

no effect on the permeation profile of the drug from ethosomes in in vitro

experiments. Efficient delivery of antibiotics to deep skin strata from ethosomal

applications could be highly beneficial, reducing possible side effects and other

drawbacks associated with systemic treatment. Further the researchers

revealed, ethosomal delivery systems could be considered for the treatment of a

number of dermal infections, requiring intracellular delivery of antibiotics,

whereby the drug must bypass two barriers: the SC and the cell membrane.

Vaibhav Dubey et al142 (2007) have prepared melatonin loaded ethonolic

liposomes and characterized for physicochemical properties and transdermal

drug delivery. The results shown that a better skin tolerability of ethosomal
suspension on rabbit skin suggested that ethosomes may offer a suitable

approach for transdermal delivery of melatonin.

Vaibhav Dubey et al143 (2007) have developed ethosomes containg methotrexate

(MTX), an anti-psoriatic, anti-neoplastic, highly hydrosoluble agent, having

limited transdermal permeation, to evaluate the transdermal potential of novel

vesicular carrier, ethosomes and characterized for the same. The results shown

that ethosomes are an efficient carrier for dermal and transdermal delivery of

MTX.

Maria manconi et al144 (2011) have developed ethosomes containing diclofenac

to study the potentiality of carriers in permeation of drugs across the skin and

characterized for the same. The results the superior ability of the PEVs to

enhance ex vivo drug transport of both hydrophilic and lipophilic diclofenac

forms.

Vijay kumar et al145 (2010) have prepared diclofenac potassium enclosed

ethosomes with an aim to evaluate the transdermal potential of novel vesicular

carrier, ethosomes, having diclofenac potassium, a potent, water soluble non‐

steroidal anti‐inflammatory drug with lesser transdermal permeation and the

results are compared with the efficiency of marketed preparation. The

pharmacodynamic studies showed enhanced anti‐inflammatory activity of

ethosomal gel than the marketed gel formulation and also confirmed that the

ethosomes are an efficient carrier for dermal and transdermal delivery of

diclofenac potassium.
Ashoniya et al146 (2011) developed ethosomes containing ketoconozole and

evaluated. The researchers concluded based on the results that the ethosomes

are prominent tools for transdermal delivery of drugs.

Xingyan Liu et al147 (2011) have prepared ligustrazine loaded ethosomes and in

vitro and in vivo evaluation was carried out and the results are compared with

conventional ligustrazine administration. The comparison concluded confirmed

that ligustrazine ethosome patches could promote better drug absorption and

increase bioavailability. This study also demonstrated that the transdermal

action of the ligustrazine ethosome patch was comparatively good.

Elisabetta Esposito et al148 (2004) have developed ethosomes and liposomes

containing azelaic acid and preformulation study was carried out and the

results are compared with each other. The researchers are concluded that the

release rate was more rapid from ethosomal systems than from liposomal

systems. They also concluded that, ethosomes produced by the highest ethanol

concentration released azelaic acid more rapidly, and the same trend was

found using viscous forms.

Verma149 (2011) has developed ethosomal systems containing ascorbic acid to

observe the effect of penetration enhancer in ethosomal formulations and the

study shown that ethosomal preparation containing penetration enhancer

shown better permeation efficiency than the ethosomal preparation alone.

Jadupati Malakar et al150 (2012) have developed transferosomal gel containing

insulin by reverse phase evaporation method for painless insulin delivery to

treat insulin dependent diabetes mellitus. The prepared formulations were


evaluated and optimized one formulation and in vivo study was carried out

with optimized formulation. The in vivo study of optimized transferosomal gel in

alloxan-induced diabetic rat has demonstrated prolonged hypoglycemic effect

in diabetic rats over 24 h after transdermal administration.

Donatella Paolino et al151 (2012) have prepared ultradeformable vesicles, by the

extrusion technique, containing asiaticoside, a natural compound obtained

from Centella asiatica and evaluated for the potentiality of ultradeformable

vesicles as a possible topical delivery system. The results shown that

ultradeformable vesicles provided the greatest in vitro skin permeation of

asiaticoside showing a 10-fold increase with respect to the free drug solution

voured and increase in in vivo collagen biosynthesis and also suggested that

ultradeformable vesicles are suitable carriers for the pharmaceutical and

cosmetic application of the natural agent asiaticoside.

Gregor Cevc et al152 (2002) have developed ultradeformable lipid vesicles to

study the stability of various aggregates in the form of lipid bilayer vesicles was

tested by three different methods before and after crossing different semi-

permeable barriers. The researchers concluded that the ultradeformable

vesicles penetrate the skin intact, that is, without permanent disintegration.

Eun kyung Oha et al153 (2011) have aimed to develop 5-amaino levulinic acid

(5-ALA) loaded ultradeformable liposomes (UDL) with different surface charges,

and to investigate their physicochemical characteristics and capability for the

skin penetration and retention of 5-ALA for topical photodynamic therapy

(PDT). The effects of surface charges of UDL on in vitro permeation of 5-ALA


and in vivo accumulation of 5-ALA_induced PpIX in viable skin were

determined and then compared with conventional neutral liposomes

(nLiposome). All UDL showed smaller particle size and better deformability

than nLiposome. However, entrapment efficiency of 5-ALA was similar to each

vesicle. Among vesicles, the cationic UDL (cUDL) demonstrated higher stability

and permeability, and could deliver 5-ALA into deep skin tissue by topical

application.

Jorge Montanari et al154 (2010) have developed with an aim independent of

artificial power sources, self administered sunlight triggered photodynamic

therapyncould be a suitable alternative treatment for cutaneous leishmaniasis,

that avoids the need for injectables and the toxic side effects of pentavalent

antimonials and they have determined the in vitro leishmanicidal activity of

sunlight triggered photodynamic ultradeformable liposomes (UDL).

B. Geusens et al155 (2009) have developed Ultradeformable cationic liposomes

for delivery of small interfering RNA (siRNA) into human primary melanocytes

and also to block the expression of a specific Myosin Va exon F containing

isoform that is physiologically involved in melanosome transport in human

melanocytes and characterized for the same. The best results were obtained

with UCLs which are promising for future in vivo experiments.

Gregor Cevc et al156 (2001) have developed and evaluated the diclofenac loaded

ultrdeformable liposomes and proved that the these vesicles are prominent tool

for topical and transdermal delivery of active principles.


Ebtessam A. Essa et al157 (2003) have developed a neutral lipophilic compound

(estradiol) enclosed transferesomes and studied the effect of electroporation on

human epidermal penetration of a model neutral lipophilic compound

(estradiol) from saturated aqueous solution and when encapsulated in

ultradeformable liposomes and Total amount penetrated and skin deposition

were compared with values obtained from passive diffusion. The results shown

that the effect of electrical pulsing on liposome size was investigated. The

action of phosphatidylcholine on skin that was structurally altered by such

pulses was determined. Electroporation did not affect liposome size. Skin

pulsing considerably increased estradiol penetration and skin deposition from

solution, relative to passive delivery, with subsequent partial recovery of skin

resistance to molecular penetration. Surprisingly, with liposomes,

electroporation did not markedly affect estradiol skin penetration. Importantly,

liposomal phosphatidylcholine applied during or after pulsing accelerated skin

barrier repair, i.e. provided an anti-enhancer or retardant effect.

Gregor Cevc et al158 (2003) have developed novel formulations of the

halogenated corticosteroid, triamcinolone-acetonide, based on ultradeformable

mixed lipid vesicles, Transfersomes, and their performance was tested in vivo

using radioactive label measurements, to study the drug biodistribution, and

murine ear edema, to determine the drug bioactivity. Sparse use of drug-loaded

TransfersomesR on the skin ensures an almost exclusive delivery of

triamcinolone-acetonide into the organ, thus arguably increasing the treatment


safety. The researchers concluded based on the results transferosomes are

efficient carriers in transportation of drugs across the skin.

P. Loan Honeywell-Nguyen et al159 (2003) developed and several aspects of

elastic vesicle transport into human skin were investigated in vivo. Surfactant

based elastic vesicles were applied onto human skin in vivo and subsequently a

series of tape-strippings were performed, which were visualised by freeze

fracture electron microscopy. Factors of investigation for non-occlusive

treatment were the duration of application and the volume of application. In

addition, occlusive vs. non-occlusive application was studied. The results have

shown a fast penetration of intact elastic vesicles into the stratum corneum

after non-occlusive treatment, frequently via channel-like regions. A higher

volume of application resulted in an increase in the presence of vesicle material

found in the deeper layers of the stratum corneum. Micrographs after occlusive

treatment revealed very few intact vesicles in the deeper layers of the stratum

corneum, but the presence of lipid plaques was frequently observed. They have

also proposed a hypothesis that the channel-like regions represent

imperfections within the intercellular lipid lamellae in areas with highly

undulating cornified envelopes.

Gregor Cevc et al160 (1998) developed transferosomes containing insulin with an

aim to deliver the insulin through intact skin and evaluated for the same. The

researchers concluded that this process can be nearly as efficient as an

injection needle, as seen from the results of experiments in mice and humans

with the insulin-carrying vesicles and also said that the carrier-mediated
transcutaneous insulin delivery is unlikely to involve shunts, lesions or other

types of skin damage. Rather than this, insulin is inferred to be transported

into the body between the intact skin cells with a bio-efficiency of at least 50%

of the s.c. dose action.

Ramesh R. Boinpally et al161 (2003) developed lecithin vesicles containing

diclofenac, anti-inflammatory agent, to treat rheumatic diseases and the

permeation of diclofenac across human cadaver epidermis in-vitro from four

lecithin vesicle formulations and a few marketed semi-solid preparations and

they concluded that the lecithin vesicles of diclofenac appear to be

advantageous for the topical delivery of diclofenac based on the results.

Yuka Hiruta et al162 (2006) have incorporated Beta-sitosterol 3-β-D-glucoside

(Sit-G), an absorption enhancer, into ultra-deformable vesicles containing

bleomycin to attenuate drug toxicity in human keratinocytes and evaluated.

Based on the results researchers concluded that the presence of Sit-G

increased drug entrapment and improved in vitro stability of ultradeformable

vesicles. They also concluded that the Ultra-deformable formulation contained

Sit-G maintained flexibility for penetration through the skin, increased

entrapment efficiency of bleomycin and stability in vitro, and significantly

increased distribution of bleomycin in epidermis and dermis compared with

those without Sit-G.

P. Loan Honeywell-Nguyen et al163 (2003) have developed pergolide from L-595–

PEG-8-L elastic vesicle formulations and studied the in vitro transport of the

same across the human skin using flow-through Franz diffusion cells to
elucidate the transport mechanism. The findings show that there was a strong

correlation between the drug incorporation to saturated levels and the drug

transport, both of which were influenced by the pH of the drug–vesicular

system. The optimal pH was found to be 5.0, giving the highest drug

incorporation as well as the highest drug transport. Non-occlusive co-treatment

with elastic vesicles improved the skin delivery of pergolide compared to the

non-occlusive buffer control by more than 2-fold. Occlusion reduces the

actions of elastic vesicles, but could increase the pergolide transport since

water is a good penetration enhancer for this particular drug. Based on the

results obtained, a mechanism of action for the elastic vesicles was proposed.

Ajay Kumar et al164 (2012) have made review on transferosomes and they

suggested that Transferosomes are novel vesicular systems that are several

times more elastic than other vesicular systems. They also given information

like regulatory aspects of excipients used in preparation, summary of clinical

investigations performed, marketed preparations available, research reports

and patent reports related to transfersomes.

Tomonobu Uchino et al165 (2013) developed vesicular carrier system enclosed

ketoprofen to study the skin permeation of ketoprofen with vesicles and

characterized for the same and also studied the effect of elasticity of the vesicle

in permeation of the enclosed drug. The researchers concluded based on the

results that the Surfactant-based vesicles enhance ketoprofen transport across

human skin, while no enhancement of ketoprofen was observed when loaded in

elastic liposomes.
Sureewan Duangjit et al166 (2011) have developed lipid vesicles viz, liposomes

and transferosomes containing meloxicum with an aim to study potential use

of liposome and transfersome vesicles in the transdermal drug delivery of

meloxicam (MX) and evaluated for the same. The results suggested that MX-

loaded transfersomes can be potentially used as a transdermal drug delivery

system rather than the liposomes.

Alpana Ram et al167 (2012) developed transferosomes containing ibuprofen, an

anti-inflammatory agent and evaluated the ability of the vesicles in permeation

of the same. The study revealed that transfersomes are a promising prolonged

delivery system for Ibuprofen and have reasonably good stability characteristics

and the researchers concluded that transferosomes are prominent tool for

transportation of ibuprofen across the skin.


Chapter - 3

THEORITICAL ANALYSIS
Chapter - 3: Theoritical Analysis

Table of contents

S.No Name of the subtitle Page No


3.1 Drug and excipient profiles  
3.2 Objective of the study  
3.3 Schematic presentation of the research work  
3.4 parameters to be characterized  
3.5 Materials  

3. THEORETICAL ANALYSIS
3.1. DRUG AND EXCIPIENTS PROFILE

3.1.1. NYSTATIN

Nystatin168 is a polyene antifungal drug to which many molds and

yeasts are sensitive, including Candida spp. Nystatin has some toxicity

associated with it when given intravenously, but it is not absorbed across

intact skin or mucous membranes. It is considered a relatively safe drug

for treating oral or gastrointestinal fungal infections.

Chemical Name: (1S,3R,4R,7R,9R,11R,15S,16R,17R,18S,19E,21E, 25E,

27E, 29E, 31E, 33R, 35S, 36R, 37S)-33-[(3-amino-3, 6-dideoxy-β-L-

mannopyranosyl)oxy]-1, 3, 4, 7, 9, 11, 17,37-octahydroxy-15, 16,18-

trimethyl-13-oxo-14, 39 dioxabicyclo [33.3.1] nonatriaconta-19,

21,25,27,29,31-hexaene-36-carboxylic acid.

Chemical structure:

Molecular formula: C47H75NO17 

Molecular weight: 926.09

Colour : Yellow to light tan powder


Odour: cereallike odor

Solubility: very slightly soluble in water (4mg/ml at 28oC & 360mg/L at

24oC), and slightly to sparingly soluble in alcohol.

Pharmacology:

Pharmacokinetics:

a) Bioavailability: 0% on oral ingestion

b) Metabolism: None (not extensively absorbed).

c) Half-life: Dependent upon GI transit time

d) Excretion: Faecal- 100 %.

Pharmacodynamics:

Nystatin is an antibiotic which is both fungistatic and fungicidal in vitro

against a wide variety of yeasts and yeast-like fungi, including Candida

albicans, C. parapsilosis, C. tropicalis, C. guilliermondi, C. pseudotropicalis, C.

krusei, Torulopsis glabrata, Tricophyton rubrum, T. mentagrophytes. Nystatin

acts by binding to sterols in the cell membrane of susceptible species resulting

in a change in membrane permeability and the subsequent leakage of

intracellular components. On repeated subculturing with increasing levels of

nystatin, Candida albicans does not develop resistance to nystatin. Generally,

resistance to nystatin does not develop during therapy. However, other species

of Candida (C. tropicalis, C. guilliermondi, C. krusei, and C. stellatoides) become

quite resistant on treatment with nystatin and simultaneously become cross

resistant to amphotericin as well. This resistance is lost when the antibiotic is


removed. Nystatin exhibits no appreciable activity against bacteria, protozoa, or

viruses.

Mechanism of action

Nystatin binds to ergosterol, a major component of the fungal cell

membrane. When present in sufficient concentrations, it forms pores in the

membrane that lead to K+ leakage, acidification, and death of the fungus. [7]

Ergosterol, is unique to fungi, so the drug does not have such catastrophic

effects on animals or plants. However, many of the systemic/toxic effects of

Nystatin are attributable to its effect on human cells via binding to mammalian

sterols, namely cholesterol. This is the effect that accounts for the

nephrotoxicity observed when high serum levels of Nystatin are achieved.

Uses:

1. It is used to treat vaginal, Cutaneous, mucosal and esophageal Candida

infections.

2. Oral nystatin is often used as a preventative treatment in people who are

at risk for fungal infections, such as AIDS patients with a low CD4 +

count and patients receiving chemotherapy.

3. It has been investigated for use in patients after liver transplantation

but fluconazole was found to be much more effective for preventing

colonization, invasive infection and death.

4. It is also used prophylactically in Very Low Birth Weight (<1500 g)

infants to prevent invasive fungal infections.


5. Liposomal Nystatinis not currently available but investigational use has

shown greater in vitro activity than colloidal formulations of

Amphotericin B.

6. It offers an intriguing possibilty for difficult to treat systemic infections,

such as invasive apergillosis, or infections that demonstrate resistance to

Amphotericin B. Additionally, liposomal Nystatin appears to cause fewer

cases of and less severe nephrotoxicity than observed with Amphotericin

B. However, at the time of this writing, plans for additional development

and investigation of liposomal nystatin appear in flux.

7. Cryptococcus is also sensitive to nystatin.

8. It is also used in cellular biology as an inhibitor of the lipid raft-caveolae

endocytosis pathway on mammalian cells, at concentrations around

3 µg/mL.

Adverse effects

The oral suspension form produces a number of adverse effects including

but not limited to

1. Diarrhea

2. Abdominal pain

3. Rarely, tachycardia, bronchospasm, facial swelling, muscle aches

Both the oral suspension and the topical form can cause

1. Hypersensitivity Reactions, including Stevens Johnsons Syndrome in

some cases

2. Rash, itching, burning.


3.1.2. CHOLSTEROL

Empirical Formula : C27H46O

Chemical structure :

Molecular Weight : 386.67

Synonyms : Cholesterin, cholesterolum.

Chemical Name : Cholest-5en-3β-ol.

Description : Cholesterol occurs as white or faintly yellow, almost

less, pearly leaflets needles, powder or granules, on prolonged exposure to light

and air cholesterol acquires a yellow to tan colour.

Boiling point : 360°C

Density : 1.052 g/cm3

Melting Point : 147-150°C.

Solubility : Chloroform 1 in 4.5; Ether 1 in 2.8 and

practically insoluble in water.


Applications : Emollient, Emulsifying agent. Cholesterol

is used in cosmetics and topical pharmaceuticals and at concentrations of

0.30%w/w imparts water absorbing power to an ointment and also has

emollient activity.

Method of manufacture:

The commercial material is normally obtained from the spinal cord of

cattle by extraction with petroleum ethers, but it may also be obtained from

wool fat. Purification is normally accomplished by repeated bromination.

Cholesterol may also be produced by entirely synthetic means.Cholesterol

produced from animal organs will always contain cholestanol and other

saturated sterols.

Safety:

Cholesterol is generally regarded as an essentially nontoxic and

nonirritant material at the levels employed as an excipient(18).It has, however,

exhibited experimental teratogenic and reproductive effects, and mutation data

have been reported. Cholesterol is often derived from animal sources and this

must be done in accordance with the regulations for human consumption. The

risk of bovine spongiform encephalopathy (BSE) contamination has caused

some concern over the use of animal-derived cholesterol in pharmaceutical

products. However, synthetic methods of cholesterol manufacture have been

developed.

Incompatibilities: Cholesterol is precipitated by digitonin.

Handling Precautions:
Observe normal precautions appropriate to the circumstances and

quantity of material handled. Rubber or plastic gloves, eye protection and a

respirator are recommended.

Regulatory status:

It is included in the FDA Inactive Ingredients Guide (injections,

ophthalmic, topical, and vaginal preparations). It is included in nonparenteral

medicines licensed in the UK andin the Canadian List of Acceptable Non-

medicinal Ingredients.
3.1.3. SPAN – 60

Empirical Formula : C24H46O6.

Chemical structure :

Molecular Weight : 431

Synonyms : Ablunol S-60, 6 - Octa deconoate

Arlaces60, Sorbitan Stearate, Spans 60.

Functional category: Emulsifying agent; nonionic surfactant; solubilizing

agent; wetting and dispersing/suspending agent.

Applications: In pharmaceutical formulation or technology Sorbitan

monoesters are a series of mixtures of partial esters of sorbitol and its mono-

and dianhydrides with fatty acids. Sorbitan diesters are a series of mixtures of

partial esters of sorbitol and its monoanhydride with fatty acids. Sorbitan

esters are widely used in cosmetics, food products, and pharmaceutical

formulations as lipophilic nonionic surfactants. They are mainly used in

pharmaceutical formulations as emulsifying agents in the preparation of

creams, emulsions, and ointments for topical application. When used alone,

sorbitan esters produce stable water-in-oil emulsions and microemulsions but

are frequently used in combination with varying proportions of a polysorbate to

produce water-in-oil or oil-in-water emulsions or creams of varying

consistencies.Sorbitan monolaurate, sorbitan monopalmitate and sorbitan


trioleate have also been used at concentrations of 0.01–0.05% w/v in the

preparation of an emulsion for intramuscular administration.

Description : Cream Solid

Typical properties

Acid value : 5-10

Flash point : >149°C

HLB value : 4.7

Hydroxyl value : 235-260

Iodine number : ≤1

Melting point : 53-57

Moisture content : ≤1

Saponification value : 147-157

Solubility: sorbitan esters are generally soluble or dispersible in oils; they are

also soluble in most organic solvents. In water, although insoluble, they are

generally dispersible.

Surface tension of 1% aqueous solution: 46mN/m

Stability and storage conditions: Gradual soap formation occurs with strong

acids or bases; sorbitan esters are stable in weak acids or bases. Sorbitan

esters should be stored in a well-closed container in a cool, dry place.

Safety: Sorbitan esters are widely used in cosmetics, food products, and oral

and topical pharmaceutical formulations and are generally regarded as

nontoxic and nonirritant materials. However, there have been occasional

reports of hypersensitive skin reactions following the topical application of


products containing sorbitan esters. When heated to decomposition, the

sorbitan esters emit acrid smoke and irritating fumes. The WHO has set an

estimated acceptable daily intake of sorbitan monopalmitate, monostearate,

and tristearate, and of sorbitan monolaurate and monooleate at up to 25mg/kg

body-weight calculated as total sorbitan esters.

LD50 (rat, oral): 31g/kg. Very mildly toxic by ingestion.

Handling precautions: Observe normal precautions appropriate to the

circumstances and quantity of material handled. Eye protection and gloves are

recommended.

Regulatory status: Certain sorbitan esters are accepted as food additives in

the UK. Sorbitan esters are included in the FDA Inactive Ingredients Guide

(inhalations; IM injections; ophthalmic, oral, topical, and vaginal preparations).

Sorbitan esters are used in nonparenteral medicines licensed in the UK.

Sorbitan esters are included in the Canadian List of Acceptable Non-medicinal

Ingredients.
3.1.4. SPAN – 80

Chemical formula : C24H44O6

Chemical structure:

Synonyms : Sorbitan oleate; Sorbitan (Z)-mono-9-

octadecenoate

Molecular weight : 428

Physical properties

Density : 0.986

Refractive Index : 1.48

Flash point : > 149o C

Stability : Stable, Combustible, Incompatible with

strong oxidizing Agents

Description : Liquid

Colour : Yellow

Odour : Fatty acid-like

Viscosity : Dynamic (25 °C) 1000 mPa*s

Boiling point : > 100 °C

Vapour pressure (20 °C) : < 1.4 hPa


Solubility : In water (20 °C), insoluble ethylacetate (20

°C), soluble acetone (20 °C).

HLB : 4.3

Storage : Store in dark. Kept under seal.

Handling : Prevention of sunlight, drench, heating

and bump, treat lightly and forbid mixed package and transport with

deleterious goods or other contaminated cargo.

Applications : Use as a w/o emulsifier or as an o/w

emulsifier for use in cosmetic formulations, oil field chemicals, plastics,

household products, coatings and textiles.


3.1.5. PHOSPHOLIPON-90H

Synonym : Hydrogenated phosphatidylcholine

Composition : Phosphatidylcholine (hydrogenated)

[g/100 g] n.l.t. 90.0

Structure formula :

Molecular formula : C43H95NO8P

Molecular weight : Average M. Wt 784.6 g/mol

Phase transition temperature : Approx. 55°C (in hydrated form)

Purity : Lysophosphatidylcholine [g/100 g]

NMT 4.0

Non-polar lipids : Triglycerides [g/100 g] NMT 2.0

Typical fatty acid composition : Σ (C16:0, C18:0) [g/100 g]n.l.t. 98

Iodine value : NMT 1


Water : [g/100 g] NMT 2.0

Residual solvents

Ethanol : [g/100 g] NMT 0.5

Physical and chemical Properties

Colour : White to whitish

Consistency : Powder

Odor : Odorless

Solubility (5% solution):

Ethanol : Soluble at 50 °C

Propylene glycol : Soluble 55 °C

Water : Dispersible 55 °C

Beeswax : Soluble 80 °C

MCT : Soluble 90 °C

Macadamia nut oil : Soluble 97 °C

Bulk density : 400–500 kg/m3

pH : 6 ± 1 at 10 g/l (20°C)

Minimum ignition energy : 3 mJ < MIE < 10 mJ

Specific resistance : 4, 32 x 10-11 Ωm

Bacteriological Data:

Total aerobic microbial count (TAMC)[/g]: NMT 100

Total combined yeasts & moulds count (TYMC) [/g]: NMT 10

E. Coli [/g] : Negative

Staphylococcus aureus [/g] : Negative


Pseudomonas aeruginosa [/g] : Negative

Packaging : 5 kg and 25 kg standard packaging

in double PE bag/aluminium foil bag

Storage

Stored in closed containers at +5 ± 3 °C. To avoid a negative impact on

the product quality by humidity, a cooled product unit must not be opened

without prior conditioning to ambient temperatures. Close opened containers

immediately.

Applications:

- Preparation of liposomes and emulsions for pharmaceuticals and

cosmetics

- Skin protectant
3.2. Objective of the study

From ancient days the search for new things, which will improve

comfortability, is going till today and will be there in future also to achieve

more and more comfortability and also there will not be end for the same.

Similarly there is lot of change has been taken place in the field of

pharmaceutical sciences also, when compared the current generation with

ancient days. Since beginning researchers trying to introduce new alternative

drug molecules and/or techniques to achieve better therapeutic effect, to

increase patient compliance, etc.

Generally researchers will have two options viz either introduction of new

molecules or using of existing molecules in better way to achieve the desired

objectives. The first one involves huge money, time, etc hence it is difficult to

follow the first option. The other option is modifying the existing drug

molecules to meet the objectives which less difficult rather the first option and

is preferable option. With the same intention researchers were adopted

different techniques to achieve the desired objectives. The different techniques

include modification of existing drug molecule, change in the route of

administration, change in the dosage form, change in drug delivery systems,

etc.

The modified drug delivery systems are classified under the class of new

drug delivery systems (NDDS). The ideal NDDS should fulfill two important

requirements. They are 1. They should capable to deliver the drug at required
concentration for required period of time. 2. These should deliver the drug to

the site of action. No NDDS were introduced till today which can were fulfill the

two requirements. To achieve the same different carrier systems were

introduced by the researchers which include cellular carrier, particulate,

macromolecular and polymeric carriers. Particulate type carrier includes lipid

particles (low and high density lipoprotein-LDL and HDL, respectively),

microspheres, nanoparticles, polymeric micelles and vesicular carriers like

liposomes, niosomes pharmacosomes, virosomes, etc.

Last three decades onwards the interest on the vesicular carrier systems

tremendously increased and huge research is carrying out as these carriers

has shown solution for many challenges like enhancing the solubility,

controlled drug release, targeting the abnormal sites, permeation of drugs

across the biological membranes, etc. These vesicular carrier systems are

highly ordered assemblies of one or several concentric lipid bilayers formed

when certain amphiphilic building blocks are confronted with water. Vesicles

can be manufactured using a diverse range of amphiphilic

molecules.Researchers developed variety of vesicles includes liposomes,

niosomes, ethosomes, transferosomes, aquasomes, pharmacosomes, etc for

different purposes. Here the primary aim of the work is to select the best

vesicular carrier system, among niosomes, ethosomes and transferosomes,

which shows enhanced transdermal delivery of active principles. The secondary

objective the research work is to enclose the nystatin, polyene antifungal

antibiotic, in the best vesicular carrier system and to characterize the same.
3.2.1. Particular goals were:

 Development of niosomal suspension using two different non-ionic

surfactants viz. span-60 and span-80 at different concentrations.

 Characterization of prepared niosomal suspensions and selection of best

formulation.

 Preparation of nystatin niosomal gel with optimized formulation and

carrying out ex vivo drug release study, stability, etc.

 Development of ethosomal suspension using different concentrations of

ethanol and lipid.

 Characterization of prepared ethosomal suspensions and selection of

best formulation.

 Preparation of nystatin ethosomal gel with optimized formulation and

carrying out ex vivo drug release study, stability, etc.

 Development of transferosomal suspension using different

concentrations of edge activators and lipid.

 Characterization of prepared transferosomal suspensions and selection

of best formulation.

 Preparation of nystatin transferosomal gel with optimized formulation

and carrying out ex vivo drug release study, stability, etc.

 Carrying out of in vivo study with optimized niosomal, ethosomal and

transferosomal gel and to determine the pharmacokinetic parameters.


 Concluding the best vesicle for enhanced transdermal drug delivery

based on the in vivo data.


3.3. SCHEMATIC PRESENTATION OF THE RESEARCH WORK
Preformulation studies

Niosomal formulations Ethosomal formulations Transferosomal formulations

Characterization Characterization Characterization


1. Vesicles morphology
2. Entrapment efficiency
3. In vitro drug release (Dialysis membrane)
4. Release kinetics

Selection of the Selection of the Selection of the


best formulation best formulation best formulation
1. Elasticity measurements.
2. Zetapotential

Preparation of gel Preparation of gel Preparation of gel


1. In vitro drug release (Dialysis membrane)
2. Ex vivo drug release (Rat skin)
3. Skin retention study
4. Stability study
5. In vivo performance

Conclusion of the best vesicular system


3.4. PARAMETERS TO BE CHARACTERIZED

1. Vesicles morphology (size, shape and surface appearance)

2. Entrapment efficiency

i. Dialysis method

ii. Ultra centrifugation method

3. In vitro drug release (Dialysis membrane)

4. Release kinetics

a. Zero order

b. First order

c. Higuchi model

d. Hixon-crowel model

e. Korsmeyer-peppas model

5. Elasticity measurements.

6. Zetapotential

7. Invitro drug release (Dialysis membrane)

8. Ex vivo drug release (Rat skin)

a. Transdermal flux.

b. Permeability co-efficient.

c. Enhancement ratio.

9. Drug retention study

10. Stability studies.

11. In vivo performance.

a. Pharmacokinetic parameters like Cmax, Tmax, AUC, etc


3.5. Materials

Table 3.5.1: List of chemicals

Sr.No. Chemical Name Grade Manufacturer

1 Phospolipon-90H HPLC Merck Ltd, Mumbai.

2 Ethanol HPLC Merck Ltd., Mumbai.

3 Hydrochloric acid AR Merck Ltd, Mumbai.

4 Dichloromethane AR Merck Ltd, Mumbai.

5 Span 80 AR Merck Ltd, Mumbai.

6 Span 60 AR Merck Ltd, Mumbai.

7 Cholesterol AR Merck Ltd, Mumbai.

8 Heavy liquid paraffin AR Merck Ltd., Mumbai.

9 Diethyl ether AR Merck Ltd., Mumbai.

10 Sodium bicarbonate AR Merck Ltd, Mumbai.

11 Ortho phosphoric acid HPLC Merck Ltd., Mumbai.

12 Anesthetic ether AR Sigma Aldrich, Mumbai

13 EDTA AR Merck Ltd., Mumbai.

14 Carbopol -971 AR Merck Ltd., Mumbai.

15 HPMC AR Merck Ltd., Mumbai.

16 Propylene glycol AR Merck Ltd., Mumbai.

17 Triethanolamine AR Merck Ltd., Mumbai.

18 Methanol HPLC Merck Ltd., Mumbai.


Table 3.5.2: LIST OF EQUIPMENTS

S. No Name of the Equipment Manufacturer

1 Digital Electronic balance Metler, Japan.

2 Roto vacuum evaporator Kshitij Innovations.

3 Magnetic stirrers Remi.

4 Refrigerated centrifuge Remi.

5 Magnetic beads Sunsil.

6 Dialysis membrane Himedia

7 Hot Air Oven Minicon

8 UV Spectrophotometer PG Instruments

9 HPLC Waters

10 Vacuum pump Kent Pvt Ltd

11 Filtration assembly Kshitij Innovations

12 Micro pippets Micro tech

13 Bath sonicator Kshitij Innovations


Chapter - 4

EXPERIMENTAL
INVESTIGATIONS
Chapter - 4: Experimental Investigations

Table of contents

S.No Title of the content Page No


4.1 Preformulation Studies  
4.2 Standard curve of nystatin in phosphate buffer saline pH 7.4  
4.3 Formulation of nystatin loaded niosomes  
4.4 Formulation of nystatin loaded ethosomes  
4.5 Preparation of nystatin loaded transferosomes.  
4.6 Characterizations  

4. EXPERIMENTAL INVESTIGATIONS
4.1. Preformulation Studies

4.1.1. Physico-chemical characterization of API

Preformulation study is the fundamental step in the development of new

formulations which involves characterization of different physic-chemical

parameters which were directly influence on the development of formulations.

In this study various physico-chemical and mechanical properties of drug and

excipients to be characterized so as to develop an effective dosage from. A

thorough understanding of the properties may ultimately provide a rational for

formulation design.

4.1.2. Identification of drug sample

The Nystatin drug, received as gift sample, was characterized and

confirmed by studying its physicochemical parameters like appearance,

solubility, absorption maximum and FTIR study. During the identification

process the drug sample was used without further purification.

4.1.3. Description

The sample of drug sample was studied for organoleptic characters such as

appearance, colour, odour and solubility. The drug sample was appeared as

yellow coloured powder with cereal like odor.

The solubility study and determination of absorption maximum (λ max) of

the drug sample was studied in phosphate buffer saline pH 7.4. The study was

carried out as follows.

4.1.4. Preparation of phosphate buffer saline pH 7.4


Accurately, 1.564gm of sodium hydroxide and 6.804gm potassium

dihydrogen phosphate was weighed and are solubilized in small volume of

distilled water. After complete solubilization of both the ingredients, the volume

was adjusted to 1000ml with distilled water. The resulting solution pH was

measured and adjusted to pH 7.4, if necessary.

4.1.5. Determination of absorption maximum (λmax) of drug sample

A stock solution of drug sample was prepared by dissolving 20mg of drug

sample in 100ml of phosphate buffer saline, pH.7.4. From the above stock

solution, 2µg/ml solution was prepared by serial dilution. The prepared

solution absorbance was recorded between the wavelengths 200-400nm by UV-

Visible spectrophotometer. From the absorbance data, wavelength at which

maximum absorbance was exhibited by the drug sample was observed. The

obesreved wavelength was considered as absorption maximum and was found

at 306nm. The absorption spectrum of given drug sample was given in figure

5.1.

4.1.6. Solubility

The solubility of drug sample was determined by adding excess amount

(100mg) of drug in the deaerated phosphate buffer saline (PBS) pH 7.4 and

kept aside for 12 hrs to acieve equilibrium with occasional shaking at room

temperature. From the above super saturated solution, 10ml of supernatant

liquid was collected and centrifuged at 3000RPM for 10min. After

centrifugation, 1ml of supernatant was transferred into the 100ml volumetric

flask and volume adjusted to 100ml with PBS pH7.4. The flask was shaken
well and the resulting solution absorbance was measured using UV-Visible

Spectrophotometer at 306nm. Based on the absorbance, the solubility of the

drug sample in PBS pH7.4 was calculated and tabulated in table 5.1.

4.1.7. FTIR study of the drug sample

FTIR spectrum of gift sample was determined to find out the

characteristic peaks for lactone carbonyl, N-H, C-OH, O-H bond stretching and

Carboxylate ion. The FTIR spectra of drug sample has given in figure 5.2.

Based on the results of the solubility, absorption maximum and FTIR

studies, the drug sample was confirmed as Nystatin.

4.2. Standard curve of nystatin in phosphate buffer saline pH 7.4

A stock solution of nystatin was prepared by dissolving 20 mg of drug in

100 ml of phosphate buffer saline pH 7.4. From the above stock solution,

solutions having concentrations of 2, 4, 6, 8 and 10µg/ml were prepared by

serial dilutions. The resultant solutions absorbances were recorded by UV –

Visible spectrophotometer at 306 nm. The absorbances of the solutions

tabulated in table 5.2 and the tandard curve has given in figure 5.3.

4.3. Formulation of nystatin loaded niosomes

Niosomal suspensions were prepared by the thin-film hydration method.

Accurately weighed quantities of drug, surfactant (Span – 60 & Span - 80), and

Cholesterol were dissolved in chloroform in a round-bottom flask. The

chloroform was evaporated at 60°C under reduced pressure using a rotary

flash evaporator (Buchi type). After chloroform evaporation, the flask was kept

under vacuum overnight to remove residual solvent. The thin film was
hydrated with 10 ml of phosphate buffer (PBS), pH 7.4, and the flask was kept

rotating at 60°C. Formulations were sonicated three times in a bath-sonicator

for 15 min with 5-min interval between successive times. Composition of all

niosomal suspensions was given in table 5.3.

4.4. Formulation of nystatin loaded ethosomes

Ethosomal suspensions were developed according to the method reported

by Touitou. Phospholipon and drug were dissolved in ethanol and the mixture

was heated to 30±1ºC. Double distilled water (consisting of dissolved

cholesterol) was heated to 30ºC±1ºC and was added slowly as a fine stream to

the lipid mixture with constant stirring (Mechanical stirrer, Remi equipment,

Mumbai) at 700rpm. The entire process was carried out in a closed vessel to

prevent the evaporation of ethanol at a temperature of 30ºC±1ºC. The stirring

was continued for 5 more minutes after addition of aqueous solution to lipid

mixture which results in the formation of ethosomal suspension. Composition

of all ethosomal suspensions was given in table 5.4.

4.5. Preparation of nystatin loaded transferosomes.

All transferosomal suspensions contanining nystatin were prepared by

the thin-film hydration method. Accurately weighed quantities of drug,

surfactant (Span – 60 & Span - 80), and lecithin were dissolved in chloroform

in a round-bottom flask. The chloroform was evaporated at 40°C under

reduced pressure using a rotary flash evaporator (Buchi type). After chloroform

evaporation, the flask was kept under vacuum overnight to remove residual

solvent. The thin film was hydrated with 10 ml of phosphate buffer saline
(PBS), pH 7.4, and the flask was kept rotating at 40°C. All the formulations

were sonicated three times in a bath-sonicator for 15 min with 5-min interval

between successive times. Composition of all transferosomal suspensions was

tabulated in table 5.5.

4.6. CHARACTERIZATION

4.6.1. Vesicle Morphology

Morphology (size, shape and surface) of the vesicles present in all

vesicular suspensions were characterized. The shape and surface of the

vesicles was observed by means of Scanning Electron Microscopy (SEM) and

the size of the vesicles was measured using optical microscope. The SEM

photographs of niosomes, ethosomes and transferosomes are given figures 5.4,

5.15 and 5.27 respectively. The vesicles present in all niosomal, ethosomal and

transferosomal formulations were tabulated in tables 5.6, 5.7 and 5.8

respectively.

4.6.2. Entrapment Efficiency

Entrapment efficiency of all vesicular suspensions was determined by

separating the unentrapped drug. Un-entrapped drug was separated out by

ultracentrifugation method.

Vesicular suspension was suitably diluted with Phosphate buffer saline

solution and centrifuged at 12000rpm for 20min at 8 oC. The supernatant liquid

was collected and analyzed for concentration of an un-entrapped drug by UV

spectrophotometer (PG instruments) at 306nm. Entrapment efficiency of all

vesicular suspensions was determined by using formulas given below. The


results were confirmed by dialysis method also. Entrapment efficiency of all

vesicular suspensions was tabulated in tables 5.6, 5.7 and 5.8.

4.6.3. In vitro Release Studies

In vitro drug release from vesicular suspensions was studied by

exhaustive dialysis method and was carried out in an artificial diffusion cell.

Two side open ended glass tube was taken and one side has been closed

with Dialysis membrane (HiMedia Laboratories Pvt. Ltd., M.Wt cut off: 12000).

The fabricated tube was used as donor compartment, in which 1ml of vesicular

suspension was taken and the entire set up placed in receptor compartment

(Beaker, 250ml capacity) containing 100 ml phosphate buffer saline. The

dialysis was carried out at 50 rpm at 37±0.5°C for 12hrs. Every hour 2ml of

diffusion fluid from receptor compartment was withdrawn and same volume of

fresh diffusion fluid was replaced to maintain sinc conditions. The withdrawn

diffusion fluids were suitably diluted and analyzed using UV spectrophotometer

at 306nm. The in vitro drug releases from all niosomal, ethosomal and

transferosomal formulations was tabulated in tables 5.9-5.17, 5.19-5.27 and

5.29-5.37 respectively and the same was graphically presented in figutes 5.5-

5.6, 5.16-5.18 and 5.28-5.29 respectively.


Specifications of the in vitro drug release study

Equipment : Artificial diffusion cell

Semipermeable membrane : Dialysis membrane (M.Wt cut off: 12000)

Diffusion medium : Phosphate buffer saline (PBS) pH 7.4

Volume of sampling fluid : 2ml

Temperature : 37±0.5°C

RPM : 50

Absorption maximum : 306nm

4.6.4. Kinetic modelling of drug release:

The order and mechanism of Nystatin release from all vesicular

suspensions was determined by fitting the in vitro drug release data in the

following mathematical models. The drug release kinetics was evaluated by

using the linear regression method.

1. Zero-order kinetics (cumulative % release vs time),

2. First-order kinetics (log % drug remaining vs time),

3. Higuchi kinetics (cumulative % drug release vs. square root of time),

4. Korsmeyer - Peppas (log cumulative % drug release vs log time) and

5. Hixson-Crowel models (cubic root of drug remaining vs time).

The r2 and n values are calculated for the linear curves obtained by

regression analysis of the above plots. The release pattern from all vesicular

suspensions was concluded based on the r 2 and n – values. The r2 and n –

values of all niosomal, ethosomal and transferosomal suspensions was

tabulated in tables 5.18, 5.28 and 5.38 respectively. Release kinetics of


niosomal, ethosomal and transferosomal suspensions are graphically presented

in figures 5.7-5.14, 5.19-5.26 and 5.30-5.37 respectively.

Based on the vesicle size, entrapment efficiency and in vitro drug release

profile from all vesicular suspensions, one vesicular suspension was selected

as an optimized formulation from each type vesicular carrier systems.

Elasticity of the vesicles membranes and surface charge of the vesicles

present in the three optimized suspensions (each from one type vesicular

suspension) was characterized as they are important factors which influence

on the drug permeability across the semipermeable membrane and physical

stability of the vesicles respectively.

4.6.5. Elasticity measurement

Three optimized vesicular suspensions were allowed to pass through the

filter paper having pore size of 50nm under vacuum conditions for 5min. After

5min, the volume of suspension permeated and the vesicles size, present in the

permeated suspension, was measured. Then elasticity of the vesicles was

calculated using the following formula. Elasticity study results of all three

optimized suspensions were tabulated in table 5.39.

Where, E – Elasticity of vesicle membrane; J – Volume of suspension

extruded in 5 min; rv - Vesicle size after extrusion and rp - Pore size of the

barrier.
4.6.6. Zeta potential

Zeta potential of the optimized formulations was measured by

instrument zetasizer nano ZS using DTS software (Malvern Instrument Limited,

UK) using M3-PALS technology. Zeta potential study results of three optimized

vesicular suspensions were tabulated in table 5.40.

4.6.7. Preparation of conventional and vesicular gels

Using raw Nystatin and optimized vesicular suspensions, four gel was

prepared having concentration of 0.01%w/w. for preparation of gels, carbopol-

971 and HPMC was used as gelling agents and are used at a ratio of 2:1. (Total

concentration of gelling agents was maintained as 1.5%w/w). Propylene glycol

was used as co-solvent and as dispersion medium for nystatin. The

performance of the gel, viscosity and physical properties was enhanced by

adding HPMC to carbopol. Neutralization and pH of the gel was adjusted by

adding triethanolamine (quantity sufficient).

Half quantity of the double distilled water was heated to 90 0C and HPMC

was added gradually to the above water with continuous stirring. The

remaining half quantity of water was cooled to 5 0C and this water added to the

HPMC dispersion with continuous stirring. Required quantity of Carbopol

which was passed through sieve no. 40 was weighed and added to the HPMC

dispersion while stirring. The resulting gel pH was adjusted by adding

triethanolamine. Required quantity of Nystatin was dispersed in propylene

glycol and transferred in to the mixture of carbopol and HPMC with continuous

stirring. Composition of all three formulations was given in table 5.41.


4.6.8. In vitro drug permeation from niosomal, ethosomal,

transferosomal and conventional gels

In vitro drug release from vesicular and conventional gels was studied by

exhaustive dialysis method and was carried out in an artificial diffusion cell.

Two side open ended glass tube was taken and one side has been closed

with Dialysis membrane (HiMedia Laboratories Pvt. Ltd., M.Wt cut off: 12000).

The fabricated tube was used as donor compartment, in which 1gm of gel was

taken and the entire set up placed in receptor compartment (Beaker, 250ml

capacity) containing 100 ml phosphate buffer saline. The dialysis was carried

out at 50 rpm at 37±0.5°C for 12hrs. Every hour 2ml of diffusion fluid from

receptor compartment was withdrawn and same volume of fresh diffusion fluid

was replaced to maintain sinc conditions. The withdrawn diffusion fluids were

suitably diluted and analyzed using UV spectrophotometer at 306nm. The in

vitro drug release from all conventional, niosomal, ethosomal and

transferosomal gels was tabulated in tables 5.42. 5.43, 5.44 and 5.45

respectively and the same were graphically presented in figutes 5.41.

Specifications of the in vitro drug release study

Equipment : Artificial diffusion cell

Semipermeable membrane : Dialysis membrane (M.Wt cut off: 12000)

Diffusion medium : Phosphate buffer saline (PBS) pH 7.4

Volume of sampling fluid : 2ml

Temperature : 37±0.5°C

RPM : 50
Absorption maximum : 306nm

4.6.9. Ex vivo drug permeation from the conventional, niosomal,

ethosomal and transferosomal gels

Ex vivo drug permeation studies of all gels were carried out using an

artificial diffusion cell consisting of effective diffusional area of 1.32 Sq.cm and

100 ml of PBS pH 7.4 was placed in receiver compartment as diffusion

medium. Between donor and receiver compartments rat abdominal skin was

mounted and is used as semipermeable membrane.

Dehaired skin of anaesthetized male albino rats weighing between

200 -250 gms was separated from the adhering tissues. Circular section of

suitable diameter was cut to place in the diffusion cell.

The subcutaneous tissue from the skin was removed surgically and the

fat content adhered to the skin was removed by washing with isopropyl alcohol

followed by distilled water.

The prepared abdominal rat skin was arranged between donor and

receptor compartments in such way that the stratum corneum, barrier of the

skin, was faced towards the donor compartment and the dermal side faced

towards the receiver compartment side.

In donor compartment, 1gm of gel was taken and the receiver

compartment was filled with 100ml of diffusion medium, Phosphate buffer

saline pH7.4 and a magnetic bead was placed in the PBS to bring the agitation.

The diffusion was carried out at 50 rpm at 37°C for 10hrs. Every hour 2ml of

diffusion fluid from receptor compartment was withdrawn and same volume of
fresh diffusion fluid was replaced to maintain sinc conditions. The withdrawn

diffusion fluids were suitably diluted and analyzed using UV spectrophotometer

at 306nm. The ex vivo drug release from all conventional, niosomal, ethosomal

and transferosomal gels was tabulated in tables 5.46. 5.47, 5.48 and 5.49

respectively and the same were graphically presented in figutes 5.42.

Specifications of ex vivo drug release study

Equipment : Artificial diffusion cell

Semipermeable membrane : Rat Skin

Diffusion medium : Phosphate buffer saline (PBS) pH 7.4

Volume of sampling fluid : 2ml

Temperature : 37±0.5°C

RPM : 50

Absorption maximum : 306nm

4.6.10. Transdermal flux

Amount of material flowing through a unit cross-sectional barrier in unit

time is said to be flux.

Mathematically it is expressed as,

Where,

J is the flux

Q is the quantity of compound traversing the membrane in time t

A is the area of exposed membrane in cm2


Flux of drug from all gels was calculated and compared with each other.

4.6.11. Data analysis of the ex vivo drug release

Cumulative amount of nystatin permeated (μg/cm 2) through the

membrane vs time (min) plot was constructed and parameters like transdermal

flux (Jss, μg/cm2/min) at steady state, permeation coefficient (Kp, cm 2/min),

etc was calculated. Slope of the linear portion of the cumulative amount of

nystatin permeated vs time was considered as transdermal flux. Permeation

coefficient (Kp) calculated by dividing the flux with the initial concentration of

the drug taken in the donor compartment and enhancement ratio (Er), extent

of increase in permeation of drug by vesicular gels than the conventional gel,

was calculated by dividing the nystatin permeation from vesicular gels by

concentration of nystatin permeated from conventional gel in a fixed time.

4.6.12. Drug retention study:

This is an important parameter for characterization of transdermal

drug delivery products which states the amount of drug reaches to the

systemic circulation and amount of drug retained in and on the skin. This

study will be carried out after completion of the ex vivo drug release study.

After completion of 12hrs ex vivo diffusion studies, the skin placed

between donor and receptor compartment was collected carefully and the gel
present on the skin was scrapped off precisely and preserved for further

studies.

The skin was sliced as very small thin pieces using surgical stainless

steel scalpel. The pieces were transferred into a 250ml beaker containing

100ml of PBS pH7.4. The beaker was kept under a mechanical stirrer and

stirring was initiated at a speed of 1000rpm and the same was continued for

24hrs. The entire process was carried out at a temperature of 37±1 0 C. After

24hrs, 10ml of solution collected from the beaker and filtered. The filtered

solution was centrifuged at 3000rpm for 30min and supernatant fluid was

collected and nystatin concentration was quantified using UV-Visible

spectrophotometer at 306nm.

The resultant nystatin concentration indicates the amount of the drug

retained in the skin. From the above data, amount of nystatin retained on the

skin was calculated by subtracting the amount of nystatin permeated (receptor

fluid concentration during ex vivo study) and retained within the skin. This was

confirmed by analyzing the collected overlying gel. i.e. gel collected from skin

surface169. The data of skin retention study was tabulated in table 5.50.

4.6.13. Stability Studies

In the present study, the stability of the vesicles was charecterized as per

ICH guidelines. Three optimized formulations preserved at refrigerated

temperature (4-8±1°C) and room temperature (25±2°C) for 180days. After 90


and 180days, shape, size and % entrapment efficiency of vesicles were

measured. The results were compared with the initial size, shape and %

entrapment efficiency of all three samples and are tabulated in tables 5.51-

5.53.

4.6.14. In vivo study of the gels in rabbits

An approved reliable reversed-phase high-performance liquid

chromatographic method170 was used for in vivo study. Four groups, consisting

of two rabbits in each group, were taken. Each group was treated with 1gm of

gels and is labled properly.

At different time points, given below, blood sapmle was collected from ear

vein of the rabbits and are analysed by HPLC for concentration of nystatin in

each sample. Nystatin is extracted by 1:2 (v/v) liquid-liquid extraction with

methanol. Separation is achieved by HPLC after direct injection on a

muBondapak C18 analytical column with a mobile phase composed of 10 mM

sodium phosphate, 1 mM EDTA, 30% methanol and 30% acetonitrile adjusted

to pH 6. Detection is by ultraviolet absorbance at 305 nm. Quantitation is

based on the sum of the peak area concentration of the two major isomers of

nystatin, which elute at 7.5-8.5 and 9.5-10.5 min. The assay was linear over

the concentration range of 0.05 to 50µg/ml. in vivo study results of

conventional, niosomal, ethosomal and transferosomal gels are tabulated in

tables 5.56, 5.57, 5.58 and 5.59 respectively. Summary of the nystatin

concentrations in blood plasma at different time intervals was tabulated in

table 5.60 and pharmacokinetic parameters are tabulated in table 5.61. HPLC
chromatograms of conventional, niosomal, ethosomal and transferosomal gels

in rabbits 1&2 are given in figures 5.53-5.68, 5.69-5.84, 5.85-5.100 and 5.101-

5.116 respectively.
Chapter - 5

EXPERIMENTAL RESULTS
5. EXPERIMENTAL RESULTS

Table 5.1: Solubility of nystatin in Phosphate buffer saline solution pH 7.4

Corresponding Actual Actual Actual


concentration concentration, concentration, concentration,
mcg/ml mcg/ml mcg/100ml mg/100ml Solubility
(corresponding (Actual of
concentration, concentration, (Actual nystatin,
mcg/ml)Xdil.Factor mcg/ml) X concentration, mg/100ml
S.No Absorbance y=0.045x+0.006 (100) 100 mcg/100ml)/1000 of PBS
1 0.163 3.4889 348.8889 34888.89 34.8889 34.89
2 0.149 3.1778 317.7778 31777.78 31.7778 31.78
3 0.158 3.3778 337.7778 33777.78 33.7778 33.78
Average solubility 33.48
Std. deviation 1.58
Table 5.2: Standard curve of nystatin in Phosphate buffer saline

solution pH 7.4

S.No Concentration, (mcg/ml) Absorbance (nm)

1 0 0.000
2 2 0.102
3 4 0.192
4 6 0.275
5 8 0.364
6 10 0.460

Table 5.3: Composition of niosomal formulations

Composition, mg
Formulation
S.No Non-ionic Surfactant
code Nystatin Cholesterol
Span-60 Span-80
1 FN1 100 50 -- 100
2 FN2 100 100 -- 100
3 FN3 100 150 -- 100
4 FN4 100 200 -- 100
5 FN5 100 -- 50 100
6 FN6 100 -- 100 100
7 FN7 100 -- 150 100
8 FN8 100 -- 200 100
Table 5.4: Composition of ethosomal formulations

Formulation Nystatin, Lecithin, Ethanol, Cholesterol,


S.No
code mg mg ml mg

1 FE1 20 100 15 --
2 FE2 20 100 30 --
3 FE3 20 100 45 --
4 FE4 20 100 60 --
5 FE5 20 200 45 --
6 FE6 20 300 45 --
7 FE7 20 400 45 --
8 FE8 20 300 45 20

Table 5.5: Composition of transferosomal formulations

Formulation Nystatin, Lecithin, Edge activator, mg


S.No
code mg mg Span-60 Span-80

1 FT1 100 960 40 --

2 FT2 100 920 80 --


3 FT3 100 880 120 --

4 FT4 100 840 160 --


5 FT5 100 960 -- 40

6 FT6 100 920 -- 80


7 FT7 100 880 -- 120

8 FT8 100 840 -- 160

Table 5.6: Vesicles size & Entrapment efficiency of all niosomal formulations

S.No Formulation Vesicle % Entrapment efficiency


code size, nm Centrifugation method Dialysis method
1 FN1 363±2.3 59.4±3.2 62.7±2.9
2 FN2 326±1.5 63.7±2.6 61.5±1.7
3 FN3 281±1.7 72.5±1.9 73.7±2.3
4 FN4 278±1.4 72.8±2.4 73.6±1.8
5 FN5 431±1.2 51.2±2.2 47.9±1.1
6 FN6 366±2.1 56.6±1.6 58.8±2.1
7 FN7 298±1.9 59.3±2.1 63.2±3.3
8 FN8 289±1.2 61.9±2.6 58.5±1.9

Table 5.7: Vesicles size & Entrapment efficiency of all ethosomal formulations

Formulation Vesicle % Entrapment efficiency


S.No
code size, nm Centrifugation method Dialysis method

1 FE1 463.2 42.6±1.6 44.2±1.5

2 FE2 416.1 64.8±1.9 62.5±2.3

3 FE3 371.7 78.7±2.1 78.3±2.7

4 FE4 291.2 61.5±1.1 64.8±3.3

5 FE5 438.2 81.3±2.4 80.7±1.6

6 FE6 412.5 88.4±1.4 87.5±2.1

7 FE7 403.3 86.8±1.7 83.9±2.8

8 FE8 391.6 87.6±2.2 85.3±2.4

Table 5.8: Vesicles size & Entrapment efficiency of all transferosomal formulations

Formulation Vesicle % Entrapment efficiency


S.No
code size, nm Centrifugation method Dialysis method
344.2
1 FT1 56.2±2.1 52.1±2.1
317.6
2 FT2 58.7±1.6 57.4±2.6
271.3
3 FT3 61.3±1.9 60.5±2.8
266.7
4 FT4 57.4±2.2 58.8±2.5
384.4
5 FT5 62.4±2.6 59.9±2.7
369.9
6 FT6 65.3±1.7 62.2±1.9
374.1
7 FT7 69.4±1.9 70.6±1.8
331.6
8 FT8 66.1±2.3 61.3±2.2
Table 5.9: Summary of in vitro drug release from all niosomal formulations across the dialysis membrane

% nystatin release from all niosomal formulations


S.No Time, Hrs FN1 FN2 FN3 FN4 FN5 FN6 FN7 FN8
1 0 0 0 0 0 0 0 0 0
2 1 9.09 7.22 6.48 4.81 11.33 6.89 6.58 5.49
3 2 17.23 14.94 13.13 9.6 21.96 14.48 15 11.12
4 3 28.77 22.06 19.38 17.39 34.42 27.39 21.95 19.64
5 4 35.19 30.76 26.48 24.82 43.27 36.36 31.76 28.52
6 5 43.11 38.56 33.87 28.72 51.34 44.66 42.14 35.02
7 6 51.54 45.45 39.09 35.03 60.37 52.08 48.55 41.68
8 7 58.47 51.54 47.04 43.55 75.06 72.21 54.75 52.51
9 8 66.22 61.21 54.34 50.89 84.81 77.97 63.77 61.82

Table 5.10: In vitro drug release from niosomal formulation FN1


amt. cumulative
in amt in amt.
TIME 5ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0 0 0 0.0 0.00 100.00 2.00 0.0
60 7.746 1.778 0.030 0.54 2.7 108 108.0 9.09 90.91 0.96 1.96 2.1
120 10.954 2.079 0.051 1.01 5.05 202 204.7 17.23 82.77 1.24 1.92 2.6
180 13.416 2.255 0.081 1.67 8.35 334 341.8 28.77 71.23 1.46 1.85 3.1
240 15.492 2.380 0.096 2.01 10.05 402 418.1 35.19 64.81 1.55 1.81 3.3
300 17.321 2.477 0.115 2.43 12.15 486 512.2 43.11 56.89 1.63 1.76 3.5
360 18.974 2.556 0.135 2.87 14.35 574 612.3 51.54 48.46 1.71 1.69 3.7
420 20.494 2.623 0.150 3.21 16.05 642 694.7 58.47 41.53 1.77 1.62 3.9
480 21.909 2.681 0.168 3.59 17.95 718 786.7 66.22 33.78 1.82 1.53 4.0

Table 5.11: In vitro drug release from niosomal formulation FN2


amt. cumulative
in amt in amt.
TIME 5ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.027 0.46 2.3 92 92.0 7.22 92.78 0.86 1.97 1.9
120 10.954 2.079 0.048 0.94 4.7 188 190.3 14.94 85.06 1.17 1.93 2.5
180 13.416 2.255 0.068 1.37 6.85 274 281.0 22.06 77.94 1.34 1.89 2.8
240 15.492 2.380 0.091 1.89 9.45 378 391.9 30.76 69.24 1.49 1.84 3.1
300 17.321 2.477 0.111 2.34 11.7 468 491.3 38.56 61.44 1.59 1.79 3.4
360 18.974 2.556 0.128 2.72 13.6 544 579.0 45.45 54.55 1.66 1.74 3.6
420 20.494 2.623 0.143 3.04 15.2 608 656.6 51.54 48.46 1.71 1.69 3.7
480 21.909 2.681 0.167 3.58 17.9 716 779.8 61.21 38.79 1.79 1.59 3.9

Table 5.12: In vitro drug release from niosomal formulation FN3


cumulative
amt. amt in amt.
TIME in 5ml 200ml. release log log cub rt
(min)t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.027 0.47 2.35 94 94.0 6.48 93.52 0.81 1.97 1.9
120 10.954 2.079 0.048 0.94 4.7 188 190.4 13.13 86.87 1.12 1.94 2.4
180 13.416 2.255 0.068 1.37 6.85 274 281.1 19.38 80.62 1.29 1.91 2.7
240 15.492 2.380 0.089 1.85 9.25 370 383.9 26.48 73.52 1.42 1.87 3.0
300 17.321 2.477 0.111 2.34 11.7 468 491.2 33.87 66.13 1.53 1.82 3.2
360 18.974 2.556 0.126 2.66 13.3 532 566.9 39.09 60.91 1.59 1.78 3.4
420 20.494 2.623 0.149 3.17 15.85 634 682.2 47.04 52.96 1.67 1.72 3.6
480 21.909 2.681 0.169 3.62 18.1 724 788.0 54.34 45.66 1.74 1.66 3.8

Table 5.13: In vitro drug release from niosomal formulation FN4


cumulative
amt. amt in amt.
TIME in 5ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.022 0.35 1.75 70 70.0 4.81 95.19 0.68 1.98 1.7
120 10.954 2.079 0.037 0.69 3.45 138 139.8 9.60 90.40 0.98 1.96 2.1
180 13.416 2.255 0.062 1.24 6.2 248 253.2 17.39 82.61 1.24 1.92 2.6
240 15.492 2.380 0.085 1.75 8.75 350 361.4 24.82 75.18 1.39 1.88 2.9
300 17.321 2.477 0.096 1.99 9.95 398 418.2 28.72 71.28 1.46 1.85 3.1
360 18.974 2.556 0.114 2.4 12 480 510.1 35.03 64.97 1.54 1.81 3.3
420 20.494 2.623 0.139 2.96 14.8 592 634.1 43.55 56.45 1.64 1.75 3.5
480 21.909 2.681 0.160 3.42 17.1 684 740.9 50.89 49.11 1.71 1.69 3.7

Table 5.14: In vitro drug release from niosomal formulation FN5


cumulative
amt. amt in amt.
TIME in 5ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.032 0.58 2.9 116 116.0 11.33 88.67 1.05 1.95 2.2
120 10.954 2.079 0.056 1.11 5.55 222 224.9 21.96 78.04 1.34 1.89 2.8
180 13.416 2.255 0.083 1.72 8.6 344 352.5 34.42 65.58 1.54 1.82 3.3
240 15.492 2.380 0.102 2.13 10.65 426 443.1 43.27 56.73 1.64 1.75 3.5
300 17.321 2.477 0.118 2.49 12.45 498 525.7 51.34 48.66 1.71 1.69 3.7
360 18.974 2.556 0.136 2.89 14.45 578 618.2 60.37 39.63 1.78 1.60 3.9
420 20.494 2.623 0.167 3.57 17.85 714 768.6 75.06 24.94 1.88 1.40 4.2
480 21.909 2.681 0.185 3.98 19.9 796 868.5 84.81 15.19 1.93 1.18 4.4

Table 5.15: In vitro drug release from niosomal formulation FN6


cumulative
amt. amt in amt.
TIME in 5ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.024 0.39 1.95 78 78.0 6.89 93.11 0.84 1.97 1.9
120 10.954 2.079 0.042 0.81 4.05 162 164.0 14.48 85.52 1.16 1.93 2.4
180 13.416 2.255 0.074 1.52 7.6 304 310.0 27.39 72.61 1.44 1.86 3.0
240 15.492 2.380 0.096 1.99 9.95 398 411.6 36.36 63.64 1.56 1.80 3.3
300 17.321 2.477 0.114 2.41 12.05 482 505.6 44.66 55.34 1.65 1.74 3.5
360 18.974 2.556 0.131 2.77 13.85 554 589.6 52.08 47.92 1.72 1.68 3.7
420 20.494 2.623 0.179 3.84 19.2 768 817.5 72.21 27.79 1.86 1.44 4.2
480 21.909 2.681 0.189 4.07 20.35 814 882.7 77.97 22.03 1.89 1.34 4.3

Table 5.16: In vitro drug release from niosomal formulation FN7

amt. amt in cumulative


TIME in 5ml 200ml. amt. log log cub rt
(min) t √t logt Abs conc (µg) (µg) release CPR CPRU CPR CPRU (CPR)
(µg)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.024 0.39 1.95 78 78.0 6.58 93.42 0.82 1.97 1.9
120 10.954 2.079 0.046 0.88 4.4 176 178.0 15.00 85.00 1.18 1.93 2.5
180 13.416 2.255 0.063 1.27 6.35 254 260.4 21.95 78.05 1.34 1.89 2.8
240 15.492 2.380 0.088 1.82 9.1 364 376.7 31.76 68.24 1.50 1.83 3.2
300 17.321 2.477 0.114 2.39 11.95 478 499.8 42.14 57.86 1.62 1.76 3.5
360 18.974 2.556 0.128 2.71 13.55 542 575.8 48.55 51.45 1.69 1.71 3.6
420 20.494 2.623 0.141 3.01 15.05 602 649.3 54.75 45.25 1.74 1.66 3.8
480 21.909 2.681 0.162 3.47 17.35 694 756.4 63.77 36.23 1.80 1.56 4.0

Table 5.17: In vitro drug release from niosomal formulation FN8

cumulative
amt. amt in amt.
TIME in 5ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 100.00 #NUM! 2.00 0.0
60 7.746 1.778 0.021 0.34 1.7 68 68.0 5.49 94.51 0.74 1.98 1.8
120 10.954 2.079 0.037 0.68 3.4 136 137.7 11.12 88.88 1.05 1.95 2.2
180 13.416 2.255 0.060 1.19 5.95 238 243.1 19.64 80.36 1.29 1.91 2.7
240 15.492 2.380 0.083 1.71 8.55 342 353.1 28.52 71.48 1.46 1.85 3.1
300 17.321 2.477 0.099 2.07 10.35 414 433.6 35.02 64.98 1.54 1.81 3.3
360 18.974 2.556 0.115 2.43 12.15 486 516.0 41.68 58.32 1.62 1.77 3.5
420 20.494 2.623 0.143 3.04 15.2 608 650.1 52.51 47.49 1.72 1.68 3.7
480 21.909 2.681 0.165 3.54 17.7 708 765.3 61.82 38.18 1.79 1.58 4.0

Table 5.18: Summary of release kinetics data of all nystatin Niosomal Formulations

Formulation Zero First Hixon


Order Order Higuchi Korsmeyer Peppas crowel Mechanism of
S.No code R² R² R² R² n R² drug release
1 FN1 0.996 0.987 0.996 0.9 0.113 0.776 Fickian diffusion
2 FN2 0.998 0.98 0.998 0.912 0.121 0.802 Fickian diffusion
3 FN3 0.999 0.983 0.999 0.922 0.122 0.805 Fickian diffusion
4 FN4 0.994 0.974 0.994 0.912 0.136 0.836 Fickian diffusion
5 FN5 0.996 0.921 0.996 0.916 0.114 0.783 Fickian diffusion
6 FN6 0.987 0.917 0.987 0.908 0.14 0.844 Fickian diffusion
7 FN7 0.997 0.98 0.997 0.898 0.129 0.816 Fickian diffusion
8 FN8 0.993 0.956 0.993 0.92 0.14 0.848 Fickian diffusion

Table 5.19: Summary of in vitro nystatin release from all ethosomal formulations across

the dialysis membrane

Cumulative amount of nystatin permeated from ethosomal suspensions


S.No TIME, Min FE1 FE2 FE3 FE4 FE5 FE6 FE7 FE8
1 15 4.46 6.48 5.97 19.84 8.49 5.09 3.69 5.48
2 30 6.38 13.65 13.02 21.66 16.57 10.35 5.68 10.10
3 60 10.91 15.79 17.09 27.89 23.00 15.20 10.12 14.77
4 120 15.47 19.65 21.83 33.37 26.43 22.70 17.01 19.59
5 180 18.91 24.31 25.73 38.74 33.58 26.77 23.29 23.44
6 240 26.61 27.95 29.03 44.81 36.98 34.50 27.90 31.32
7 300 31.80 32.23 35.29 48.82 41.03 39.14 34.04 35.05
8 360 36.10 37.33 40.33 53.35 44.38 45.51 40.48 39.38
9 420 40.68 40.46 44.91 56.95 48.25 54.55 47.79 44.56
10 480 43.88 45.17 49.66 62.86 51.29 62.43 54.47 49.32
11 540 46.65 47.46 53.44 65.89 55.84 67.44 60.41 55.73
12 600 51.79 51.15 57.00 69.44 59.19 72.83 68.13 60.83
13 660 56.98 56.72 61.73 75.46 64.91 77.15 72.01 64.83
14 720 61.29 62.04 63.58 78.11 73.64 79.61 78.49 69.25

Table 5.20:In vitro drug release from ethosomal formulation FE1

cumulative
amt. in amt in amt.
TIME Conc, 2ml, 200ml, release log log cub rt
(min) t √t logt Abs (µg) (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.015 0.19 0.38 38 38.0 4.46 95.54 0.65 1.98 1.6
30 5.477 1.477 0.018 0.27 0.54 54 54.4 6.38 93.62 0.80 1.97 1.9
60 7.746 1.778 0.027 0.46 0.92 92 92.9 10.91 89.09 1.04 1.95 2.2
120 10.954 2.079 0.035 0.65 1.3 130 131.8 15.47 84.53 1.19 1.93 2.5
180 13.416 2.255 0.042 0.79 1.58 158 161.1 18.91 81.09 1.28 1.91 2.7
240 15.492 2.380 0.056 1.11 2.22 222 226.7 26.61 73.39 1.43 1.87 3.0
300 17.321 2.477 0.065 1.32 2.64 264 270.9 31.80 68.20 1.50 1.83 3.2
360 18.974 2.556 0.073 1.49 2.98 298 307.6 36.10 63.90 1.56 1.81 3.3
420 20.494 2.623 0.081 1.67 3.34 334 346.6 40.68 59.32 1.61 1.77 3.4
480 21.909 2.681 0.087 1.79 3.58 358 373.9 43.88 56.12 1.64 1.75 3.5
540 23.238 2.732 0.091 1.89 3.78 378 397.5 46.65 53.35 1.67 1.73 3.6
600 24.495 2.778 0.100 2.09 4.18 418 441.3 51.79 48.21 1.71 1.68 3.7
660 25.690 2.820 0.109 2.29 4.58 458 485.4 56.98 43.02 1.76 1.63 3.8
720 26.833 2.857 0.117 2.46 4.92 492 522.2 61.29 38.71 1.79 1.59 3.9

Table 5.21: In vitro drug release from ethosomal formulation FE2

cumulative
amt. amt in amt.
TIME in 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.025 0.42 0.84 84 84 6.48 93.52 1.97 1.86
30 5.477 1.477 0.046 0.88 1.76 176 176.84 13.65 86.35 1.13 1.94 2.39
60 7.746 1.778 0.051 1.01 2.02 202 204.6 15.79 84.21 1.20 1.93 2.51
120 10.954 2.079 0.062 1.25 2.5 250 254.62 19.65 80.35 1.29 1.91 2.70
180 13.416 2.255 0.075 1.54 3.08 308 315.12 24.31 75.69 1.39 1.88 2.90
240 15.492 2.380 0.085 1.76 3.52 352 362.2 27.95 72.05 1.45 1.86 3.03
300 17.321 2.477 0.097 2.02 4.04 404 417.72 32.23 67.77 1.51 1.83 3.18
360 18.974 2.556 0.111 2.33 4.66 466 483.76 37.33 62.67 1.57 1.80 3.34
420 20.494 2.623 0.119 2.51 5.02 502 524.42 40.46 59.54 1.61 1.77 3.43
480 21.909 2.681 0.132 2.79 5.58 558 585.44 45.17 54.83 1.65 1.74 3.56
540 23.238 2.732 0.137 2.91 5.82 582 615.02 47.46 52.54 1.68 1.72 3.62
600 24.495 2.778 0.146 3.12 6.24 624 662.84 51.15 48.85 1.71 1.69 3.71
660 25.690 2.820 0.161 3.45 6.9 690 735.08 56.72 43.28 1.75 1.64 3.84

Table 5.22: In vitro drug release from ethosomal formulation FE3

cumulative
amt. in amt in amt.
TIME 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.027 0.47 0.94 94 94 5.97 94.03 1.97 1.81
30 5.477 1.477 0.052 1.02 2.04 204 204.94 13.02 86.98 1.11 1.94 2.35
60 7.746 1.778 0.066 1.33 2.66 266 268.98 17.09 82.91 1.23 1.92 2.58
120 10.954 2.079 0.082 1.69 3.38 338 343.64 21.83 78.17 1.34 1.89 2.79
180 13.416 2.255 0.095 1.98 3.96 396 405.02 25.73 74.27 1.41 1.87 2.95
240 15.492 2.380 0.106 2.22 4.44 444 456.98 29.03 70.97 1.46 1.85 3.07
300 17.321 2.477 0.127 2.69 5.38 538 555.42 35.29 64.71 1.55 1.81 3.28
360 18.974 2.556 0.144 3.06 6.12 612 634.8 40.33 59.67 1.61 1.78 3.43
420 20.494 2.623 0.159 3.39 6.78 678 706.92 44.91 55.09 1.65 1.74 3.55
480 21.909 2.681 0.174 3.73 7.46 746 781.7 49.66 50.34 1.70 1.70 3.68
540 23.238 2.732 0.186 3.99 7.98 798 841.16 53.44 46.56 1.73 1.67 3.77
600 24.495 2.778 0.196 4.23 8.46 846 897.14 57.00 43.00 1.76 1.63 3.85
660 25.690 2.820 0.211 4.56 9.12 912 971.6 61.73 38.27 1.79 1.58 3.95
720 26.833 2.857 0.216 4.66 9.32 932 1000.72 63.58 36.42 1.80 1.56 3.99

Table 5.23:In vitro drug release from ethosomal formulation FE4

cumulative
amt. amt in amt.
TIME in 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.061 1.22 2.44 244 244 19.84 80.16 1.30 1.90 2.71
30 5.477 1.477 0.065 1.32 2.64 264 266.44 21.66 78.34 1.34 1.89 2.79
60 7.746 1.778 0.082 1.69 3.38 338 343.08 27.89 72.11 1.45 1.86 3.03
120 10.954 2.079 0.096 2.01 4.02 402 410.46 33.37 66.63 1.52 1.82 3.22
180 13.416 2.255 0.110 2.32 4.64 464 476.48 38.74 61.26 1.59 1.79 3.38
240 15.492 2.380 0.126 2.67 5.34 534 551.12 44.81 55.19 1.65 1.74 3.55
300 17.321 2.477 0.136 2.89 5.78 578 600.46 48.82 51.18 1.69 1.71 3.65
360 18.974 2.556 0.147 3.14 6.28 628 656.24 53.35 46.65 1.73 1.67 3.76
420 20.494 2.623 0.156 3.33 6.66 666 700.52 56.95 43.05 1.76 1.63 3.85
480 21.909 2.681 0.171 3.66 7.32 732 773.18 62.86 37.14 1.80 1.57 3.98
540 23.238 2.732 0.177 3.81 7.62 762 810.5 65.89 34.11 1.82 1.53 4.04
600 24.495 2.778 0.186 3.99 7.98 798 854.12 69.44 30.56 1.84 1.49 4.11
660 25.690 2.820 0.200 4.32 8.64 864 928.1 75.46 24.54 1.88 1.39 4.23
720 26.833 2.857 0.206 4.44 8.88 888 960.74 78.11 21.89 1.89 1.34 4.27

Table 5.24: In vitro drug release from ethosomal formulation FE5

cumulative
amt. in amt in amt.
TIME 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.037 0.69 1.38 138 138 8.49 91.51 0.93 1.96 2.04
30 5.477 1.477 0.066 1.34 2.68 268 269.38 16.57 83.43 1.22 1.92 2.55
60 7.746 1.778 0.089 1.85 3.7 370 374.06 23.00 77.00 1.36 1.89 2.84
120 10.954 2.079 0.101 2.11 4.22 422 429.76 26.43 73.57 1.42 1.87 2.98
180 13.416 2.255 0.126 2.67 5.34 534 545.98 33.58 66.42 1.53 1.82 3.23
240 15.492 2.380 0.137 2.92 5.84 584 601.32 36.98 63.02 1.57 1.80 3.33
300 17.321 2.477 0.151 3.22 6.44 644 667.16 41.03 58.97 1.61 1.77 3.45
360 18.974 2.556 0.162 3.46 6.92 692 721.6 44.38 55.62 1.65 1.75 3.54
420 20.494 2.623 0.174 3.74 7.48 748 784.52 48.25 51.75 1.68 1.71 3.64
480 21.909 2.681 0.184 3.95 7.9 790 834 51.29 48.71 1.71 1.69 3.72
540 23.238 2.732 0.199 4.28 8.56 856 907.9 55.84 44.16 1.75 1.65 3.82
600 24.495 2.778 0.209 4.51 9.02 902 962.46 59.19 40.81 1.77 1.61 3.90
660 25.690 2.820 0.228 4.93 9.86 986 1055.48 64.91 35.09 1.81 1.55 4.02
720 26.833 2.857 0.258 5.59 11.18 1118 1197.34 73.64 26.36 1.87 1.42 4.19

Table 5.25: In vitro drug release from ethosomal formulation FE6

cumulative
amt. in amt in amt.
TIME 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.026 0.45 0.9 90 90 5.09 94.91 0.71 1.98 1.72
30 5.477 1.477 0.047 0.91 1.82 182 182.9 10.35 89.65 1.01 1.95 2.18
60 7.746 1.778 0.066 1.33 2.66 266 268.72 15.20 84.80 1.18 1.93 2.48
120 10.954 2.079 0.095 1.98 3.96 396 401.38 22.70 77.30 1.36 1.89 2.83
180 13.416 2.255 0.110 2.32 4.64 464 473.34 26.77 73.23 1.43 1.86 2.99
240 15.492 2.380 0.140 2.98 5.96 596 609.98 34.50 65.50 1.54 1.82 3.26
300 17.321 2.477 0.157 3.36 6.72 672 691.94 39.14 60.86 1.59 1.78 3.40
360 18.974 2.556 0.181 3.89 7.78 778 804.66 45.51 54.49 1.66 1.74 3.57
420 20.494 2.623 0.215 4.65 9.3 930 964.44 54.55 45.45 1.74 1.66 3.79
480 21.909 2.681 0.245 5.3 10.6 1060 1103.74 62.43 37.57 1.80 1.57 3.97
540 23.238 2.732 0.262 5.69 11.38 1138 1192.34 67.44 32.56 1.83 1.51 4.07
600 24.495 2.778 0.281 6.11 12.22 1222 1287.72 72.83 27.17 1.86 1.43 4.18
660 25.690 2.820 0.295 6.43 12.86 1286 1363.94 77.15 22.85 1.89 1.36 4.26
720 26.833 2.857 0.303 6.61 13.22 1322 1407.42 79.61 20.39 1.90 1.31 4.30

Table 5.26: In vitro drug release from ethosomal formulation FE7

cumulative
amt. in amt in amt.
TIME 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.020 0.32 0.64 64 64 3.69 96.31 0.57 1.98 1.54
30 5.477 1.477 0.028 0.49 0.98 98 98.64 5.68 94.32 0.75 1.97 1.78
60 7.746 1.778 0.045 0.87 1.74 174 175.62 10.12 89.88 1.01 1.95 2.16
120 10.954 2.079 0.072 1.46 2.92 292 295.36 17.01 82.99 1.23 1.92 2.57
180 13.416 2.255 0.096 1.99 3.98 398 404.28 23.29 76.71 1.37 1.88 2.86
240 15.492 2.380 0.113 2.37 4.74 474 484.26 27.90 72.10 1.45 1.86 3.03
300 17.321 2.477 0.136 2.88 5.76 576 591 34.04 65.96 1.53 1.82 3.24
360 18.974 2.556 0.159 3.41 6.82 682 702.76 40.48 59.52 1.61 1.77 3.43
420 20.494 2.623 0.186 4.01 8.02 802 829.58 47.79 52.21 1.68 1.72 3.63
480 21.909 2.681 0.211 4.55 9.1 910 945.6 54.47 45.53 1.74 1.66 3.79
540 23.238 2.732 0.232 5.02 10.04 1004 1048.7 60.41 39.59 1.78 1.60 3.92
600 24.495 2.778 0.260 5.64 11.28 1128 1182.74 68.13 31.87 1.83 1.50 4.08
660 25.690 2.820 0.272 5.92 11.84 1184 1250.02 72.01 27.99 1.86 1.45 4.16
720 26.833 2.857 0.296 6.44 12.88 1288 1362.5 78.49 21.51 1.89 1.33 4.28

Table 5.27: In vitro drug release from ethosomal formulation FE8

cumulative
amt. in amt in amt.
TIME 2ml 200ml. release log log cub rt
(min) t √t logt Abs conc (µg) (µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.028 0.48 0.96 96 96 5.48 94.52 0.74 1.98 1.76
30 5.477 1.477 0.046 0.88 1.76 176 176.96 10.10 89.90 1.00 1.95 2.16
60 7.746 1.778 0.064 1.28 2.56 256 258.72 14.77 85.23 1.17 1.93 2.45
120 10.954 2.079 0.082 1.69 3.38 338 343.28 19.59 80.41 1.29 1.91 2.70
180 13.416 2.255 0.096 2.01 4.02 402 410.66 23.44 76.56 1.37 1.88 2.86
240 15.492 2.380 0.127 2.68 5.36 536 548.68 31.32 68.68 1.50 1.84 3.15
300 17.321 2.477 0.140 2.98 5.96 596 614.04 35.05 64.95 1.54 1.81 3.27
360 18.974 2.556 0.156 3.33 6.66 666 690 39.38 60.62 1.60 1.78 3.40
420 20.494 2.623 0.175 3.75 7.5 750 780.66 44.56 55.44 1.65 1.74 3.55
480 21.909 2.681 0.192 4.13 8.26 826 864.16 49.32 50.68 1.69 1.70 3.67
540 23.238 2.732 0.215 4.65 9.3 930 976.42 55.73 44.27 1.75 1.65 3.82
600 24.495 2.778 0.233 5.05 10.1 1010 1065.72 60.83 39.17 1.78 1.59 3.93
660 25.690 2.820 0.247 5.35 10.7 1070 1135.82 64.83 35.17 1.81 1.55 4.02
720 26.833 2.857 0.263 5.71 11.42 1142 1213.24 69.25 30.75 1.84 1.49 4.11

Table 5.28: Summary of Release kinetics data of all nystatin ethosomal suspension

Formulation Zero First Hixon- Korsmeyer-


order order Higuchi crowel peppas Mechanism of
S.No Code R2 R2 R2 R2 R2 n drug release
1 FE1 0.992 0.992 0.979 0.924 0.994 0.684 Non-Fickian diffusion
2 FE2 0.99 0.984 0.973 0.925 0.959 0.492 Non-Fickian diffusion
3 FE3 0.986 0.993 0.982 0.907 0.974 0.517 Non-Fickian diffusion
4 FE4 0.988 0.988 0.989 0.948 0.973 0.367 Fickian diffusion
5 FE5 0.971 0.958 0.976 0.887 0.976 0.483 Non-Fickian diffusion
6 FE6 0.991 0.979 0.975 0.921 0.991 0.685 Non-Fickian diffusion
7 FE7 0.996 0.948 0.996 0.961 0.895 0.95 Non-Fickian diffusion
8 FE8 0.994 0.985 0.975 0.929 0.99 0.624 Non-Fickian diffusion
Table 5.29: Summary of in vitro nystatin release from all transferosomal formulations across the dialysis membrane

In released from all transferosomal formulations


S.No Time, Min FT1 FT2 FT3 FT4 FT5 FT6 FT7 FT8

1 15 1.7 1.0 3.1 1.0 1.8 7.7 19.7 5.3


2 30 3.2 2.9 6.8 5.5 5.2 15.3 24.1 10.6

3 60 5.8 5.5 10.8 11.4 10.0 21.5 28.9 15.7


4 120 9.7 9.7 14.8 12.9 16.0 28.3 35.9 20.3

5 180 14.8 14.3 19.9 16.7 20.6 32.4 39.9 25.8


6 240 19.0 19.4 24.4 21.3 24.9 38.0 44.9 30.4

7 300 23.4 23.2 27.6 25.3 28.9 43.9 49.5 34.2


8 360 27.1 26.7 32.1 30.0 33.6 48.5 53.5 39.5

9 420 31.8 30.8 37.0 32.9 40.1 53.4 56.6 45.4


10 480 36.6 35.9 41.3 35.4 44.4 56.7 60.3 49.5
11 540 41.9 39.9 45.1 40.9 48.3 59.6 66.3 55.6

12 600 45.1 43.2 49.0 46.8 54.3 63.2 69.2 61.6


13 660 48.5 45.8 51.8 51.2 60.0 65.5 72.3 66.9

14 720 51.1 54.2 58.3 55.3 65.2 69.2 75.2 72.1

Table 5.30: In vitro drug release from transferosomal formulation FT1

cumulative
amt. cub
TIME amt. in amt in release log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.018 0.27 0.54 54 54 4.98 95.02 0.70 1.98 1.71
30 5.477 1.477 0.034 0.62 1.24 124 124.54 11.49 88.51 1.06 1.95 2.26
60 7.746 1.778 0.056 1.1 2.2 220 221.78 20.46 79.54 1.31 1.90 2.74
120 10.954 2.079 0.078 1.6 3.2 320 323.98 29.89 70.11 1.48 1.85 3.10
180 13.416 2.255 0.086 1.77 3.54 354 361.18 33.32 66.68 1.52 1.82 3.22
240 15.492 2.380 0.093 1.93 3.86 386 396.72 36.60 63.40 1.56 1.80 3.32
300 17.321 2.477 0.099 2.07 4.14 414 428.58 39.54 60.46 1.60 1.78 3.41
360 18.974 2.556 0.103 2.16 4.32 432 450.72 41.58 58.42 1.62 1.77 3.46
420 20.494 2.623 0.106 2.23 4.46 446 469.04 43.27 56.73 1.64 1.75 3.51
480 21.909 2.681 0.109 2.29 4.58 458 485.5 44.79 55.21 1.65 1.74 3.55
540 23.238 2.732 0.110 2.31 4.62 462 494.08 45.58 54.42 1.66 1.74 3.57
600 24.495 2.778 0.111 2.34 4.68 468 504.7 46.56 53.44 1.67 1.73 3.60
660 25.690 2.820 0.115 2.42 4.84 484 525.38 48.47 51.53 1.69 1.71 3.65
720 26.833 2.857 0.120 2.54 5.08 508 554.22 51.13 48.87 1.71 1.69 3.71

Table 5.31: In vitro drug release from transferosomal formulation FT2

cumulative
amt. cub
TIME amt. in amt in release log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.026 0.44 0.88 88 88 7.57 92.43 0.88 1.97 1.96
30 5.477 1.477 0.036 0.67 1.34 134 134.88 11.61 88.39 1.06 1.95 2.26
60 7.746 1.778 0.052 1.02 2.04 204 206.22 17.75 82.25 1.25 1.92 2.61
120 10.954 2.079 0.061 1.23 2.46 246 250.26 21.54 78.46 1.33 1.89 2.78
180 13.416 2.255 0.092 1.91 3.82 382 388.72 33.45 66.55 1.52 1.82 3.22
240 15.492 2.380 0.105 2.19 4.38 438 448.54 38.60 61.40 1.59 1.79 3.38
300 17.321 2.477 0.117 2.46 4.92 492 506.92 43.62 56.38 1.64 1.75 3.52
360 18.974 2.556 0.119 2.52 5.04 504 523.84 45.08 54.92 1.65 1.74 3.56
420 20.494 2.623 0.123 2.59 5.18 518 542.88 46.72 53.28 1.67 1.73 3.60
480 21.909 2.681 0.124 2.62 5.24 524 554.06 47.68 52.32 1.68 1.72 3.63
540 23.238 2.732 0.124 2.63 5.26 526 561.3 48.30 51.70 1.68 1.71 3.64
600 24.495 2.778 0.125 2.64 5.28 528 568.56 48.93 51.07 1.69 1.71 3.66
660 25.690 2.820 0.126 2.66 5.32 532 577.84 49.73 50.27 1.70 1.70 3.68
720 26.833 2.857 0.140 2.98 5.96 596 647.16 55.69 44.31 1.75 1.65 3.82
Table 5.32: In vitro drug release from transferosomal formulation FT3

cumulative
amt. cub
TIME amt. in amt in release log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.025 0.43 0.86 86 86 7.06 92.94 0.85 1.97 1.92
30 5.477 1.477 0.035 0.64 1.28 128 128.86 10.58 89.42 1.02 1.95 2.20
60 7.746 1.778 0.051 1.01 2.02 202 204.14 16.76 83.24 1.22 1.92 2.56
120 10.954 2.079 0.062 1.24 2.48 248 252.16 20.70 79.30 1.32 1.90 2.75
180 13.416 2.255 0.110 2.32 4.64 464 470.64 38.64 61.36 1.59 1.79 3.38
240 15.492 2.380 0.114 2.4 4.8 480 491.28 40.33 59.67 1.61 1.78 3.43
300 17.321 2.477 0.124 2.62 5.24 524 540.08 44.34 55.66 1.65 1.75 3.54
360 18.974 2.556 0.130 2.76 5.52 552 573.32 47.07 52.93 1.67 1.72 3.61
420 20.494 2.623 0.133 2.83 5.66 566 592.84 48.67 51.33 1.69 1.71 3.65
480 21.909 2.681 0.135 2.86 5.72 572 604.5 49.63 50.37 1.70 1.70 3.67
540 23.238 2.732 0.137 2.92 5.84 584 622.22 51.09 48.91 1.71 1.69 3.71
600 24.495 2.778 0.142 3.02 6.04 604 648.06 53.21 46.79 1.73 1.67 3.76
660 25.690 2.820 0.149 3.17 6.34 634 684.1 56.17 43.83 1.75 1.64 3.83
720 26.833 2.857 0.153 3.26 6.52 652 708.44 58.16 41.84 1.76 1.62 3.87
Table 5.33: In vitro drug release from transferosomal formulation FT4

cumulative cub
TIME amt. in amt in amt. release log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.017 0.24 0.48 48 48 4.13 95.87 0.62 1.98 1.60
30 5.477 1.477 0.023 0.38 0.76 76 76.48 6.58 93.42 0.82 1.97 1.87
60 7.746 1.778 0.036 0.66 1.32 132 133.24 11.47 88.53 1.06 1.95 2.25
120 10.954 2.079 0.039 0.74 1.48 148 150.56 12.96 87.04 1.11 1.94 2.35
180 13.416 2.255 0.061 1.22 2.44 244 248.04 21.35 78.65 1.33 1.90 2.77
240 15.492 2.380 0.069 1.4 2.8 280 286.48 24.65 75.35 1.39 1.88 2.91
300 17.321 2.477 0.070 1.42 2.84 284 293.28 25.24 74.76 1.40 1.87 2.93
360 18.974 2.556 0.082 1.69 3.38 338 350.12 30.13 69.87 1.48 1.84 3.11
420 20.494 2.623 0.089 1.84 3.68 368 383.5 33.00 67.00 1.52 1.83 3.21
480 21.909 2.681 0.095 1.97 3.94 394 413.18 35.56 64.44 1.55 1.81 3.29
540 23.238 2.732 0.108 2.26 4.52 452 475.12 40.89 59.11 1.61 1.77 3.45
600 24.495 2.778 0.116 2.44 4.88 488 515.64 44.38 55.62 1.65 1.75 3.54
660 25.690 2.820 0.122 2.58 5.16 516 548.52 47.20 52.80 1.67 1.72 3.61
720 26.833 2.857 0.125 2.64 5.28 528 565.68 48.68 51.32 1.69 1.71 3.65
Table 5.34: In vitro drug release from transferosomal formulation FT5

cumulative
amt. cub
TIME amt. in amt in release log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.011 0.11 0.22 22 22 1.80 98.20 0.25 1.99 1.22
30 5.477 1.477 0.030 0.54 1.08 108 108.22 8.84 91.16 0.95 1.96 2.07
60 7.746 1.778 0.049 0.95 1.9 190 191.3 15.63 84.37 1.19 1.93 2.50
120 10.954 2.079 0.073 1.48 2.96 296 299.2 24.44 75.56 1.39 1.88 2.90
180 13.416 2.255 0.087 1.81 3.62 362 368.16 30.08 69.92 1.48 1.84 3.11
240 15.492 2.380 0.093 1.93 3.86 386 395.78 32.33 67.67 1.51 1.83 3.19
300 17.321 2.477 0.096 1.99 3.98 398 411.64 33.63 66.37 1.53 1.82 3.23
360 18.974 2.556 0.109 2.28 4.56 456 473.62 38.69 61.31 1.59 1.79 3.38
420 20.494 2.623 0.111 2.34 4.68 468 490.18 40.05 59.95 1.60 1.78 3.42
480 21.909 2.681 0.117 2.46 4.92 492 518.86 42.39 57.61 1.63 1.76 3.49
540 23.238 2.732 0.140 2.98 5.96 596 627.78 51.29 48.71 1.71 1.69 3.72
600 24.495 2.778 0.147 3.13 6.26 626 663.74 54.23 45.77 1.73 1.66 3.79
660 25.690 2.820 0.163 3.48 6.96 696 740 60.46 39.54 1.78 1.60 3.92
720 26.833 2.857 0.174 3.74 7.48 748 798.96 65.27 34.73 1.81 1.54 4.03

Table 5.35: In vitro drug release from transferosomal formulation FT6


cumulative
amt. cub
TIME amt. in amt in release log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) (µg) CPR CPRU CPR CPRU (CPR)
15 3.873 1.176 0.030 0.53 1.06 106 106 8.31 91.69 0.92 1.96 2.03
30 5.477 1.477 0.050 0.97 1.94 194 195.06 15.29 84.71 1.18 1.93 2.48
60 7.746 1.778 0.067 1.36 2.72 272 275 21.55 78.45 1.33 1.89 2.78
120 10.954 2.079 0.083 1.72 3.44 344 349.72 27.41 72.59 1.44 1.86 3.02
180 13.416 2.255 0.097 2.02 4.04 404 413.16 32.38 67.62 1.51 1.83 3.19
240 15.492 2.380 0.112 2.36 4.72 472 485.2 38.03 61.97 1.58 1.79 3.36
300 17.321 2.477 0.128 2.71 5.42 542 559.92 43.88 56.12 1.64 1.75 3.53
360 18.974 2.556 0.140 2.98 5.96 596 619.34 48.54 51.46 1.69 1.71 3.65
420 20.494 2.623 0.153 3.26 6.52 652 681.3 53.39 46.61 1.73 1.67 3.77
480 21.909 2.681 0.161 3.44 6.88 688 723.82 56.73 43.27 1.75 1.64 3.84
540 23.238 2.732 0.168 3.59 7.18 718 760.7 59.62 40.38 1.78 1.61 3.91
600 24.495 2.778 0.176 3.78 7.56 756 805.88 63.16 36.84 1.80 1.57 3.98
660 25.690 2.820 0.181 3.89 7.78 778 835.44 65.47 34.53 1.82 1.54 4.03
720 26.833 2.857 0.190 4.09 8.18 818 883.22 69.22 30.78 1.84 1.49 4.11

Table 5.36: In vitro drug release from transferosomal formulation FT7

TIME amt. in amt in cumulative log log cub


(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) amt. CPR CPRU CPR CPRU rt
release
(µg) (CPR)
15 3.873 1.176 0.062 1.24 2.48 248 248 17.71 82.29 1.25 1.92 2.61
30 5.477 1.477 0.082 1.68 3.36 336 338.48 24.18 75.82 1.38 1.88 2.89
60 7.746 1.778 0.105 2.21 4.42 442 447.84 31.99 68.01 1.50 1.83 3.17
120 10.954 2.079 0.126 2.66 5.32 532 542.26 38.73 61.27 1.59 1.79 3.38
180 13.416 2.255 0.146 3.1 6.2 620 635.58 45.40 54.60 1.66 1.74 3.57
240 15.492 2.380 0.167 3.58 7.16 716 737.78 52.70 47.30 1.72 1.67 3.75
300 17.321 2.477 0.186 3.99 7.98 798 826.94 59.07 40.93 1.77 1.61 3.89
360 18.974 2.556 0.203 4.38 8.76 876 912.92 65.21 34.79 1.81 1.54 4.03
420 20.494 2.623 0.210 4.54 9.08 908 953.68 68.12 31.88 1.83 1.50 4.08
480 21.909 2.681 0.215 4.64 9.28 928 982.76 70.20 29.80 1.85 1.47 4.13
540 23.238 2.732 0.217 4.69 9.38 938 1002.04 71.57 28.43 1.85 1.45 4.15
600 24.495 2.778 0.220 4.75 9.5 950 1023.42 73.10 26.90 1.86 1.43 4.18
660 25.690 2.820 0.222 4.81 9.62 962 1044.92 74.64 25.36 1.87 1.40 4.21
720 26.833 2.857 0.224 4.84 9.68 968 1060.54 75.75 24.25 1.88 1.38 4.23

Table 5.37: In vitro drug release from transferosomal formulation FT8

cumulative cub
TIME amt. in amt in amt. log log rt
(min) t √t logt Abs conc 2ml(µg) 200ml.(µg) release CPR CPRU CPR CPRU (CPR)
(µg)
15 3.873 1.176 0.021 0.34 0.68 68 68 5.34 94.66 0.73 1.98 1.75
30 5.477 1.477 0.036 0.67 1.34 134 134.68 10.57 89.43 1.02 1.95 2.19
60 7.746 1.778 0.051 0.99 1.98 198 200.02 15.70 84.30 1.20 1.93 2.50
120 10.954 2.079 0.063 1.27 2.54 254 258 20.25 79.75 1.31 1.90 2.73
180 13.416 2.255 0.078 1.61 3.22 322 328.54 25.79 74.21 1.41 1.87 2.95
240 15.492 2.380 0.091 1.89 3.78 378 387.76 30.44 69.56 1.48 1.84 3.12
300 17.321 2.477 0.101 2.11 4.22 422 435.54 34.19 65.81 1.53 1.82 3.25
360 18.974 2.556 0.115 2.43 4.86 486 503.76 39.54 60.46 1.60 1.78 3.41
420 20.494 2.623 0.131 2.78 5.56 556 578.62 45.42 54.58 1.66 1.74 3.57
480 21.909 2.681 0.141 3.01 6.02 602 630.18 49.46 50.54 1.69 1.70 3.67
540 23.238 2.732 0.158 3.37 6.74 674 708.2 55.59 44.41 1.74 1.65 3.82
600 24.495 2.778 0.173 3.72 7.44 744 784.94 61.61 38.39 1.79 1.58 3.95
660 25.690 2.820 0.187 4.02 8.04 804 852.38 66.91 33.09 1.83 1.52 4.06
720 26.833 2.857 0.200 4.31 8.62 862 918.42 72.09 27.91 1.86 1.45 4.16

Table 5.38: Summary of Release kinetics data of all nystatin transferosomal suspension

Zero First Hixon- Korsmeyer-


Formulation Higuchi Mechanism of
S.No order order crowel peppas
Code drug release
R2 R2 R2 R2 R2 n
1 FT1 0.862 0.915 0.862 0.738 0.66 0.055 Fickian diffusion
Fickian diffusion
2 FT2 0.88 0.916 0.88 0.811 0.765 0.056
Fickian diffusion
3 FT3 0.886 0.932 0.886 0.81 0.763 0.061
Fickian diffusion
4 FT4 0.99 0.991 0.99 0.934 0.873 0.074
Fickian diffusion
5 FT5 0.974 0.965 0.974 0.841 0.7 0.082
Fickian diffusion
6 FT6 0.981 0.998 0.981 0.911 0.85 0.059
Fickian diffusion
7 FT7 0.999 0.989 0.999 0.981 0.96 0.078
Fickian diffusion
8 FT8 0.998 0.962 0.998 0.96 0.896 0.074

Table 5.39: Elasticity measurements of three optimized vesicular formulations

Volume of
liquid Vesicle Pore
Name of the permeated size size of
vesicular after filter Elasticity,
carrier Formulation in 5min, extrusion, paper,
systems code ml nm (rv) µm (rp) (rv/rp) (rv/rp)2 nm.ml/µm Average ±SD
1.9 241 50 4.82 23.2324 44.1
Niosomes FN3 1.8 248 50 4.96 24.6016 44.3 45.2 1.7
2.1 237 50 4.74 22.4676 47.2
2.7 276 50 5.52 30.4704 82.3
2.5 269 50 5.38 28.9444 72.4
Ethosomes FE6 2.8 274 50 5.48 30.0304 84.1 79.6 6.3
3.3 292 50 5.84 34.1056 112.5
Transferosome 3.2 288 50 5.76 33.1776 106.2
s FT7 3.5 293 50 5.86 34.3396 120.2 113.0 7.0

Table 5.40: Zeta potential of three optimized vesicular formulations

Name of the vesicular carrier systems


Niosomes Ethosomes, Transferosomes,
S.No FN3 FE6 FT7
Zeta potential, mV 24.2 -38.1 -20

Table 5.41: Composition of conventional, niosomal, ethosomal &transferosomal gels

Ingredients Conventional gel Niosomal gel Ethosomal gel Transferosomal gel

Nystatin 0.01%w/w 0.01%w/w 0.01%w/w 0.01%w/w


Carbopol -971 1%w/w 1%w/w 1%w/w 1%w/w
HPMC 0.5% w/w 0.5% w/w 0.5% w/w 0.5% w/w
Propylene glycol 2% 2% 2% 2%
Triethanolamine q.s q.s q.s q.s
Double distilled
100% w/w 100% w/w 100% w/w 100% w/w
water q.s

Table 5.42: In vitro drug release from conventional gel across the dialysis membrane

cumulative
cumulative amt
TIME amt. in amt in amt.
√t logt Abs conc CPR permeated/
(min) t 2ml(µg) 100ml.(µg) release
Unit area
(µg)
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.00
60 7.746 1.778 0.007 0.021 0.042 2.1 2.1 2.10 1.59
120 10.954 2.079 0.007 0.032 0.064 3.2 3.2 3.24 2.46
180 13.416 2.255 0.008 0.037 0.074 3.7 3.8 3.81 2.88
240 15.492 2.380 0.008 0.047 0.094 4.7 4.9 4.88 3.70
300 17.321 2.477 0.008 0.055 0.11 5.5 5.8 5.77 4.37
360 18.974 2.556 0.009 0.06 0.12 6 6.4 6.38 4.84
420 20.494 2.623 0.009 0.062 0.124 6.2 6.7 6.70 5.08
480 21.909 2.681 0.009 0.064 0.128 6.4 7.0 7.03 5.32
540 23.238 2.732 0.009 0.067 0.134 6.7 7.5 7.46 5.65
600 24.495 2.778 0.009 0.067 0.134 6.7 7.6 7.59 5.75

Table 5.43: In vitro drug release from niosomal gel across the dialysis membrane

cumulative cumulative amt


TIME amt. in amt in amt.
√t logt Abs conc CPR
(min) t 2ml(µg) 100ml.(µg) release permeated/
(µg) Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.00
60 7.746 1.778 0.007 0.012 0.024 1.2 1.2 1.20 0.91
120 10.954 2.079 0.008 0.043 0.086 4.3 4.3 4.32 3.28
180 13.416 2.255 0.011 0.1 0.2 10 10.1 10.11 7.66
240 15.492 2.380 0.012 0.13 0.26 13 13.3 13.31 10.08
300 17.321 2.477 0.016 0.22 0.44 22 22.6 22.57 17.10
360 18.974 2.556 0.020 0.3 0.6 30 31.0 31.01 23.49
420 20.494 2.623 0.024 0.39 0.78 39 40.6 40.61 30.77
480 21.909 2.681 0.025 0.43 0.86 43 45.4 45.39 34.39
540 23.238 2.732 0.026 0.45 0.9 45 48.3 48.25 36.55
600 24.495 2.778 0.027 0.46 0.92 46 50.2 50.15 37.99

Table 5.44: In vitro drug release from ethosomal gel across the dialysis membrane

cumulative cumulative amt


TIME amt. in amt in amt.
√t logt Abs conc CPR
(min) t 2ml(µg) 100ml.(µg) release permeated/
(µg) Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.0
60 7.746 1.778 0.017 0.25 0.5 25 25.0 25.00 18.9
120 10.954 2.079 0.024 0.41 0.82 41 41.5 41.50 31.4
180 13.416 2.255 0.029 0.51 1.02 51 52.3 52.32 39.6
240 15.492 2.380 0.030 0.53 1.06 53 55.3 55.34 41.9
300 17.321 2.477 0.030 0.53 1.06 53 56.4 56.40 42.7
360 18.974 2.556 0.030 0.54 1.08 54 58.5 58.46 44.3
420 20.494 2.623 0.033 0.59 1.18 59 64.5 64.54 48.9
480 21.909 2.681 0.033 0.59 1.18 59 65.7 65.72 49.8
540 23.238 2.732 0.034 0.63 1.26 63 70.9 70.90 53.7
600 24.495 2.778 0.035 0.64 1.28 64 73.2 73.16 55.4

Table 5.45: In vitro drug release from transferosomal gel across the dialysis membrane

cumulative cumulative amt


TIME amt. in amt in amt.
√t logt Abs conc CPR
(min) t 2ml(µg) 100ml.(µg) release permeated/
(µg) Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.0
60 7.746 1.778 0.015 0.2 0.4 20 20.0 20.00 15.2
120 10.954 2.079 0.020 0.32 0.64 32 32.4 32.40 24.5
180 13.416 2.255 0.025 0.42 0.84 42 43.0 43.04 32.6
240 15.492 2.380 0.028 0.49 0.98 49 50.9 50.88 38.5
300 17.321 2.477 0.029 0.52 1.04 52 54.9 54.86 41.6
360 18.974 2.556 0.031 0.55 1.1 55 58.9 58.90 44.6
420 20.494 2.623 0.031 0.55 1.1 55 60.0 60.00 45.5
480 21.909 2.681 0.031 0.55 1.1 55 61.1 61.10 46.3
540 23.238 2.732 0.031 0.56 1.12 56 63.2 63.20 47.9
600 24.495 2.778 0.031 0.56 1.12 56 64.3 64.32 48.7

Table 5.46: Ex vivo drug release from conventional gel across the rat skin

cumulative amt
cumulative
TIME amt. in amt in
√t logt Abs conc amt. release CPR
(min) t 2ml(µg) 100ml.(µg) permeated
(µg)
/Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.00
60 7.746 1.778 0.007 0.018 0.036 1.8 1.8 1.80 1.36
120 10.954 2.079 0.007 0.029 0.058 2.9 2.9 2.94 2.22
180 13.416 2.255 0.008 0.037 0.074 3.7 3.8 3.79 2.87
240 15.492 2.380 0.008 0.046 0.092 4.6 4.8 4.77 3.61
300 17.321 2.477 0.008 0.055 0.11 5.5 5.8 5.76 4.36
360 18.974 2.556 0.009 0.062 0.124 6.2 6.6 6.57 4.98
420 20.494 2.623 0.009 0.064 0.128 6.4 6.9 6.89 5.22
480 21.909 2.681 0.009 0.066 0.132 6.6 7.2 7.22 5.47
540 23.238 2.732 0.009 0.067 0.134 6.7 7.5 7.45 5.65
600 24.495 2.778 0.009 0.068 0.136 6.8 7.7 7.69 5.82

Table 5.47: Ex vivo drug release from niosomal gel across the rat skin

cumulative amt
cumulative
TIME amt. in amt in
√t logt Abs conc amt. release CPR
(min) t 2ml(µg) 100ml.(µg) permeated
(µg)
/Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.00
60 7.746 1.778 0.006 0.01 0.02 1 1.0 1.00 0.76
120 10.954 2.079 0.008 0.04 0.08 4 4.0 4.02 3.05
180 13.416 2.255 0.010 0.09 0.18 9 9.1 9.10 6.89
240 15.492 2.380 0.012 0.13 0.26 13 13.3 13.28 10.06
300 17.321 2.477 0.015 0.21 0.42 21 21.5 21.54 16.32
360 18.974 2.556 0.019 0.29 0.58 29 30.0 29.96 22.70
420 20.494 2.623 0.024 0.39 0.78 39 40.5 40.54 30.71
480 21.909 2.681 0.025 0.42 0.84 42 44.3 44.32 33.58
540 23.238 2.732 0.026 0.44 0.88 44 47.2 47.16 35.73
600 24.495 2.778 0.026 0.44 0.88 44 48.0 48.04 36.39

Table 5.48: Ex vivo drug release from ethosomal gel across the rat skin

cumulative amt
cumulative
TIME amt. in amt in
√t logt Abs conc amt. release CPR
(min) t 2ml(µg) 100ml.(µg) permeated
(µg)
/Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.0
60 7.746 1.778 0.017 0.24 0.48 24 24.0 24.00 18.2
120 10.954 2.079 0.025 0.42 0.84 42 42.5 42.48 32.2
180 13.416 2.255 0.028 0.49 0.98 49 50.3 50.32 38.1
240 15.492 2.380 0.029 0.52 1.04 52 54.3 54.30 41.1
300 17.321 2.477 0.029 0.52 1.04 52 55.3 55.34 41.9
360 18.974 2.556 0.030 0.54 1.08 54 58.4 58.38 44.2
420 20.494 2.623 0.032 0.58 1.16 58 63.5 63.46 48.1
480 21.909 2.681 0.032 0.58 1.16 58 64.6 64.62 49.0
540 23.238 2.732 0.034 0.62 1.24 62 69.8 69.78 52.9
600 24.495 2.778 0.034 0.62 1.24 62 71.0 71.02 53.8

Table 5.49: Ex vivo drug release from transferosomal gel across the rat skin

cumulative amt
cumulative
TIME amt. in amt in
√t logt Abs conc amt. release CPR
(min) t 2ml(µg) 100ml.(µg) permeated
(µg)
/Unit area
0 0.000 0.000 0.000 0 0 0 0.0 0.00 0.0
60 7.746 1.778 0.015 0.19 0.38 19 19.0 19.00 14.4
120 10.954 2.079 0.020 0.31 0.62 31 31.4 31.38 23.8
180 13.416 2.255 0.025 0.42 0.84 42 43.0 43.00 32.6
240 15.492 2.380 0.028 0.48 0.96 48 49.8 49.84 37.8
300 17.321 2.477 0.029 0.51 1.02 51 53.8 53.80 40.8
360 18.974 2.556 0.030 0.53 1.06 53 56.8 56.82 43.0
420 20.494 2.623 0.030 0.53 1.06 53 57.9 57.88 43.8
480 21.909 2.681 0.031 0.55 1.1 55 60.9 60.94 46.2
540 23.238 2.732 0.031 0.55 1.1 55 62.0 62.04 47.0
600 24.495 2.778 0.031 0.55 1.1 55 63.1 63.14 47.8

Table 5.50: Nystatin retention from all gels during ex vivo drug release study in and on skin
Concentrations, µg/ml
Area
Conventional gel Niosomal gel Ethosomal gel Transferosomal gel

Drug permeated
4.3 28.6 16.3 21.9
in to the skin layers
Drug permeated in to the
5.8 36.4 53.8 47.8
receptor fluid

drug retained on the skin 89.9 35 29.9 30.3


Table 5.51: Stability studies of optimized niosomal formulation gel (FN3)

S.No Refrigerated temperature Room temperature


Formulation
FN3 Vesicles Vesicle size Vesicles Vesicle
% EE % EE
shape (nm) shape size (nm)

1 Freshly prepared Spherical 281±1.7 72.5±1.9 Spherical 281±1.7 72.5±1.9

2 After 3 month Spherical 282±1.2 71.9±1.6 Spherical 288±1.4 70.2±1.7

3 After 6 month Spherical 285±1.3 71.3±1.2 Spherical 290±2.7 67±2.3

Table 5.52: Stability studies of optimized ethosomal formulation gel (FE6)

Refrigerated temperature Room temperature


Formulation
S.No
FE6 Vesicles Vesicle Vesicles Vesicle
% EE % EE
shape size (nm) shape size (nm)

1 Freshly prepared Spherical 403.3 88.4 Spherical 403.3 88.4

2 After 3 month Spherical 414.5 85.2 Spherical 416.8 84.2

3 After 6 month Spherical 438.1 82.8 Spherical 485.2 71.1

Table 5.53: Stability studies of optimized transferosomal formulation gel (FT7)


Refrigerated temperature Room temperature
Formulation
S.No
FT7 Vesicle Vesicle
Vesicles shape % EE Vesicles shape % EE
size (nm) size (nm)

Freshly Approximately Approximately


1 374.1 70 374.1 70
prepared Spherical shape Spherical shape

After 3 Approximately Approximately


381.6 68.4 391.9 65.7
month Spherical shape Spherical shape

After 6 Approximately Approximately


2 419.3 62.8 446.2 59.4
month Spherical shape Spherical shape
Table 5.54: Analysis of ex vivo drug release study of three optimized
vesicular formulations

Kp,
S.No Nystatin gels Slope Area Jss sq.cm/hr Er
1 Conventional gel 0.948 1.32 0.72 0.007
2 Niosomal gel 2.297 1.32 1.74 0.017 2.4
3 Transferosomal gel 10.71 1.32 8.11 0.081 11.3
4 Ethosomal gel 12.83 1.32 9.72 0.097 13.5

Table 5.55: Calibration curve of the nystatin by HPLC

S.No Concentration, ng/ml Area of the peak


1 0 0
2 300 214265
3 600 428531
4 900 642797
5 1200 857063
6 1500 1071328
7 1800 1285594
8 2100 1499860
Table 5.56: In vivo study of the conventional gel in rabbits

Rabbit -I Rabbit-II Concentration,


Time, Avearge
Hrs Isomer -a Isomer-b Total area Isomer -a Isomer-b Total area area ng/ml
0 0 0 0 0 0 0 0 0
1 7755 5170 12925 7158 4772 11930 12427 17.40
3 9404 6269 15673 8680 5787 14467 15070 21.10
5 9092 6061 15153 8392 5595 13987 14570 20.40
7 9003 6002 15004 8310 5540 13850 14427 20.20
9 8869 5913 14781 8187 5458 13644 14213 19.90
11 8334 5556 13890 7693 5129 12822 13356 18.70
24 6373 4249 10622 5883 3922 9805 10213 14.30

Table 5.57: In vivo study of the niosomal gel in rabbits

Rabbit -I Rabbit-II Avearge Concentration,


Time,
Hrs Isomer -a Isomer-b Total area Isomer -a Isomer-b Total area area ng/ml
0 0 0 0 0 0 0 0 0
1 39531 26354 65885 36490 24327 60817 63351 88.70
3 194670 129780 324450 179695 119797 299492 311971 436.81
5 196274 130850 327124 181176 120784 301961 314542 440.41
7 149702 99801 249503 138186 92124 230310 239906 335.91
9 148498 98999 247497 137075 91384 228459 237978 333.21
11 53614 35743 89357 49490 32994 82484 85921 120.30
24 25225 16817 42042 23285 15523 38808 40425 56.60
Table 5.58: In vivo study of the ethosomal gel in rabbits

Rabbit -I Rabbit-II Avearge Concentration,


Time,
Hrs Isomer -a Isomer-b Total area Isomer -a Isomer-b Total area area ng/ml
0 0 0 0 0 0 0 0 0
1 132008 88006 220014 121854 81236 203090 211552 296.21
3 306133 204088 510221 282584 188389 470973 490597 686.92
5 573759 382506 956266 529624 353083 882707 919486 1287.44
7 757466 504977 1262443 699199 466133 1165332 1213887 1699.65
9 783983 522656 1306639 723677 482451 1206128 1256383 1759.15
11 761120 507413 1268534 702573 468382 1170954 1219744 1707.85
24 144665 96444 241109 133537 89025 222562 231836 324.61

Table 5.59: In vivo study of the transferosomal gel in rabbits

Rabbit -I Rabbit-II Concentration,


Time, Avearge
Hrs Isomer -a Isomer-b Total area Isomer -a Isomer-b Total area area ng/ml
0 0 0 0 0 0 0 0 0
1 87486 58324 145809 80756 53837 134593 140201 196.31
3 221143 147429 368572 204132 136088 340220 354396 496.21
5 483421 322281 805702 446235 297490 743725 774714 1084.73
7 636777 424518 1061296 587795 391863 979658 1020477 1428.84
9 663696 442464 1106160 612643 408428 1021071 1063616 1489.24
11 645468 430312 1075780 595817 397211 993028 1034404 1448.34
24 88377 58918 147295 81579 54386 135965 141630 198.31
Table 5.60: Summary of the nystatin concentrations in blood plasma at
different time intervals (In vivo study)

Concentration of nystatin, ng/ml


Time,Hrs Conventional gel Niosomal gel Transferosomal gel Ethosomal gel
0 0 0 0 0
1 17.4 88.7 196.3 296.2
3 21.1 436.8 496.2 686.9
5 20.4 440.4 1084.7 1287.4
7 20.2 335.9 1428.8 1699.6
9 19.9 332.2 1489.2 1759.1
11 18.7 120.3 1448.3 1707.8
24 14.3 56.6 198.3 324.6

Table 5.61: Bioavailability studies of nystatin from all gels

Formulations Cmax, ng/ml Tmax, Hrs AUC, ngh/ml

Conventional gel 21.9 3 589.21

Niosomal gel 442.8 5 1092.7

Transferosomal gel 1471.6 9 2136.3

Ethosomal gel 1759.1 9 2449.2

Figure 5.1: Absorption spectrum of Nystatin between 200-400nm


Figure 5.2: FTIR graph of Nystatin

FTIR showed the characteristic peak for NH, OH stretch – 3350-3331cm-1, Lactone carbonyl stretch-
1701cm-1, Carboxylate ion –1566-1546cm-1, C-OH stretch-1068cm-1 (Fig.2)
Figure 5.3: Standard curve of nystatin in PBS pH 7.4
Figure 5.4: SEM images of the nystatin loaded niosomes
Figure 5.5: In vitro drug release from niosomal preparations prepared
using span-60

Figure 5.6: In vitro drug release from niosomal preparations prepared


using span-80
Figure 5.7: Release kinetics of niosomal suspension FN1
Figure 5.8: Release kinetics of niosomal suspension FN2
Figure 5.9: Release kinetics of niosomal suspension FN3
Figure 5.10: Release kinetics of niosomal suspension FN4
Figure 5.11: Release kinetics of niosomal suspension FN5
Figure 5.12: Release kinetics of niosomal suspension FN6
Figure 5.13: Release kinetics of niosomal suspension FN7
Figure 5.14: Release kinetics of niosomal suspension FN8
Figure 5.15: SEM Photographs of ethosomes
Figure 5.16: Effect of ethanol concentration on drug release from
ethosomes

Figure 5.17: Effect of lecithin concentration on drug release from


ethosomes.
Figure 5.18: Effect of cholesterol on drug release from ethosomes
Figure 5.19: Release kinetics of ethosomal formulation FE1
Figure 5.20: Release kinetics of ethosomal formulation FE2
Figure 5.21: Release kinetics of ethosomal formulation FE3
Figure 5.22: Release kinetics of ethosomal formulation FE4
Figure 5.23: Release kinetics of ethosomal formulation FE5
Figure 5.24: Release kinetics of ethosomal formulation FE6
Figure 5.25: Release kinetics of ethosomal formulation FE7
Figure 5.26: Release kinetics of ethosomal formulation FE8
Figure 5.27: SEM photographs of transferosomes
Figure 5.28: In vitro drug release from transferosomal formulations
prepared using span - 60

Figure 5.29: In vitro drug release from transferosomal formulations


prepared using span - 80
Figure 5.30: Release kinetics of transferosomal formulation FT1
Figure 5.31: Release kinetics of transferosomal formulation FT2
Figure 5.32: Release kinetics of transferosomal formulation FT3
Figure 5.33: Release kinetics of transferosomal formulation FT4
Figure 5.34: Release kinetics of transferosomal formulation FT5
Figure 5.35: Release kinetics of transferosomal formulation FT6
Figure 5.36: Release kinetics of transferosomal formulation FT7
Figure 5.37: Release kinetics of transferosomal formulation FT8
Figure 5.38: Zeta potential measurement of nystatin loaded niosomes of
an optimized formulation (FN3)
Figure 5.39: Zeta potential measurement of nystatin loaded ethosomes of
an optimized formulation (FE6)
Figure 5.40: Zeta potential measurement of nystatin loaded
transferosomes of an optimized formulation (FT7)
Figure 5.41: In vitro drug release from all gels across the dialysis
membrane

Figure 5.42: Ex vivo drug release from all gels across the rat skin
Figure 5.43: calculation of flux at steady state and other parameters from ex vivo drug release data
Figure 5.44: Calibration curve of nystatin by HPLC

Figure 5.45: In vivo drug release from all gels


Figure 5.46: HPLC chromatogram of nystatin (300ng/ml)
Figure 5.47: HPLC chromatogram of nystatin (600ng/ml)
Figure 5.48: HPLC chromatogram of nystatin (900ng/ml)
Figure 5.49: HPLC chromatogram of nystatin (1200ng/ml)
Figure 5.50: HPLC chromatogram of nystatin (1500ng/ml)
Figure 5.51: HPLC chromatogram of nystatin (1800ng/ml)
Figure 5.52: HPLC chromatogram of nystatin (2100ng/ml)
Figure 5.53: Conventional gel HPLC chromatograms of 0th hour blood sample in
Rabbit -1

Figure 5.54: Conventional gel HPLC chromatograms of 0 th hour blood sample in Rabbit -2
Figure 5.55: Conventional gel HPLC chromatograms of 1 st hour blood sample in Rabbit -1
Figure 5.56: Conventional gel HPLC chromatograms of 1 st hour blood sample in Rabbit -2
Figure 5.57: Conventional gel HPLC chromatograms of 3 rd hour blood sample in Rabbit -1
Figure 5.58: Conventional gel HPLC chromatograms of 3 rd hour blood sample in Rabbit -2
Figure 5.59: Conventional gel HPLC chromatograms of 5 th hour blood sample in Rabbit -1
Figure 5.60: Conventional gel HPLC chromatograms of 5 th hour blood sample in Rabbit -2
Figure 5.61: Conventional gel HPLC chromatograms of 7 th hour blood sample in Rabbit -1
Figure 5.62: Conventional gel HPLC chromatograms of 7 th hour blood sample in Rabbit -2
Figure 5.63: Conventional gel HPLC chromatograms of 9 th hour blood sample in Rabbit -1
Figure 5.64: Conventional gel HPLC chromatograms of 9 th hour blood sample in Rabbit -2
Figure 5.65: Conventional gel HPLC chromatograms of 11 th hour blood sample in Rabbit -1
Figure 5.66: Conventional gel HPLC chromatograms of 11 th hour blood sample in Rabbit -2
Figure 5.67: Conventional gel HPLC chromatograms of 24 th hour blood sample in Rabbit -1
Figure 5.68: Conventional gel HPLC chromatograms of 24 th hour blood sample in Rabbit -2
Figure 5.69: Niosomal gel HPLC chromatogram of nystatin at 0 th hr blood sample in rabbit - 1
Figure 5.70: Niosomal gel HPLC chromatogram of nystatin at 0 th hr blood sample in rabbit - 2
Figure 5.71: Niosomal gel HPLC chromatogram of nystatin at 1 st hr blood sample in rabbit - 1
Figure 5.72: Niosomal gel HPLC chromatogram of nystatin at 1 st hr blood sample in rabbit - 2
Figure 5.73: Niosomal gel HPLC chromatogram of nystatin at 3 rd hr blood sample in rabbit - 1
Figure 5.74: Niosomal gel HPLC chromatogram of nystatin at 3 rd hr blood sample in rabbit - 2
Figure 5.75: Niosomal gel HPLC chromatogram of nystatin at 5 th hr blood sample in rabbit - 1
Figure 5.76: Niosomal gel HPLC chromatogram of nystatin at 5 th hr blood sample in rabbit - 2
Figure 5.77: Niosomal gel HPLC chromatogram of nystatin at 7 th hr blood sample in rabbit - 1
Figure 5.78: Niosomal gel HPLC chromatogram of nystatin at 7 th hr blood sample in rabbit - 2
Figure 5.79: Niosomal gel HPLC chromatogram of nystatin at 9 th hr blood sample in rabbit - 1
Figure 5.80: Niosomal gel HPLC chromatogram of nystatin at 9 th hr blood sample in rabbit - 2
Figure 5.81: Niosomal gel HPLC chromatogram of nystatin at 11 th hr blood sample in rabbit - 1
Figure 5.82: Niosomal gel HPLC chromatogram of nystatin at 11 th hr blood sample in rabbit - 2
Figure 5.83: Niosomal gel HPLC chromatogram of nystatin at 24 th hr blood sample in rabbit - 1
Figure 5.84: Niosomal gel HPLC chromatogram of nystatin at 24 th hr blood sample in rabbit - 2
Figure 5.85: Ethosomal gel HPLC chromatogram of nystatin at 0 th hr blood sample in rabbit - 1
Figure 5.86: Ethosomal gel HPLC chromatogram of nystatin at 0 th hr blood sample in rabbit - 2
Figure 5.87: Ethosomal gel HPLC chromatogram of nystatin at 1 st hr blood sample in rabbit - 1
Figure 5.88: Ethosomal gel HPLC chromatogram of nystatin at 1 st hr blood sample in rabbit - 2
Figure 5.89: Ethosomal gel HPLC chromatogram of nystatin at 3 rd hr blood sample in rabbit - 1
Figure 5.90: Ethosomal gel HPLC chromatogram of nystatin at 3 rd hr blood sample in rabbit - 2
Figure 5.91: Ethosomal gel HPLC chromatogram of nystatin at 5 th hr blood sample in rabbit - 1
Figure 5.92: Ethosomal gel HPLC chromatogram of nystatin at 5 th hr blood sample in rabbit - 2
Figure 5.93: Ethosomal gel HPLC chromatogram of nystatin at 7 th hr blood sample in rabbit - 1
Figure 5.94: Ethosomal gel HPLC chromatogram of nystatin at 7 th hr blood sample in rabbit - 2
Figure 5.95: Ethosomal gel HPLC chromatogram of nystatin at 9 th hr blood sample in rabbit - 1
Figure 5.96: Ethosomal gel HPLC chromatogram of nystatin at 9 th hr blood sample in rabbit - 2
Figure 5.97: Ethosomal gel HPLC chromatogram of nystatin at 11 th hr blood sample in rabbit - 1
Figure 5.98: Ethosomal gel HPLC chromatogram of nystatin at 11 th hr blood sample in rabbit - 2
Figure 5.99: Ethosomal gel HPLC chromatogram of nystatin at 24 th hr blood sample in rabbit - 1
Figure 5.100: Ethosomal gel HPLC chromatogram of nystatin at 24 th hr blood sample in rabbit - 2
Figure 5.101: Transferosomal gel HPLC chromatogram of nystatin at 0 th hr blood sample in rabbit - 1
Figure 5.102: Transferosomal gel HPLC chromatogram of nystatin at 0 th hr blood sample in rabbit - 2
Figure 5.103: Transferosomal gel HPLC chromatogram of nystatin at 1 st hr blood sample in rabbit - 1
Figure 5.104: Transferosomal gel HPLC chromatogram of nystatin at 1 st hr blood sample in rabbit - 2
Figure 5.105: Transferosomal gel HPLC chromatogram of nystatin at 3 rd hr blood sample in rabbit - 1
Figure 5.106: Transferosomal gel HPLC chromatogram of nystatin at 3 rd hr blood sample in rabbit - 2
Figure 5.107: Transferosomal gel HPLC chromatogram of nystatin at 5 th hr blood sample in rabbit - 1
Figure 5.108: Transferosomal gel HPLC chromatogram of nystatin at 5 th hr blood sample in rabbit - 2
Figure 5.109: Transferosomal gel HPLC chromatogram of nystatin at 7 th hr blood sample in rabbit - 1
Figure 5.110: Transferosomal gel HPLC chromatogram of nystatin at 7 th hr blood sample in rabbit - 2
Figure 5.111: Transferosomal gel HPLC chromatogram of nystatin at 9 th hr blood sample in rabbit - 1
Figure 5.112: Transferosomal gel HPLC chromatogram of nystatin at 9 th hr blood sample in rabbit - 2
Figure 5.113: Transferosomal gel HPLC chromatogram of nystatin at 11 th hr blood sample in rabbit - 1
Figure 5.114: Transferosomal gel HPLC chromatogram of nystatin at 11 th hr blood sample in rabbit - 2
Figure 5.115: Transferosomal gel HPLC chromatogram of nystatin at 24 th hr blood sample in rabbit - 1
Figure 5.116: Transferosomal gel HPLC chromatogram of nystatin at 24 th hr blood sample in rabbit - 2
Chapter - 6
DISCUSSION ON RESULTS
Chapter - 6: DISCUSSION ON RESULTS

Table of contents

S.No Name of the subtitle Page No


6.1 Morphology of vesicles (size, shape and texture)  
6.2 Entrapment efficiency  
6.3 In vitro drug release  
6.4 Release kinetics  
6.5 Zeta potential  
6.6 Elasticity measurement  
6.7 In vitro and ex vivo drug release  
6.8 Skin retention study  
6.9 Stability study  
6.10 In vivo study  
6. DISCUSSION ON RESULTS

6.1. Morphology of vesicles (size, shape and texture)

The shape and size of the vesicles present in all formulations was

determined by using optical microscope. All vesicles appeared spherical shape.

But, transfersomes appeared not spherical as much as niosomes and

ethosomes. The surface of the vesicles was determined by SEM analysis.

Niosomes and transferosomes appeared with rough surface and ethosomes

appeared with smooth surface.

The average size of niosomes was found in the range of 278±1.4 to

431±1.2nm. The size of vesicles was decreased with increase in the

concentration of surfactant. This may due to decrease in surface tension with

increase in the concentration of surfactant.

The average size of ethosomes present in all formulations was found in

the range of 291.2 – 463.2nm. The size of the ethosomes was inversely

proportional to the concentration of ethanol. Vesicles size was decreased with

an increase in the concentration of ethanol from 15- 60%. This is may be due

to the modification in net charge of the system by the ethanol and some degree

of stearic stabilization.

The average size of the transferosomes present in all formulations was

found in the range of 266.7 – 374.1nm. The size of the vesicles in

transferosomal formulations prepared using span – 60 is less than the vesicles

present in transferosomal formulations prepared using span – 80. This is


because the size of the vesicles depends on the liposphilicty of the edge

activators. i.e. the size of the vesicles is influencing by their HLB values. The

particle size is inversely proportional to the HLB value of the surfactant. The

reason behind to this concept is that there is increase in surface free energy

with increasing the lipophilicity of surfactants. The surface free energy will

increase due to the interaction of the edge activators with the lipid head groups

present in the membrane which leads increase in packing density of the layer.

This increased surface free energy causes fusion between lipid bilayers and

also increase in size of the vesicles.

The size of the vesicles deceased with increase in the concentration of the

surfactant which may be due to the decrease in surface tension with increase

in concentration of surfactant. Among all formulations the vesicles size was

found asfollows Ethosomes > Transferosomes > Niosomes

6.2. Entrapment efficiency

Entrapment efficiency in niosomal formulation was observed that

the percentage entrapment efficiency increased with increase in surfactant

concentration. Entrapment efficiency is more in case of span-60 niosomes

rather than the niosomes prepared using span-80. This may be due to that the

Span-80 has the lowest transition temperature (Tc = -12ºC) and span-60 has

high transition temperature (Tc =53ºC).

In ethosomal formulations the Entrapment efficiency of Nystatin was

increased with an increase in ethanol concentration up to 45% and on further

increasing the concentration of ethanol, entrapment efficiency was decreased.


Increased entrapment of Nystatin with an increase in concentration of ethanol

owing to increase in fluidity of membranes and increased solubility of Nystatin

in ethanol. On increasing the concentration of ethanol further, the entrapment

efficiency was decreased. This may due to the leakage of the drug from bilayer

of the vesicles.

EE was also increased with an increase in the concentration of lipid to

certain extent. But after specific concentration, the percent EE was decreased.

This may be due to the saturation effect on increase in solubility of Nystatin in

lipid.

Formulations prepared with span – 80 have shown more entrapment

efficiency than the formulations prepared using span – 60. The influential

factor on the entrapment efficiency of nystatin in the transferosomes may be

the lipophilicity of the edge activators (span – 60 and span – 80).

Span – 80 is more lipophilic than the span – 60 than the span – 80. The

HLB values also exhibiting the same that the span – 60 (HLB value: 4.7) is less

liposphilic than the span – 80 (HLB value: 4.3). As nystatin is lipophilic in

nature, it is more soluble in the span – 80 than the span – 60. Hence the

nystatin has entrapped more in the lipid bilayers of the span – 80 than the

lipid bilayers of the span – 60 in transferosomes.

All the transferosomal formulations prepared using span – 80 and span –

60 showed increased entrapment efficiency with increased concentration of

edge activators, initially. Later on further increasing the concentration of edge

activators leads to decrease in the entrapment efficiency. This is may be due to


that, at initial the edge activators (monomers) may be associated with lipid

bilayers of the transferosomes and on increasing the concentration of edge

activators the association of monomers with lipid bilayers may increased which

increases the partitioning of the drug and increases in the entrapment

efficiency. On further increasing the concentration of the edge activators, the

association of edge activators with lipid bilayers may be saturated and some

amount of edge activators left in bulk solution. The unassociated monomers

might begin formation of micelles in lipid bilayers of the transferosomes which

results in the formation of pores in transferosomes and finally bilayers

conversion will takes place to mixed micelles. These mixed micelles are not able

to accommodate more quantity of the drug and are not capable to penetrate the

deeper layers of the skin because of their structural features.

6.3. In vitro drug release

In vitro diffusion studies of all formulations were carried out in fabricated

diffusion cell. The drug release from niosomes decreases with increase in the

concentration of surfactant and also from the data we shall conclude that

niosomal formulations prepared using Span 60 yielded a lower rate of drug

release compared to Span 80 niosomes. This can be explained by the fact that

niosomes exhibit an alkyl chain length-dependent release. The higher the chain

length, the lower the release rate. The nystatin release from FN4 formulation is

highly sustained than that of other niosomal preparations.

In ethosomal formulations, the release of the drug from vesicles was

increased as the concentration of ethanol increases. This may be due to


increased fluidity of the bilayers of the vesicles with increased concentration of

ethanol. However drug release from vesicles was decreased as the

concentration of lecithin increased above 300mg, due to decrease in the bilayer

malleability and deformability and increased rigidity of vesicles. Effect of

incorporation of cholesterol on drug release was also studied and was found

that decreased drug release with addition of cholesterol. This may be due to

the increased rigidity of the vesicle bilayer.

In vitro drug release from all transferosomal formulations showed

increased drug release with increase concentration of edge activators at lower

concentrations. On further increase in the concentration of edge activators the

drug release was decreased. This was observed in all formulations prepared

using both edge activators. This may be due to fact that the transferosomes

lipid bilayers are more ordered and less leaky at low concentration of edge

activators which results in less drug release. Whereas on increasing the

concentration of edge activators slowly, the drug release has increased. This

may be due to the increased deformability of the bilayers. On further increasing

the concentration of edge activators, the vesicle may lose their structure and

also may be due to the formation of mixed micelles which results in decrease in

drug release.

6.4. Release kinetics

All ethosomal, niosomal and transferosomal formulations in vitro drug

release data fitted in various release kinetic models and they showed that all

formulations following zero order drug release. The r 2 values of higuchi and
hixon crowel models showed that drug release from all formulations following

diffusion rather than the dissolution. This was further confirmed by fitting the

in vitro drug release data in Korsmeyer-Peppas model. The r 2 values of

Korsmeyer-Peppas model also showed the same. The n values of Korsmeyer-

Peppas model (n>0.5) showed that niosomes and transferosomal formulations

following fickian diffusion.

But the n-value in ethosomal formulation was found between 0.48-0.95

except FT4 formulation (0.37). this indicates that the ethosomal formulations

following non-fickian diffusion and zero order release.

Based on the entrapment efficiency, in vitro drug release and release

mechanism, one optimized formulation i.e. FN3, FE6 and FT7 from all

niosomal, ethosomal and transferosomal suspension was selected respectively.

The selected optimized suspensions were subjected to zeta potential and

elasticity measurements.

6.5. Zeta potential

The selected optimized formulations FN3, FE6 and FT7 was shown 24.2,

-38.1 and -20.0 mV respectively which substantiating that the ethosomal

formulations are more stable than other two vesicles. i.e. niosomes and

transferosomes. The stability patterns of all the vesicles are as follows based on

the zeta potential.

Ethosomes > Niosomes > Transferosomes

6.6. Elasticity measurement


The elasticity of the lipid/surfactant bilayers of the niosomes, ethosomes

and transferosomes present in three optimized vesicular suspensions was

measured and was found to be 45.2±1.7, 79.6±6.9, 113.0±7.0 nm.ml/µm

respectively. The results stated that the transferosomes more elastic than

others two vesicles followed by ethosomes. Least elasticity was exhibited by the

niosomes.

6.7. In vitro and ex vivo drug release

The nystatin release from skin was lesser than the dialysis membrane

which may be due to the complexicity of skin membrane. But, there is a good

correlation between the in vitro and ex vivo release of nystatin from all gels.

The ex vivo drug release data was analyzed for transdermal flux,

permeation co-efficient and enhancement ratio. The transdermal flux at steady

state for transferosomal, ethosomal, niosomal and conventional gel was found

to be 8.11, 9.72, 1.74 and 0.72 Jss, μg/cm2/min respectively. The permeation

co-efficient of transferosomal, ethosomal, niosomal and conventional gel was

found to be 0.081, 0.097, 0.017 and 0.007 sq.cm/hr respectively. The extent of

drug permeation from vesicular gels was compared with the drug permeation

from conventional gel was measured by calculating the enhancement ratio. The

enhancement ratio of niosomal, transferosomal and ethosomal gels was found

to be 2.4, 11.3 and 13.5 respectively. The effectiveness of the vesicles based on

the above data concluded as follows for administration of drugs across the

skin.

Ethosomal gel > Transferosomal gel > Niosomal gel > Conventional gel
6.8. Skin retention study

The skin retention study revealed that more amount of drug retained on

the skin in case of conventional gel and highest amount of nystatin permeated

from ethosomal gel. In ethosomal gel, less amount of drug retained on the skin

as well as within the skin. This may be due to the optimized flexibility of the

vesicle lipid bilayers. In case of transferosomes less quantity of the nystatin

retained on the skin and less quantity of the drug permeated into the systemic

circulation when compared with ethosomes but more amount of drug retained

within the skin in case of transferosomes than the ethosomes. This may be due

to the fact that the excessive flexibility of transferosomes. Due to high flexibility

of the lipid bilayers of transferosomes, they penetrated easily into the skin than

the ethosomes and may undergo rapture within the skin while penetrating into

the deeper layers of the skin. This may be the reason for accumulating more

quantity of the nystatin with in the skin in transferosomes than the ethosomes.

This was also confirmed in elasticity measurements of the vesicles. In niosomal

gel, neither nystatin permeated into the systemic circulation nor into the skin.

But, when compared with conventional gel, niosomal gel permeated more

quantity of the nystatin into the systemic circulation.The study revealed that

the extent of drug release in to the systemic circulation is as follows

Ethosomal gel > Transferosomal gel > Niosomal gel > Conventional gel

6.9. Stability study

All prepared gels were subject stability studies at room and refrigerated

temperatures for 6 months. The study was shown that there is no change in
the vesicles shape in all gels after 3 rd and 6th months also at both temperatures.

The change in the size and entrapment efficiency of the vesicles in all gels was

negligible after 6th month when compared with initial size and entrapment

efficiency at refrigerated temperature. But the variation in the size and

entrapment efficiency of vesicles in all gels at room temperature was more

when compared with the change that has been taken place at refrigerated

temperature.

If on comparison of the change that has taken place in size and

entrapment efficiency of the vesicles, within the gels, the change was as

follows.

Transferosomes > Ethosomes > Niosomes

The reason may be for this is their bilayer flexibility. The study was

shown that the stability is inversely proportional to the flexibility of the

bilayers.

6.10. In vivo study

The in vivo study and HPLC analysis revealed that the ethosomes are

prominent vesicular carrier system for enhanced transdermal delivery of

nystatin as it has shown highest C max (1759ng.ml) and AUC 2449.2 ngh/ml

than other gels. The study was shown that the in vivo performance of all gels

was as follows.

Ethosomal gel > Transferosomal gel > Niosomal gel > Conventional gel.
Chapter - 7

SUMMARY, CONCLUSION AND

RECOMMENDATIONS
7. SUMMARY, CONCLUSION AND RECOMMENDATIONS

The present research work aimed to select the best vesicular carrier

system among niosomes, ethosomes and transferosomes (primary objective) for

administration of drugs across the skin to produce systemic effect. Nystatin, a

polyene antifungal agent, was used as model drug in the present study. The

secondary objective of the study is enhancement of bioavailability of the

nystatin and to decrease the adverse effects associated with higher doses of the

same by administering through skin.

Niosomal and transferosomal suspensions of nystatin prepared by using

thin film hydration Technique whereas ethosomal suspensions of nystatin was

prepared method reported by Touitou et al. Totally 24 vesicular suspensions

(eight formulations from each type of vesicular systems) were prepared.

Niosomal suspensions were prepared using two different non-ionic

surfactants i.e span-80 and span-60. Each surfactant used at four different

concentrations (eight niosomal suspensions) was prepared. Similarly, eight

transferosomal and eight ethosomal suspensions were prepared.

Transferosomal suspensions were prepared using two different edge activators

i.e. span-60 and span-80 at four different concentrations whereas in

preparation of ethosomal suspensions ethanol and lecithin concentrations has

been changed for preparing the same (each at four different concentrations).

The prepared niosomal, ethosomal and transferosomal suspensions (24

formulations) evaluated for different parameters like vesicles morphology (size,


shape and surface texture), entrapment efficiency and in vitro drug release. The

drug release data fitted in different mathematical models such as Zero order,

First order, Higuchi, Hixon-crowel, Korsmeyer-peppas to find out the order and

mechanism of drug release from all formulations.

Based on the size, entrapment efficiency and invitro drug release results,

one optimized vesicular suspension from each type of vesicular formulation

was selected.

Elasticity and surface charge (Zeta potential) of the vesicles present in

three optimized vesicular suspensions was studied.

After performing the zetapotential and elasticity studies, using the

optimized niosomal, ethosomal and transferosomal suspensions niosomal,

ethosomal and transfersosmal gels having concentration of 0.01%w/w was

prepared respectively. Along with three vesicular nystatin gels, conventional

nystatin gel having same concentration was also prepared with pure nystatin

sample.

The prepared gels were characterized for in vitro and ex vivo drug release,

skin retention, stability studies. Finally in vivo studies also carried out to find

out the systemic availability of nystatin from individual gels in rabbits.

In vitro, ex vivo drug release data showed a good correlation between

each other. The study revealed that ethosomes are effective for permeation of

nystatin across the dialysis membrane and skin than other two vesicles.

Transdermal flux, permeation coefficient and enhancement ratio was

calculated from ex vivo drug release data of all three vesicular carrier gels. The
data showed higher efficiency to the ethosomes than other two vesicular gels.

Skin retention study was performed to find out the quantity of nystatin that

reached systemic circulation and retained in and on the skin after ex vivo drug

release study. The skin retention study also showed that ethosomes are

efficient than other two vesicular carriers i.e. transferosomes and niosomes.

In vivo study was also carried out in rabbits to find out the efficiency of

all vesicular gels and conventional gel in production of systemic effect. Highest

Cmax and AUC was exhibited by the ethosomes than other two vesicular and

conventional gels which stating that ethosomes are effective carriers to produce

systemic effect than other two carriers.

Our findings contribute to the evidence base for enhancing the

permeability of nystatin across the skin and to produce the systemic anti

fungal activity. (Secondary objective)

The present study demonstrates the utility of ethosomes as a tool for

administration of drugs across the skin to produce the systemic effect and

further studies such as clinical trials are required to conclude the same.
REFERENCES
REFERENCES

1. Sylvia Miriyam Findlay. Drug Delivery Markets - An Outlook, 3 rd

edition, Frost & Sullivan Market Insight, 2008, 111-113.

2. Monkhouse, D.C.; and Huq, A.S.; Drug development and industrial

pharmacy. 14,1988,183-188.

3. Stan-Posthumd,J.J.; Vink, J.Lecessies and Bruijn, J. A.; Asian

Journal of Pharmacy and Clinical Research. 54(3), 1988, 1342-1346.

4. Lyn Margetts,; and Richard Sawyer.; Continuing Education in

Anaesthesia, Critical Care & Pain, 7 (5), 2007, 171-176.

5. Mark R. Prausnitz.; and Robert Langer.; Nat Biotechnol. 26 (11),

2008, 1261–1268.

6. Paul A.J. Kolarsick.; and Maria Ann Kolarsick B.S. Anatomy and

Physiology of the Skin. Andrews’ Diseases of the Skin: Clinical

Dermatology, 10th edition, Phila-delphia, 2006, 1-121.

7. Vikas Chander Jhawat.; Vipin Saini.; Sunil Kamboj.; and Nancy

Maggon.; Int. J. Pharm. Sci. Rev. Res., 20(1), 2013, 47-56.

8. Higuchi .T.; J Soc Cos Chem. 11, 1960, 85-97.

9. Valenta. C.; Siman. U.; and Kratzel. M.; Int J Pharm. 97 (3), 2000,

77–85.

10. Stott, P.W.; Williams, A.C.; and Barry, B.W.; J Control Release. 50,

1998, 297–308.
11. Woolfson, A.D et al.; J Control Release. 67, 2000, 395–408.

12. Stott, P.W.; Williams, A.C.; and Barry, B.W.; Int J Pharm. 219, 2001,

161– 176.

13. Kang, L.; Jun, H.W.; and McCall, J.W.; Int J Pharm. 206, 2000, 35–

42.

14. Behl, C.R.; et al.; J. Invest. Dermatol. 75, 1980, 346-52.

15. McKenzie, A.W.; Stoughton, R.B.; Arch. Dermatol.86, 1962, 608- 610.

16. Scheuplein, R.J.; Blank, I.H.; J. Invest Dermatol.60, 1973, 286-296.

17. Roberts, M.S.; Walker, M. Pharmaceutical Skin Penetration

Enhancement, 4th Edition. Marcel Dekker, 1993, 1-30.

18. Prausnitz, M.R.; Mitragotri, S.; and Langer, R.; Nat Rev Drug Discov.

3, 2004, 115-124.

19. Finnin, B.C.; and Morgan, T.M.; J. Pharm. Sci. 88, 1999, 955-958.

20. Santus, G.C.; and Baker, R.W.; J Control Release. 25, 1993,1-20.

21. Osborne, D.W.; et al. Pharm. Tech.21, 1997, 58-66.

22. Barry, B.W.; Dermatological formulation: Percutaneous absorption. 7 th

Edition, Marcel Dekker, New York, 1983, 22-56.

23. Kligman,A.M.; J Am Med Assoc. 193, 1965, 796-804.

24. Southwell, D.; and Barry, B.W.; J Invest Dermatol. 82, 1984, 507-

515.

25. Barry, B.W.; Southwell, D.; and Woodford, R.; J Invest Dermatol. 82,

1984, 49-52.
26. Bennett, S.L.; Barry, B.W.; and Woodford, R.; J Pharm Pharmacol. 37,

1984, 294- 304.

27. Hori, M et al.; J. Pharm. Sci.80, 1991, 32-35.

28. Melendres, J.L et al.; Int. J. Pharm. 2, 1993, 243-248.

29. William, A.C.; and Barry, B.W.; Adv Drug Deliv. 56, 2004, 603-618.

30. Hadgraft, J et al.; Int. J Pharma.141 (1-2), 1996, 17-25.

31. Phillips, C. A. ; and Michniak, B. B.; J. Pharm. Sci.84,1995,1427.

32. Park, E.S et al.; Drug Deve Ind Pharmacy.27, 2001, 975-980.

33. Jungbauer, F.H.W.; Coenraods, P.J.; and Kardaun, S.H.; Contact

Dermatitis. 45, 2001, 303- 304.

34. Wong, O et al.; Int. J. Pharm. 52, 1989, 191–192.

35. Aungst BJ, Rogers NJ, Shefter E. Enhancement of naloxone

penetration through human skin in vitro using fatty acid, fatty

alcohol, surfactant, sulfoxide and amide. Int J Pharm 1986; 33: 244-

247.

36. Aungst BJ. Structure effect studies of fatty acid isomers as skin

penetration enhancers and skin irritant. Pharm Res 1989; 6:244-247.

37. Shin, S.C.; and Lee, H.J.; Int J Pharm. 54, 2002, 201-206.

38. Funke, A.P et al.; Pharm Res 2002; 19: 661-668.

39. Goodman, M.; and Barry, B.W.; Int J Pharm. 57, 1989, 29-40.

40. Chien, Y.W et al.; Pharm. Rcs.5, 1998, 103- 106.

41. Friend, D et al.; J. Control. Release. 7, 1998, 243-250.

42. Sugibayashi, K. et al.; J. Pharm. Sci.81, 1992, 58-65.


43. Mollgaard, B.; and Hoclgaard, A.; Acta Pharm. Suet. 20, 1983, 433-

442.

44. Zatz, J.L.; and Lee, B.; Skin penetration enhancement by surfactants,

4th edition, Marcel Dekker, 1997, 546-68.

45. Scheuplein, R.J.; and Ross, L.; Journal of the Society of Cosmetic

Chemists. 21(13), 1970, 853-873.

46. William, A.C.; and Barry, B.W.; Int J Pharm.57, 1989,R7-R9

47. Cornwell, P.A.; and Barry, B.W.; J Pharm Pharmacol.46, 1994, 261-

269.

48. EL-Kuttan, A.F.; Asbill, C.S.; Michniak, B.B.; Int J Pharm.198, 2000,

179-189.

49. Vollmer, U et al.; Int. J.Pharm. 21(13), 1999, 51-58.

50. Rajadhyaksha, V.; and Pfister, W.R.; Drug Cosmet Ind. 1, 1996, 36-

47.

51. Goldberg, E. P. Eds. Targeted Drugs, 2nd Edition, Wiley, 1983, 312.

52. Gregoriadis, G.; Nature, 265, 1977, 407-419.

53. Helmstädter, A.; Pharmazie. 56, 2001, 583-587.

54. Subramony, J.A.; Sharma, A.; and Phipps,J.B.; Int. J. Pharm. 317 (1),

2006, 1-6.

55. Kanikkannan, N.; Biodrugs. 16, 2002, 339-347.

56. Mitragotri, S.; Blankschtein, D.; and Langer, R.; (1996) Pharm. Res.

13, 1996, 411-420.

57. Smith, N.B et al.; Ultrasound. Med. Biol. 29, 2003, 311-317.
58. Sintov, A et al.; J. Control. Rel. 89, 2003, 311-320.

59. Akomeah, F et al.; Eur. J. Pharm Sci. 21, 2004, 337-345.

60. Paranjape, M et al.; Sens Actuators A. 104, 2003, 195-204.

61. Gerstel, M.S.; and Place, V.A.; Drug delivery device, 1976, Patent

(Serial Number US 3,964, 482).

62. Allen, M.G et al.; Microneedle devices and methods of manufacture

and use thereof, 2002, Patent (Serial Number US 6, 334,856).

63. Godshall, N.; 1996, Patent (Serial Number WO 9637256).

64. Kamen, D.; 1998 Patent Serial Number WO 98/ 11937.

65. Jang, K.; 1998, Patent Serial Number US 5,843,114.

66. Longbridge, D.J et al.; Control. Rel Bioact. Mat. 25, 1998, 964.

67. Burkoth, T.L et al.; Crit. Rev Ther. Drug Carrier. Syst. 16, 1999, 331-

384.

68. Treffel, P et al.; Acta. Derm. Venereol (Stockh). 73, 1993, 200-202.

69. Cormier, M et al.; 2001, Patent (Serial Number WO 01/41864 A1).

70. Neukermans, A.P et al.; Device and method for enhancing

microprotrusion skin piercing. 2001, Patent (Serial Number WO

0141863).

71. Bangham, A. D.; Standish, M.; and Watkins, J.G.; J. Mol. Biol. 13,

1965, 238.

72. Wagner , A.; and Uhl, K.V.; J Drug Deliv. 201(1), 2011,1-9.

73. Doijad, R.C et al.; J Global Pharm Technol. 1(1), 2009, 94-100.

74. Bhatt,D.A.; and Pethe, A.M.; Int J Pharm Res Dev. 2 (7), 2010, 1-11.
75. Gangwar, S.; Singh, S.; and Garg, G.; J Pharm Res. 3(4), 2010, 688-

691.

76. Cevc, G.; and Blume, G.; Biochemicaet Biophysica Acta. 78 (1), 2001,

1-15.

77. Patel, J.L.; and Bharadia, P.D.; World J Pharm Res. 1(3), 2012, 456-

469.

78. Kumar, D et al.; Int Scholar Res Network Pharm. 6 (3), 2012, 1-14.

79. Namdeo, A.; and Jain, N.K.; Indian J Pharm Sci. 58(2), 1996, 41-46.

80. Arora, D et al.; Int J Recent Adv Pharm Res. 1, 2011, 45-51.

81. Chaudhari, M.J et al.; Int J Pharm Res Scholar. 1(2), 2012, 485-489.

82. Baillie, A.J et al.; J. Pharma Pharmacol. 37(12), 1985, 863-868.

83. Rogerson, A et al.; J. Pharma Pharmacol. 40(5), 1988, 337-342.

84. Chandra Prakash, K.S et al.; Int J Pharm. 61(6), 1990, R1-R3.

85. Raja Naresh, R.A et al.; Indian drugs. 30(6), 1993, 275-278.

86. Chandra Prakash, K.S et al.; Drug Dev Ind Pharm. 19(11), 1993,

1331-1342.

87. Malhotra, M.; and Jain, N.K.; Indian drugs. 31(3), 1994, 81-86.

88. Partha Sarathi, G.; Udupa, N.; and Pillai G.K.; J Drug Target; 2(2),

1994, 173-182.

89. Partha Sarathi, G.; Udupa, N.; and Pillai, G.K.; Indian J Pharm Sci.

56(3), 1994, 90-94.

90. Naresh, R.A.; and Udupa, N.; STP Pharm Sci. 6(1), 1996, 61-71.

91. Raja Naresh, R.A.; Udupa, N.; and Umadevi, P.; Indian J Pharm Sci.
58(6), 1996, 230-235.

92. Arul, B.; East Pharm. 41(11), 1998, 115-116.

93. Erdogan, S.; Ozer, A.Y.; and Hincal, A.A.; STP Pharm Sci. 8(2), 1998,

127-132.

94. Sheena, I.P et al.; Indian J Pharm Sci. 60(1), 1998, 45-48.

95. Namdeo, A et al.; Indian drugs. 36(6), 1999, 378-380.

96. Devi, S.G.; Venkatesh.; and Udupa, N.; Indian J Pharm Sci. 62(6),

2000, 479-481.

97. Ruckmani. K.; Jayakar. B.; and Ghoshal, S.K.; Drug Dev Ind Pharm26

(2), 2000, 217-222.

98. Fang, J.Y et al.; Int J Pharm. 219(5), 2001, 61-72.

99. Ravi Chandran, V et al.; East Pharm. 44(2), 2001, 113-15.

100.Ruckmani,K.; and Ghoshal, S. K.; STP Pharm Sci. 11(4), 2001, 301-

303.

101.Alia Sagar Shashiwala.; and Ambikananadan Mishra.; J Pharm

Pharmaceut Sci. 5(3), 2002, 220-225.

102.Balasubramaniam,A.; Kumar, V.A.; and Pillai, K.J.; Drug Dev Ind

Pharm. 28(102), 2002, 1181-1193.

103.Dattu Shivani et al.; Indian drugs. 39(3), 2002, 163-165.

104.Joseph, V.M.; Balamurugan, M.; and Ezhilmuthu, P.R.; Indian drugs.

40(7), 2002, 385-389.

105.Manconi, M et al.; Int J Pharm. 234(3), 2002, 237-248.

106.Roux, E et al.; Int J Pharm. 242(1-2), 2002, 257-36.


107.Satturwar, P.M et al.; Indian J Pharm Sci. 64(2), 2002, 155-158.

108.Varshosaz, J.; Pardakhty, A.; and Hajhashemi, V.; Drug deliv. 10(4),

2003, 251-262.

109.Aggarwal, D.; Garg, A.; and Kaur, I.P.; J. Pharma Pharmacol. 56(12),

2004, 1509-1517.

110.Dufes, C et al.; Pharm Res. 21(1), 2004, 101-107.

111.Mullaicharam, A.R.; and Murthy, R.S. STP Pharm Sci. 14(2), 2004, 99-

104.

112.Aggarwal, D.; and Kaur, I.P.; Int J Pharm. 290(1-2), 2005, 155-159.

113.Alsara, I.A et al.; Sci Pharm. 73(3), 2005, 81-93.

114.Guinedi, A.S et al.; Int J Pharm. 306(1-2), 2005, 71-82.

115.Huang, Y.Z et al.; Pharmazie. 60(6), 2005, 473-474.

116.Jain, S.; and Vyas, S.P.; J. Pharma Pharmacol. 57(9), 2005, 1177-

1184.

117.Nasseri. B.; Int J Pharm. 300(1-2), 2005, 95-101.

118.Ning, M.Y et al.; Drug Dev Ind Pharm. 331(4-5), 2005, 75-383.

119.Jain, C.P.; Vyas, S.P.; and Dixit, V.K.; Indian J Pharm Sci. 68(5),

2006, 575-578.

120.Manconi.M et al.; Int J Pharm. 311(1-2), 2006, 11-19.

121.Manivannan Rangasamy et al.; Journal of Pharmacy Research. 1(2),

2008, 163-168.

122.Naresh Ahuja et al.; Rasayan J. Chem. 1(3), 2008, 561-563.

123.Sankar, V et al.;RJPBCS. 2 (3), 2009, 223 – 230.


124. Gupta Naveen et al.; IJPLS, 1(7), 2010, 413-418.

125. Srinivas, S et al.; Digest Journal of Nanomaterials and Biostructures.

5 (1), 2010, 249 – 254

126. Kandasamy Ruckmani.; and Veintramuthu Sankar.; AAPS

PharmSciTech. 11(3), 2010, 1119–1127.

127. Pavala Rani, N.; Suriyaprakash, T.N.K.; and Senthamarai, R.;

International Journal of Pharma and Bio Sciences. 1 (4), 2010, 379-

387.

128. Srinivas, S et al.; Digest Journal of Nanomaterials and Biostructures.

5 (1), 2010, 249 – 254.

129. Akhilesh, D.; Anoop, V. N.; and Rao. B.P.; International Journal of

Research in Pharmaceutical and Biomedical Sciences. 2 (4), 2011,

1582-1589.

130. Chawda Himmat Singh.; Jain C.P.; and Bairwa Narendra Kumar.;

Pharmacophore. 2 (3), 2011, 168 -185.

131. Mohamed Firthouse, P.U et al.; Int.J. PharmTech Res. 3(2), 2011,

1019-1022.

132. Abraham lingan,M et al.; Sci. Revs. Chem. Commun.1 (1), 2011, 7-

17.

133. Vyas Jigar et al.; IJPBCS, 3 (3), 2011, 1714-1718.

134. Sathyavathi, V et al.; International journal of life sciences & pharma

research. 2 (1), 2012, L82- L95.

135. Pardakhty, A et al. J Microencapsul. 29(3), 2012, 219-24.


136. Disha Mehtani.; and Rajesh Kumar Maheshwari.; Lambert

Publications. 4 (4), 2012, 12-19.

137. Rajesh, Z.; Mujoriya.; and Ramesh Babu Bodla.; Advances in Life

Science and Technology. 3 (2), 2012, 1 – 13.

138. Guiling, Li et al.; European Journal of Pharmaceutics and

Biopharmaceutics 82, 2012, 49–57

139. Manish K.Chourasia.; LifengKang.; and SuiYungChan.; Results in

Pharma Sciences. 1, 2011, 60–67.

140. Limsuwan, T.; and Amnuaikit, T.; Procedia Chemistry. 4, 2012, 328 –

335.

141. Godin, B.; and Touitou, E.; Journal of Controlled Release. 94,

2004, 365– 379.

142. Vaibhav Dubey.; Dinesh Mishra.; and Jain, N.K.; European Journal of

Pharmaceutics and Biopharmaceutics. 67, 2007, 398–405

143. Vaibhav Dubey et al.; Journal of Controlled Release. 123, 2007, 148–

154.

144. Maria Manconi et al.; European Journal of Pharmaceutics and

Biopharmaceutics. 78, 2011, 27–35.

145. Vijayakumar M.R et al.; Int J Pharm Pharm Sci, 2(4), 2010, 82-86.

146. Ashoniya Sheer.; and Meenakshi Chauhan.; Journal of

pharmaceutics and cosmetology. 1(3), 2011, 1-14.

147. Xingyan, Liu et al.; International Journal of Nanomedicine. 6 2011,

241–247.
148. Elisabetta Esposito.; and Enea Menegatti, J. Cosmet. Sci. 55,

2004, 253-264.

149. Verma P.; IJPSR. 2(9), 2011, 2472-2474.

150. Jadupati Malakar et al.; Saudi Pharmaceutical Journal. 20, 2012,

355–363.

151. Donatella Paolino et al.; Journal of Controlled Release. 162, 2012,

143–151.

152. Gregor Cevc.; Andreas Schatzlein.; and Holger Richardsen.;

Biochimica et Biophysica Acta. 156(4), 2002, 21– 30.

153. Eun kyung Oh et al.; European Journal of Pharmaceutical Sciences.

44, 2011, 149–157.

154. Jorge Montanari et al.; Journal of Controlled Release. 147, 2010,

368–376.

155. Geusens, B et al.; Journal of Controlled Release. 133, 2009, 214–220.

156. Gregor Cevc.; and Gabriele Blume.; Biochimica.Biophysica Acta.

1514, 2001, 191-205.

157. Ebtessam A. Essa et al.; Journal of Controlled Release. 92, 2003,

163–172.

158. Gregor Cevc.; and Gabriele Blume.; Biochimica et Biophysica Acta

1614, 2003, 156– 164.

159. Loan Honeywell-Nguyen, P et al.; Journal of Controlled Release. 90,

2003, 243–255
160. Gregor Cevc, et al.; Biochimica et Biophysica Acta. 1256, 1998, 201–

215.

161. Ramesh R. Boinpally et al.; European Journal of Pharmaceutics

and Biopharmaceutics. 56, 2003, 389–392.

162. Yuka Hiruta et al.; Journal of Controlled Release. 113, 2006, 146–

154.

163. Loan Honeywell-Nguyen, P.; and Joke A. Bouwstra.; Journal of

Controlled Release. 86, 2003, 145–156.

164. Ajay Kumar.; Kamla Pathak.; and Vikas Bali.; Drug Discovery Today.

17, 2012, 123-127.

165. Tomonobu Uchino et al.; European Journal of Pharmaceutics and

Biopharmaceutics 2014 (In Press)

166. Sureewan Duangjit et al.; Journal of Drug Delivery, 2011, Article ID

418316, doi : 10.1155 / 2011 / 418316.

167. Mohammed Irfan.; Sushma Verma.; and Alpana Ram.; Asian J Pharm

Clin Res. 5 (3), 2012, 162-165.

168. http://en.wikipedia.org/wiki/Nystatin

169. Hardevinder palsingh.; Ashok kumar Tiwari.; and Subeet Jain. The

pharmaceutical society of Japan. 130 (3), 2010, 397-407.

170. Groll, A.Het al.;  J Chromatogr B Biomed Sci Appl. 735(1), 1999,51-

62
INDEX
INDEX

A Epidermis

Application of Pressure Ethosomes

Aquasomes Eutectic systems

B Ex vivo drug release

Bilosomes F

Bingam bodies First order

C H

Chemical penetration enhancers Higuchi

Chemical potential adjustment Hixon-crowel

Cholesterol HPLC

D Hydration

Dermis I

Dialysis membrane In vitro drug release

Dialysis method In vivo study

Dialysis method Intercellular route

E Ion pairs and complex coacervates

Elasticity of the vesicles Iontophoresis

Electrophoresis K

Enhancement ratio Keratinocytes

Entrapment efficiency Korsmeyer-Peppas

Enzymosomes L
Langerhans cells

Laser radiation and photochemical S

waves Skin

Liposomes Skin abrasion

M Skin puncture and perforation

Magnetophoresis Skin stretching

Melanocytes Span-60

Merkel cells Span-80

Microneedle based devices Sphingosomes

Morphology Stability studies

N Stratum basale

Needleless injection Stratum corneum

Niosomes Stratum granulosum

Nystatin Stratum lucidem

P Stratum spinosum

Permeability co-efficient. Suction ablation

Pharmaceutical carrier systems T

Pharmacosomes Temperature (“thermophoresis”)

Phosphate buffer saline pH 7.4 Thin film hydration technique

Phospolipon-90H Topical drug delivery systems

R Transcellular route

Radio frequency Transdermal drug delivery systems

Routes of administration Transdermal flux.


Transferosomes V

Transfollicular route Vesicular Carrier Systems

U Virosomes

Ultra centrifugation method Z

Ultrasound ( Phonophoresis, Zero order

Sonophoresis) Zeta potential

UV-Visible spectrophotometer
LIST OF PUBLICATIONS &
CONFERENCES

LIST OF PUBLICATIONS & CONFERENCES


1. K. Srikanth, V. Rama Mohan Gupta, N. Devanna. Formulation and Evaluation

of Nystatin loaded Niosomes. International Journal of Pharmaceutical Sciences

and Research, 4(5), 2013, 2015 – 2020. (ISSN No: 0975 – 8232).

2. K. Srikanth, V. Rama Mohan Gupta, N. Devanna. Ethanolic Vesicles of

Nystatin for enhanced Transdermal Drug Delivery: Invitro & Exvivo Evaluation.
Indo-American Journal of Pharmaceutical Research, 3(9), 2013, 7316 – 7324.

(ISSN No: 2231 - 6876).

3. K. Srikanth, V. Rama Mohan Gupta, Sundaraj Manvi, N. Devanna. Particulate

Carrier Systems: A Review. International Research Journal of Pharmacy. 3(11),

2012, 22 -26. (ISSN No: 2230 - 8407).

4. Presented a Poster entitled “Formulation and Evaluation of Nystatin loaded

Niosomes” in an International Conference ‘Indo-American Pharmaceutical

Regulatory Symposium’ held at JNTUH, Hyderabad in 2012.

5. Gave an Oral Presentation entitled “Formulation and Evaluation of Nystatin

loaded Transferosomes” in AICTE sponsored a two days National Level

Conference “INNOVATE – 13” at Pulla Reddy Institute of Pharmacy, Medak in

2013.

You might also like