29.04.2013 Views

FORMULATION AND EVALUATION OF CERTAIN TOPICALLY ...

FORMULATION AND EVALUATION OF CERTAIN TOPICALLY ...

FORMULATION AND EVALUATION OF CERTAIN TOPICALLY ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

Turn your PDF publications into a flip-book with our unique Google optimized e-Paper software.

<strong>FORMULATION</strong> <strong>AND</strong> <strong>EVALUATION</strong> <strong>OF</strong><br />

<strong>CERTAIN</strong> <strong>TOPICALLY</strong> APPLIED DRUGS<br />

A Thesis Submitted for the<br />

Degree of Master<br />

In<br />

Pharmaceutical Sciences (Pharmaceutics)<br />

By<br />

Rasha Ali AL-Hussiny<br />

Under the Supervision of<br />

Prof. Dr. Fakhr El-Din S. Ghazy<br />

Professor of Pharmaceutics<br />

Faulty of Pharmacy<br />

Zagazig University<br />

Dr. Mohamed A. Hammad Dr. Nagia A. El-Megrab<br />

Assistant Professor of Assistant Professor of<br />

Pharmaceutics Pharmaceutics<br />

Faulty of Pharmacy Faulty of Pharmacy<br />

Zagazig University Zagazig University<br />

Department of Pharmaceutics<br />

And Industrial Pharmacy<br />

Faculty of Pharmacy<br />

Zagazig University<br />

2010<br />

- 1 -


- 2 -


- 3 -


ACKNOWLEDGEMENTS<br />

I am deeply thankful to GOD, by the grace of whom the progress and<br />

success of this work was possible.<br />

I would like to express my heartfelt gratitude and profound indebtedness<br />

to my guide Prof. Dr. F. S. Ghazy, Professor of Pharmaceutices, Faculty<br />

of Pharmacy, Zagazig University; the greatest supporting person for this<br />

work. Under his guidance I have worked. His constant enlightening<br />

support, timely advice all throughout my work and encouragement have<br />

been instrumental in the completion of this study.<br />

Also, I have to thank Dr. M.A. Hammad, Assistant Professor of<br />

Pharmaceutices, Faculty of Pharmacy, Zagazig University; for<br />

supervising the work, for his encouragement and for his great efforts to<br />

make this work possible.<br />

Also, I thank Dr. N. A. EL-Megrab, Assistant Professor of<br />

Pharmaceutices, Faculty of Pharmacy, Zagazig University; appreciating<br />

her continous encouragement and help supporting me with much scientific<br />

materials and with valuable instructions.<br />

I also extend my sincere thanks to all my colleagues and members of the<br />

department of Pharmaceutics, Faculty of Pharmacy, Zagazig University<br />

for their help.<br />

And finally I would like to thank my family, for their support during this<br />

study …Thank You.<br />

Rasha<br />

2010<br />

- 4 -


ABBREVIATIONS<br />

ABBREVIATION THE WORD<br />

Glz Gliclazide<br />

Glib Glibenclamide<br />

PEG Polyethylene glycol<br />

UR Urea<br />

glu Glucose<br />

O/W Oil in water<br />

W/O Water in oil<br />

HPMC Hydroxypropylmethyl cellulose<br />

WSB Water soluble base<br />

IPP Isopropyl palmitate<br />

IPM Isopropyl myristate<br />

OA Oleic acid<br />

LOA Linoleic acid<br />

Lab Labrafil<br />

Tc Transcutol<br />

SLS Sodium lauryl sulphate<br />

Tw 80 Tween 80 ( Polyoxyethylene Sorbitan<br />

Monooleate)<br />

PG Propylene glycol<br />

Span 80 Sorbitan mono-oleate<br />

i.p intraperitoneal<br />

NIDDM Non insulin dependant diabetes mellitus<br />

- 5 -


List of Tables<br />

List of Figures<br />

Contents<br />

Abstract ………………………………………………………………. i<br />

General Introduction …………………………………………………. 1<br />

Scope of work ………………………………………………………... 35<br />

Part One<br />

Formulation and Evaluation of Topically Applied Gliclazide<br />

- Introduction ………………………………………………………… 37<br />

Chapter (I)<br />

Formulation and Characterization of Gliclazide Solid Dispersions<br />

-Introduction …………………………………………………………. 40<br />

-Experimental and methodology …………………………………….. 67<br />

-Results and discussion ……………………………………………… 74<br />

-Conclusion ………………………………………………………….. 117<br />

Chapter (II)<br />

In Vitro and In Vivo Studies on Topical Applications of Gliclazide<br />

Solid Dispersions<br />

-Introduction ………………………………………………………. 118<br />

-Experimental and methodology ………………………………….. 119<br />

-Results and discussion …………………………………………… 134<br />

-Conclusion ……………………………………………………….. 157<br />

- 6 -


Part Two<br />

Formulation and Evaluation of Topically Applied Glibenclamide<br />

-Introduction …………………………………………………… 158<br />

-Experimental and methodology ………………………………… 176<br />

-Results and discussion …………………………………………… 186<br />

-Conclusion ……………………………………………………….. 235<br />

General Conclusion …………………………………………………. 237<br />

References …………………………………………………………… 238<br />

Arabic Summary …………………………………………………….. <br />

- 7 -


Figure<br />

List of Figures<br />

Number Description<br />

- 8 -<br />

Page<br />

Number<br />

1 Diagrammatic representation of the skin structure. 3<br />

2 Diagrammatic representation of the stratum<br />

corneum and the intercellular and transcellular<br />

routes of penetration<br />

3 Schematic representation of types of external<br />

medicines.<br />

4 Structure of gliclazide<br />

5 Diagrammatic representation of process of<br />

solubilization<br />

6 Phase diagram for eutectic system 55<br />

7 Phase diagram for Discontinuous solid solutions 56<br />

8 Substitutional crystalline solid solutions<br />

9 Interstitial crystalline solid solutions.<br />

10 Amorphous crystalline solid solution 58<br />

11 UV spectra of gliclazide in methanol. 74<br />

12 Calibration curve of gliclazide in methanol at max<br />

227 nm.<br />

13 Calibration curve of gliclazide in phosphate buffer<br />

(7.4)at max 227 nm .<br />

14 Phase solubility diagram of gliclazide in water at<br />

25°C in presence of PEG 4000 and PEG 6000.<br />

15 Phase solubility diagram of gliclazide in water<br />

at 25°C in presence of glucose and urea.<br />

16 Dissolution profile of gliclazide-PEG 6000 systems. 82<br />

17 Dissolution profile of gliclazide-PEG 4000 systems. 84<br />

10<br />

20<br />

37<br />

41<br />

57<br />

58<br />

75<br />

75<br />

78<br />

78


18 Dissolution profile of gliclazide-glucose systems. 87<br />

19 Dissolution profile of gliclazide-urea systems 89<br />

20 Ratio between % of gliclazide dissolved from (A)<br />

drug in different solid dispersions and (B) drug<br />

alone at t = 60 min.<br />

21 FTIR spectra of gliclazide –PEG 6000 systems.<br />

22 FTIR spectra of gliclazide –PEG 4000 systems. 100<br />

23 FTIR spectra of gliclazide –glucose systems.<br />

- 9 -<br />

92<br />

99<br />

101<br />

24 FTIR spectra of gliclazide –urea systems. 102<br />

25 DSC spectra of gliclazide –PEG 6000 systems. 106<br />

26 DSC spectra of gliclazide –PEG 4000 systems. 107<br />

27 DSC spectra of gliclazide –glucose systems.<br />

108<br />

28 DSC spectra of gliclazide –urea systems. 109<br />

29 X-ray spectra of gliclazide –PEG 6000 systems. 113<br />

30 X-ray spectra of gliclazide –PEG 4000 systems. 114<br />

31 X-ray spectra of gliclazide –glucose systems. 115<br />

32 X-ray spectra of gliclazide –urea systems. 116<br />

33 Diagrammatic representation of the drug diffusion<br />

apparatus.<br />

34 In vitro release profile of gliclazide from different<br />

topical preparations.<br />

35 In vitro release profile of gliclazide and (8:92)<br />

gliclazide –PEG 6000 solid dispersion from<br />

different topical bases.<br />

36 In vitro release profile of gliclazide and (1:10)<br />

gliclazide –glucose solid dispersion from different<br />

topical bases.<br />

37 In vitro release profile of gliclazide and (8:92)<br />

gliclazide –PEG 4000 solid dispersion from<br />

different topical bases.<br />

125<br />

136<br />

141<br />

143<br />

145


38 In vitro release profile of gliclazide and (1:10)<br />

gliclazide –urea solid dispersion from different<br />

topical bases.<br />

39 Release of gliclazide from different bases with<br />

different solid dispersions.<br />

40 Percent reduction in blood glucose levels after oral<br />

and topical administration of gliclazide in normal<br />

rats.<br />

41 Percent reduction in blood glucose levels after oral<br />

and topical administration of gliclazide in diabetic<br />

rats.<br />

- 10 -<br />

147<br />

148<br />

153<br />

156<br />

42 Glibenclamide structure. 158<br />

43 Techniques to optimize drug permeation across the<br />

skin.<br />

44 UV absorption spectra for glibenclamide in<br />

methanol.<br />

45 Calibration curve of glibenclamide in phosphate<br />

buffer (7.4) at max 227 nm.<br />

46 Release profile of glibenclamide from different<br />

topical bases.<br />

47 Percentage drug released from different topical<br />

bases.<br />

48 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of<br />

cetrimide.<br />

49 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of SLS.<br />

163<br />

186<br />

188<br />

192<br />

194<br />

199<br />

201


50 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of Tween<br />

80.<br />

51 Release profile of glibenclamide from water<br />

soluble base containing different concentrations of<br />

labrafil.<br />

52 Percentage drug released from water soluble base<br />

containing different concentrations of different<br />

surfactants<br />

53 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of oleic<br />

acid.<br />

54 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of linoleic<br />

acid.<br />

55 Percentage drug released from water soluble base<br />

containing different concentrations of fatty acids.<br />

56 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of<br />

isopropyl myristate.<br />

57 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of<br />

isopropyl palmitate .<br />

58 Release profile of glibenclamide from water soluble<br />

base containing different concentrations of<br />

Transcutol.<br />

- 11 -<br />

203<br />

205<br />

206<br />

209<br />

211<br />

212<br />

215<br />

217<br />

220


59 . Percentage drug released from water soluble base<br />

containing different concentrations of fatty acid<br />

esters and Transcutol.<br />

60 Percentage drug released from water soluble base<br />

containing the best concentrations of different<br />

penetration enhancers used.<br />

61 Percent reduction in blood glucose levels after oral<br />

and topical administration of glibenclamide in<br />

normal rats.<br />

62 Percent reduction in blood glucose levels after oral<br />

and topical administration of glibenclamide in<br />

diabetic rats.<br />

- 12 -<br />

221<br />

222<br />

231<br />

234


Table<br />

Number<br />

List of Tables<br />

Description Page<br />

1 Methods for the characterization of solid<br />

dispersion.<br />

2 Types of carriers and their ratios in gliclazide solid<br />

dispersions and physical mixtures.<br />

3 Solubility enhancement data of gliclazide in various<br />

carrier solutions at 25°C.<br />

4 Effect of change in pH on the solubility of<br />

gliclazide.<br />

5 Dissolution parameters (±SD) of gliclazide in<br />

distilled water from different gliclazide - PEG 6000<br />

systems.<br />

6 Dissolution parameters (±SD) of gliclazide in<br />

distilled water from different gliclazide - PEG 4000<br />

systems.<br />

7 Dissolution parameters (±SD) of gliclazide in<br />

distilled water from different gliclazide – glucose<br />

systems.<br />

8 Dissolution parameters (±SD) of gliclazide in<br />

distilled water from different gliclazide –urea<br />

systems.<br />

9 Collective data for dissolution of gliclazide<br />

obtained from different carriers used.<br />

10 FTIR spectra of gliclazide solid dispersions and<br />

physical mixtures compared with individual<br />

components.<br />

- 13 -<br />

Number<br />

64<br />

69<br />

77<br />

79<br />

81<br />

83<br />

86<br />

88<br />

91<br />

95


11 Fusion temperatures (Tc) and heat of fusion (<br />

of gliclazide solid dispersions and physical mixtures<br />

compared with individual components.<br />

12 º)<br />

for some gliclazide solid dispersions and physical<br />

mixtures compared with individual components.<br />

- 14 -<br />

105<br />

111<br />

13 Composition of different topical bases 124<br />

14 Amounts of sample and standard used 131<br />

15 In vitro release data of gliclazide from<br />

different topical bases<br />

135<br />

16 Viscosity of different topical bases. 138<br />

17 In vitro release of gliclazide and (8:92) gliclazide-<br />

PEG 6000 solid dispersion from different topical<br />

bases<br />

18 In vitro release of gliclazide and (1:10) gliclazide-<br />

glucose solid dispersion from different topical<br />

bases.<br />

19 In vitro release of gliclazide and (8:92) gliclazide-<br />

PEG 4000 solid dispersion from different topical<br />

bases.<br />

20 In vitro release of gliclazide and (1:10) gliclazide-<br />

urea solid dispersion from different topical bases.<br />

21 Kinetic data of the release of gliclazide and its solid<br />

dispersions from different topical bases.<br />

22 Reduction in blood glucose level after oral and<br />

topical application of gliclazide and 10:90<br />

gliclazide- PEG 6000 solid dispersion in normal<br />

rats. All values are expressed as mean ± sd.<br />

140<br />

142<br />

144<br />

146<br />

149<br />

152


23 Reduction in blood glucose level after oral and<br />

topical application of gliclazide and 10:90<br />

gliclazide- PEG 6000 solid dispersion in diabetic<br />

rats. All values are expressed as mean ± sd.<br />

- 15 -<br />

155<br />

24 Composition of different topical formulations. 180<br />

25 Types of penetration enhancers and percentages<br />

used.<br />

26 In vitro release of glibenclamide from different<br />

topical bases.<br />

27 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of<br />

cetrimide<br />

28 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of Sodium<br />

lauryl sulphate (SLS).<br />

29 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of Tween<br />

80.<br />

30 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of labrafil.<br />

31 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of oleic<br />

acid.<br />

32 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of linoleic<br />

acid.<br />

33 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of<br />

Isopropylmyristate (IPM).<br />

182<br />

198<br />

200<br />

202<br />

204<br />

208<br />

210<br />

214


34 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of<br />

Isopropylpalmitate (IPP).<br />

35 In vitro release of glibenclamide from water soluble<br />

base containing different concentrations of<br />

Transcutol.<br />

36 Kinetic data of the release of Glib from different<br />

topical bases<br />

37 Reduction in blood glucose level after oral and<br />

topical application of glibenclamide and<br />

glibenclamide with 1% oleic acid in normal rats.<br />

38 Reduction in blood glucose level after oral and<br />

topical application of glibenclamide and<br />

glibenclamide with 1% cetrimide in normal rats<br />

39 Reduction in blood glucose level after oral and<br />

topical application of glibenclamide and<br />

glibenclamide with 1% isopropyl myristate (IPM) in<br />

normal rats..<br />

40 Reduction in blood glucose level after oral and<br />

topical application of glibenclamide and<br />

glibenclamide with 5 % Labrafil in normal rats.<br />

41 Reduction in blood glucose level after oral and<br />

topical application of glibenclamide and<br />

glibenclamide with 1% cetrimide in diabetic rats.<br />

- 16 -<br />

216<br />

219<br />

224<br />

227<br />

228<br />

229<br />

230<br />

233


- 17 -


Abstract<br />

Part One<br />

Formulation and Evaluation of Topically Applied Gliclazide.<br />

Chapter One<br />

Formulation and Characterization of Gliclazide Solid Dispersions.<br />

The purpose of this study was to improve the dissolution of Gliclazide<br />

(Glz) for enhancing its bioavailability and therapeutic efficacy.<br />

Physical mixtures (PMs) and solid dispersions (SDs) of Glz with each of<br />

polyethylene glycol 4000 (PEG 4000) and polyethylene glycol 6000 (PEG<br />

6000) in ratios 10: 90, 8: 92, 5: 95 and 1: 99 (drug-to-carrier w/w) were<br />

prepared. Glucose (glu) and urea (UR) in ratios 1:1, 1:2, 1: 3, 1: 5 and 1: 10<br />

(drug-to-carrier w/w) were also prepared. All SDs were prepared by solvent<br />

evaporation method. The equilibrium solubility of Glz in presence of<br />

different concentrations of the above mentioned carriers was determined at<br />

25°C and the influence of different pH on the solubility of Glz was also<br />

examined. The dissolution of all prepared samples (PMs and SDs) was<br />

carried out in media of pure distilled water pH 6.5. All SDs and PMs as well<br />

as individual components were subjected to inspection by FTIR<br />

spectroscopy, DSC and X-ray powder diffraction.<br />

The results revealed that, the aqueous solubility of Glz was favoured<br />

by the presence of PEG 4000 and PEG 6000 while the aqueous solubility<br />

was slightly improved when glu or UR was used as a carrier. The solubility<br />

of Glz increased with increasing pH (higher in alkaline medium rather than<br />

acidic one). The type of carrier and drug to carrier ratio had great influence<br />

on the rate and extent of dissolution of Glz from its SDs. All the investigated<br />

carriers improved the dissolution rate of Glz. The highest rates were obtained<br />

from PEG 6000 followed by PEG 4000, glu and finally UR SDs at mixing<br />

- 18 -


atios of (1:99), (1:99), (1:10) and (1:10) respectively. Physical<br />

characterization of all systems prepared revealed structural changes in the<br />

prepared SDs from the plain drug, which may account for increased<br />

dissolution rates.<br />

It was concluded that SDs showed increased dissolution rate as compared<br />

to the pure drug.<br />

Chapter Two<br />

In Vitro and In Vivo Studies on Topical Application of<br />

Gliclazide Solid Dispersions<br />

The aim of this study to enhance the release of Glz from topical<br />

preparations by incorporating it in the form of solid dispersion with water<br />

soluble carriers. Another aim was to determine whether a Glz would be<br />

absorbed through the skin and consequently lower blood glucose levels.<br />

Glz was formulated in different topical formulations. For this<br />

purpose, a set of traditional formulations such as ointment bases, cream<br />

bases and gel bases were utilized. The traditional classes of ointment<br />

bases studied were water soluble base (WSB), emulsion bases and<br />

absorption base. The gel base studied was hydroxylpropyl<br />

methylcellulose gel (HPMC gel). The emulsion bases chosen were oil in<br />

water (O/W) and water in oil (W/O) emulsions. Investigation of the<br />

release studies from topical formulation bases were carried in vitro over a<br />

period of six hours at a thermostatically controlled water bath operating at<br />

37°C and 100 rpm using the rate limiting membrane technique , at<br />

concentration of 1 % w/w Glz for all topical preparations. The receptor<br />

media employed throughout this investigation was sörensen’phosphate<br />

buffer of pH 7.4. The release studies of drug from (8:92) PEG 6000,<br />

- 19 -


(8:92) PEG 4000, (1:10) glu and (1:10) UR w/w drug to carrier ratio SDs<br />

from WSB, HPMC gel and O/W emulsion were investigated. In vitro skin<br />

permeation of Glz and its SDs from different topical formulations was<br />

studied. The blood glucose reducing hypoglycemic activity of Glz<br />

systems was studied in both normal and diabetic rats.<br />

The results revealed that, the percentage amount of drug released<br />

from WSB, gel base are greater than that released from other bases. The<br />

rate of drug release can be arranged in the following descending order:<br />

WSB (64.15 %) > HPMC gel (43.38 %) > O/W emulsion base (8.43 %).<br />

There is no drug is released fromabsorption base and W/O emulsion<br />

base. The amount of drug released from topical bases incorporating SDs<br />

can be arranged in the following descending order: Topical preparations<br />

containing drug: PEG 6000 (8:92) SD > (1:10) drug: glu (1/10) SD ><br />

drug: PEG 4000 (8:92) SD > drug: UR (1:10) SD > pure drug.Isolated<br />

skin permeation studies indicated that, the amount of Glz permeated<br />

across hairless rabbit skin was too small to be measured<br />

spectrophotometrically. The present study showed that Glz was absorbed<br />

through the skin and lowered the blood glucose levels. Topical<br />

preparations of Glz or its SDs exhibited better control of blood glucose<br />

level than oral Glz administration in rats as topical route effectively<br />

maintained normoglycemic level in contrast to the oral group which<br />

produced remarkable hypoglycemia. The blood glucose reducing activity<br />

of ointment contained (10:90) Glz –PEG 6000 solid dispersions was<br />

significantly more when compared to ointment contained Glz alone.<br />

The results suggest the possibility of transdermal administration of<br />

Glz for the treatment of NIDDM.<br />

- 20 -


- 21 -


General introduction<br />

Skin anatomy and physiology<br />

Skin is the largest organ of the body and, in addition to its primary<br />

function as a barrier for protection of the internal biological milieu from<br />

the external environment, has a variety of roles in the maintenance of<br />

physiological homeostasis (Monteiro-Riviere, 2001a) .<br />

1. The main funcnion of the skin:<br />

There are many different structures within the skin. Together these<br />

structures impart many protective properties to the skin that help to avoid<br />

damage to the body from outside influences. In this way, the skin serves<br />

many purposes:<br />

Protects the body from water loss and from injury due to bumps,<br />

chemicals, sunlight or microorganisms, and some glands (sebaceous)<br />

may have weak anti-infective properties.<br />

Helps to control body temperature through sweat glands.<br />

Is the sensor to inform the brain of changes in immediate environment.<br />

Produces vitamin D in the epidermal layer, when it is exposed to the<br />

sun's rays.<br />

Uses specialized pigment cells to protect us from penetration of<br />

ultraviolet rays of the sun.<br />

Act as channel for communication to the outside world.<br />

Plays an important role in regulation of body blood pressure (Chine,<br />

1982).<br />

- 22 -


2. Skin anatomy:<br />

As shown in (Figure 1), anatomically, skin is comprised of two principal<br />

components: a stratified, a vascular epidermis and the underlying dermis.<br />

The epidermis is further classified into layers called the stratum corneum,<br />

stratum lucidum, stratum granulosum, stratum spinosum, and the stratum<br />

basale. Together, these cell layers function to anchor the epidermis to the<br />

underlying dermis, to replenish cells that are naturally sloughed off from<br />

the surface epidermis, and to form a permeability barrier that protects the<br />

internal biological environment from the external milieu. The dermis<br />

consists of a dense irregular network of collagen, elastic, and reticular<br />

fibers that provides mechanical support for the tissue. An extensive<br />

network of capillaries, nerves, and lymphatics also located in the dermis<br />

facilitate the exchange of metabolites between blood and tissues, fat<br />

storage, protection against infections, and tissue repair. Below the dermis<br />

is the hypodermis, which anchors skin to underlying muscle or bone by<br />

loose connective tissue of collagen and elastic fibers (Monteiro-Riviere,<br />

2001a, 2004, 2006; Taylor et al., 2006).<br />

2.1. The epidermis:<br />

The epidermis is derived from ectoderm and consists of stratified<br />

squamous keratinized epithelium. The thickness and number of stratified<br />

layers varies among mammalian species and anatomical location. In<br />

general, porcine skin in the thoracolumbar area is an acceptable model for<br />

percutaneous absorption studies and has an epidermal thickness of about<br />

52 (Monteiro-<br />

Riviere, 2004). The vascular epidermis continuously undergoes an<br />

orderly process of proliferation, differentiation, and keratinization to<br />

- 23 -


eplenish the epidermis as stratum corneum cells are naturally sloughed<br />

from the skin’s surface (Monteiro-Riviere, 2006).<br />

Figure 1: Diagrammatic representation of the skin structure.<br />

- 24 -


Keratinocytes are the predominate cell type of the epidermis, accounting<br />

for approximately 80% of the cell population (Monteiro-Riviere, 2004).<br />

These cells originate in the stratum basale and, upon mitosis, undergo a<br />

continual differentiation process, known as keratinization. During this<br />

process, the epidermal cells migrate upward, increase in size, and produce<br />

differentiation products such as tonofilaments, keratohyalin granules, and<br />

lamellated bodies. Epidermal layers are easily identified by distinct<br />

differences in cell morphology and differentiation products that result due<br />

to keratinization. The remaining group of epidermal cells, known as<br />

nonkeratinocytes, consists of melanocytes, Langerhans cells, and Merkel<br />

cells and do not participate in the process of keratinization (Smack, et al.,<br />

1994).<br />

Stratum basale:<br />

The stratum basale is the layer of skin located closest to the dermis<br />

and is comprised of a single layer of columnar or cuboidal cells that are<br />

attached to the overlying stratum spinosum cells and to adjacent basale<br />

cells by desmosomes and to the underlying basement membrane by<br />

hemidesmosomes. Desmosomes are small, localized adhesion sites that<br />

mediate direct cell-to-cell contact by providing anchoring sites for<br />

intermediate filaments of the cellular cytoskeletons. Hemidesmosomes,<br />

on the other hand, function to provide strong attachment sites between the<br />

intermediate filaments of cells and the extracellular matrix of the<br />

underlying basal lamina (Taylor et al., 2006). In addition to their role in<br />

synthesizing the basement membrane, basale cells also function as stem<br />

cells to continuously produce keratinocytes that subsequently undergo<br />

keratinization. Immature keratinocytes of the stratum basale are capable<br />

of engaging in the synthesis of keratin, which are later assembled into<br />

keratin filaments called tonofilaments. Other nonkeratinocytes cells are<br />

also present in the stratum basale. Merkel cells are closely associated with<br />

- 25 -


nerve fibers and function as mechanoreceptors capable of relaying<br />

sensory information to the brain. Additionally, melanocytes, which<br />

produce and secrete melanin and provide protection from ultraviolet<br />

irradiation, reside near the basement membrane and are responsible for<br />

transferring melanin to surrounding keratinocytes.<br />

Stratum spinosum:<br />

The stratum spinosum or “prickle cell layer” is located above the<br />

stratum basale and consists of several layers of irregularly shaped<br />

polyhedral cells. Tight junctions and desmosomes connect adjacent cells<br />

and the underlying stratum basale. Additionally, Langerhans cells,<br />

important for the skin’s immune response, are found in this epidermal<br />

layer. This layer is morphologically distinguished from other epidermal<br />

layers by the presence of tonofilaments. As keratinocytes mature and<br />

move upward through this layer, the cells increase in size and become<br />

flattened in a plane parallel to the surface of the skin. Keratinocytes<br />

within the upper part of the stratum spinosum begin to produce<br />

keratohyalin granules and lamellar bodies, which are distinctive features<br />

of the cells in the stratum granulosum.<br />

Stratum granulosum:<br />

The next epidermal layer, the stratum granulosum, contains several<br />

layers of flattened cells positioned parallel to skin’s surface. The<br />

numerous granules those are present in the cells of this layer contain<br />

precursors for the protein filaggrin, which is responsible for the<br />

aggregation of keratin filaments present within the cornified cells of the<br />

stratum corneum. These granules fuse with the cell membrane and secrete<br />

their contents via exocytosis into the intercellular spaces between the<br />

stratum granulosum and stratum corneum layers. The lipid contents of the<br />

- 26 -


granules then form the intercellular lipid component of the stratum<br />

corneum barrier.<br />

Stratum lucidum:<br />

Present only in areas of thick skin, such as the palms of the hands<br />

and soles of the feet, is a subdivision of the stratum corneum called the<br />

stratum lucidum. This epidermal layer is a thin, translucent layer of cells<br />

devoid of nuclei and cytoplasmic organelles. These cells are keratinized<br />

and contain a viscous fluid, eleidin, which is analogous to keratin.<br />

tratum corneum:<br />

The stratum corneum is the outermost layer of the epidermis and its<br />

composition and organization significantly contribute to the skin’s<br />

permeability barrier. The stratum corneum consists of terminally<br />

differentiated cells arranged in multicellular stacks perpendicular to the<br />

surface of the skin. The cells are devoid of nuclei and cytoplasmic<br />

organelles and are almost completely filled with keratin filaments. The<br />

interlocking columns of cells are embedded in a structured lamellar<br />

matrix that consists of specialized lipids secreted from the granules of the<br />

stratum granulosum cells. This barrier functions to restrict the penetration<br />

of hydrophilic substances and large entities through the skin and to<br />

prevent excess loss of body fluids (Mackenzie, 1975; Menton, 1976;<br />

Monteiro-Riviere, 1991, 2001a, 2001b, 2006; Smack et al., 1994;<br />

Taylor et al., 2006)<br />

2.2. The dermis<br />

Collagen, elastic, and reticular fibers embedded in an amorphous<br />

ground substance of proteoglycans create a network of dense connective<br />

tissue that makes up the dermis. Fibroblasts, mast cells, and macrophages<br />

are the predominate cell types found in the dermis; however, plasma cells,<br />

- 27 -


fat cells, chromatophores, and extravasated leukocytes are often also<br />

present. The more superficial layer of the dermis, the papillary layer, lies<br />

immediately beneath the basement membrane and contains a less dense,<br />

irregular framework of type I and type III collagen molecules and elastic<br />

fibers. This region also contains blood and lymphatic vessels that serve<br />

but do not enter the epidermis and nerve processes that either terminate in<br />

the dermis or penetrate into the epidermis. Fingerlike protrusions of the<br />

dermal connective tissue into the underside of the epidermis are called<br />

dermal papillae. Likewise, epidermal ridges are similar protrusions of the<br />

epidermis into the dermis. Increased mechanical stress on the skin<br />

increases the depth of the epidermal ridges and length of the dermal<br />

papillae, thus, creating a more extensive interface between the dermis and<br />

epidermis. The reticular layer of the dermis lies beneath the papillary<br />

layer. This layer is substantially thicker than its superficial layer and is<br />

characterized by thick bundles of mostly type I collagen, coarser elastic<br />

fibers and fewer cells (Monteiro-Riviere, 1991, 2001a, 2001b, 2006).<br />

2.3. The hypodermis:<br />

The hypodermis is superficial fascia that lies below the skin and helps to<br />

anchor the dermis to underlying muscle and bone. It is comprised of<br />

connective tissue containing a loose arrangement of collagen and elastic<br />

fibers that allows for flexibility and free movement of the skin over the<br />

underlying structures (Monteiro-Riviere, 2006).<br />

2.4. Skin appendages:<br />

Hair follicles, associated sebaceous glands, arrector pili muscles,<br />

and sweat glands are appendageal structures commonly found in skin.<br />

Hairs are produced by hair follicles and are keratinized structures derived<br />

from epidermal invaginations that traverse the dermis and may extend<br />

- 28 -


into the hypodermis. Although skin penetration through a hair follicle still<br />

requires a compound to traverse the stratum corneum, follicles represent<br />

regions of greater surface areas and can, therefore, contribute to increased<br />

transdermal absorption (Monteiro-Riviere, 2004). Connective tissue at<br />

the base of the hair follicle provides an attachment site for the arrector pili<br />

muscle, which upon contraction not only erects the hair but also assists in<br />

emptying the sebaceous glands. Sebaceous glands release their secretory<br />

product, sebum, into ducts that empty into the canal of the hair follicle.<br />

Sebum is an oily secretion that acts as an antibacterial agent. Apocrine<br />

and eccrine sweat glands are also located in skin and function to produce<br />

secretions involved in communication and thermoregulation, respectively<br />

(Monteiro-Riviere and Stinsons, 1993).<br />

- 29 -


Percutaneous absorption<br />

The primary barrier against the passage of foreign hydrophilic<br />

substances into the skin is the stratum corneum. The stratum corneum<br />

consists of 10-15 layers of nonviable, protein rich cells surrounded by an<br />

extracellular lipid matrix. The intercellular lipid lamellae, composed<br />

mainly of ceramides, cholesterol, and fatty acids, are primarily<br />

responsible for restricting the passage of aqueous entities through the skin<br />

(Wertz, 2004). The importance of the lipid moieties in barrier function<br />

has been demonstrated by the removal of lipids from the stratum corneum,<br />

which subsequently results in an increased penetration of compounds<br />

(Hadgraft, 2001; Monteiro-Riviere et al., 2001). The stratum corneum<br />

serves as the rate-limiting barrier to percutaneous absorption because the<br />

underlying epidermal layers are much more aqueous in nature and, thus,<br />

allow the passage of substances to occur more easily. Once penetration<br />

through the epidermis occurs, there is little resistance to diffusion, and<br />

substances have access to systemic circulation via absorption into the<br />

blood and lymphatic vessels located in the dermis. Additionally,<br />

keratinocytes possess metabolizing enzymes that interact with the<br />

diffused compound and produce metabolites that can easily be absorbed<br />

by cutaneous vasculature (Monteiro-Riviere, 2001a; Riviere, 1990;<br />

Bronaugh et al., 1989).<br />

1. Pathways for transdermal drug delivery:<br />

Drugs can be diffused through the following pathways:<br />

1.1. Transappendagel:<br />

Diffusion occurs through hair follicle, sebaceous glands and<br />

eccrine glands<br />

- 30 -


1.2. Transepidermal:<br />

It is the most important pathway of drug permeation. As shown in<br />

(Figure 2) it is divided into:<br />

1.2.1. Intercellular bathway:<br />

It is the main route for permeation of the most drugs through<br />

intercellular spaces between the cells of stratum corneum, which is filled<br />

with a lipid, based lamellar crystalline structure (Moghimi et al., 1996,<br />

1997, and 1998).<br />

1.2.2. Transcellular pathway:<br />

Transport via corneocytes e.g. through protein-filled cell cytoplasm<br />

and protein-lipid cellular envelope (Moghimi et al., 1999).<br />

Figure 2: Diagrammatic representation of the stratum<br />

corneum and the intercellular and transcellular routes of penetration<br />

(Barry, 2001)<br />

- 31 -


2. Factors affecting percutaneous absorption:<br />

2.1. Physicochemical properties of the penterant molecules:<br />

2.1.1. Partition coefficient:<br />

The majority of topically applied drugs are covalent compounds in<br />

nature. Regardless of the types of vehicle used, at some point during the<br />

process of transdermal penetration the drug molecules have to dissolve<br />

and diffuse within the endogenous hydrated tissues of the stratum<br />

corneum. Drugs possessing both water and lipid solubility are favorably<br />

absorbed through the skin. Transdermal permeability coefficient a linear<br />

dependency on partition coefficient .A lipid/ water partition coefficient of<br />

one or greater is generally required for optimal tarnsdermal permeability.<br />

The drug substances should have a greater physicochemical attraction to<br />

the skin than to the vehicle in which it is presented (Chine, 1982).<br />

Molecules showing intermediate partition coefficients (log P<br />

octanol/water of 1-3) have adequate solubility within the lipid domains of<br />

the stratum corneum to permit diffusion through this domain whilst still<br />

having sufficient hydrophilic nature to allow partitioning into the viable<br />

tissues of the epidermis (Heather, 2005).<br />

2.1.2. pH conditions:<br />

The pH condition of the skin surface and in the drug delivery<br />

systems affect the extent of dissociation of ionogenic drug molecules and<br />

their transdermal permeability. The pH dependence of the transdermal<br />

permeability was related to the effect of the solution pH on the<br />

concentration of lipophilic, nonionized species of the drugs.<br />

- 32 -


2.1.3. Penetrant concentration :<br />

Transdermal permeability across mammalian skin is passive<br />

diffusion process and thus, depends on the concentration of penetrant<br />

molecules on the surface layers of the skin<br />

2.1.4. Penetrant solubility:<br />

According to Meyer-Overton theory of absorption , lipid soluble<br />

drugs pass through cell membrane owing to its lipid content while water<br />

soluble substances pass after hydration of protein particles in the cell<br />

wall which leaves the cell permeable to water soluble substances.<br />

2.1.5. Penetrant molecular weight:<br />

Rate of drug penetration is inversely proportional to its molecular<br />

weight, low molecular weight drugs penetrate faster than high molecular<br />

weight drugs.<br />

2.2. Physiological and pathological conditions of the skin:-<br />

2.2.1. Skin hydration:<br />

The moisture balance in the stratum corneum has been attributed to<br />

the presence of a combination of water soluble substances, known as<br />

natural moisturizing factor in the superfacial barrier layers .This factor is<br />

produced in the skin and is responsible for the hydration of the skin.<br />

Hydration of stratum corneum can enhance the transdermal permeability.<br />

Skin hydration can be achieved simply by covering or occluding the skin<br />

with pasting sheeting, leading to accumulation of sweat and condensed<br />

transpired water vapor .Increased hydration of stratum corneum appears<br />

to open up its dense, closely packed cells and increase its porosity<br />

resulting into increased permeation of drug molecules (Scheuplein and<br />

Ross, 1974).<br />

- 33 -


2.2.2. Skin temperature:<br />

A rise in skin temperature has been shown to have a definite effect<br />

on the percutaneous absorption of the drugs .This temperature-depentant<br />

increase in transdermal permeability was rationalized as due to the<br />

thermal energy required diffusivity and solubility of the drug in the skin<br />

tissues. Rises in skin temperature may also increase vasodilatation of the<br />

skin vessels leading to an increase in percutaneous absorption.<br />

2.2.3. Regional variation<br />

The permeation of water varies in different regions of the skin due<br />

to difference in the nature and thickness of the barrier layer (Wester and<br />

Maibach, 1999).<br />

2.2.4. Traumatic and pathologic injury to the skin:<br />

Injuries to the skin that disrupt the continuity of the stratum<br />

corneum are reported to increase skin permeability .The observed<br />

increase in the permeability may be due to the noticeable vasodilatation<br />

caused by the removal of barrier layer (Scott, 1991).<br />

<br />

2.2.5. Lipid film:<br />

The lipid film on the skin surface which contains emulsifying<br />

agents may provide a protective film to prevent the removal of natural<br />

moisturizing factor from the skin and play some limited role in<br />

maintaining the barrier function of the stratum corneum .<br />

2.3. Physicochemical properties of drug delivery system:<br />

2.3.1. Release characteristics:<br />

The affinity of the vehicle for the drug molecules can influence the<br />

release of the drug molecules from the vehicle. Solubility in the vehicle<br />

- 34 -


will determine the release rate of the drug. Generally, the more easily the<br />

drug is released from the drug delivery system, the higher the rate of<br />

tranedermal permeability. The mechanisms of the drug release depend on<br />

whether the drug molecules are dissolved or suspended in the delivery<br />

system and on the interfacial partition coefficient of the drug from the<br />

delivery system to the skin tissue.<br />

2.3.2. Composition of drug delivery system :<br />

The composition of drug delivery systems has a great influence on<br />

the percutaneous absorption of drug species. It may affect not only the<br />

rate of drug release, but also the permeability of the stratum corneum by<br />

means of hydration, mixing with lipids, or other sorption-promoting<br />

effects (Howes et al., 1999).<br />

2.3.3. Enhancement of transdermal permeation:<br />

Transdermal permeation of drugs can be improved by the addition<br />

of sorption or permeations promoters into the drug delivery systems.<br />

Sorption and permeation promoters are agents that have no therapeutic<br />

properties of their own but can promote the absorption of the drugs from<br />

the drug delivery systems onto the skin. Examples of permeation<br />

promoters are organic solvents and surface active agents<br />

3. Methods for studying percutaneous absorption:<br />

3.1. In vitro methods:<br />

The general advantage of in vitro method is to control the<br />

laboratory environment and so elucidate the individual factors, which<br />

modify drug penetration. Those methods are valuable for deducing<br />

physicochemical parameters such as fluxes, partition coefficient, and<br />

diffusion coefficient.<br />

- 35 -


3.1.1. Release method without a rate-limiting membrane :<br />

These procedures record the kinetics from a formulation to a<br />

simple immiscible phase, which is supposed to corresponding properties<br />

with human skin .Such techniques measure drug-vehicle interactions and<br />

the release characteristics of the formulation.<br />

3.1.2. Diffusion methods with a rate controlling membrane :<br />

3.1.2.1. Simulated skin membrane:<br />

Because human skin may be difficult to obtain and varies in its<br />

permeability, many workers use other materials to simulate it such as<br />

cellulose acetate membrane (Gary-Bobo et al., 1969; Diplo et al., 1970),<br />

silicone rubber (Flynn and Roseman, 1971; Bottari et al., 1977; Di colo<br />

et al., 1980), collagen (Nakano et al., 1976), and egg shell membrane.<br />

3.1.2.2. Natural skin membrane:<br />

Excised skin from a variety of animal including rats, mice, rabbits,<br />

guinea pigs has been used. Skin may be used immediately or stored at -24<br />

°C for a long time, and it may be subjected to greater extreme of heat,<br />

humidity, pH, and various fluids other biological tissues without<br />

irreversibly changing its barrier properties. Storage up to 6 months at -<br />

20°C leaves human skin permeability unaffected (Astley and Levine,<br />

1976). Elias et al., (1981) claims that a temperature as low as -70°C does<br />

not affect barrier properties.<br />

3.2. In-vivo methods:<br />

These include:-<br />

3.2.1. Animal models:<br />

3.2.2. Techniques:<br />

3.2.2.1. Observation of physiological or pharmacological response :<br />

- 36 -


If the penetrant stimulates a biological reaction when it reaches the<br />

viable tissue, then this response may provide the basis for determining the<br />

penetrant kinetics. The most productive technique in terms of<br />

biopharmaceutical application is the vasoconstrictor or balancing<br />

response to topical steroids.<br />

3.2.2.2. Physical properties of the skin:<br />

There are several methods used for measuring physical properties<br />

of the skin such as determination of transepidermal water loss, in addition<br />

to thermal determinations, mechanical analysis, and spectral analysis.<br />

3.2.2.3. Analysis of body tissues or fluids :<br />

Urinary analysis is often used to study percutaneous absorption<br />

(Wurster and Kramer, 1961; Butler, 1966; Fledmann and Maibach ,<br />

1965, 1966, 1967, 1968, 1969, 1970 ). Combination of blood, urine, and<br />

faeces analysis was used with rats, monkeys, and human volunteers to<br />

examine the percutaneous absorption and excretion of tritium-labeled<br />

diflorasone diacetate (Wickrema sinha et al., 1978).<br />

3.2.2.4. Surface loss :<br />

Measurements of the rate of loss of penetrant from an applied<br />

vehicle should lead to a determination of the flux of the material into the<br />

skin. The main use of a loss technique has been to monitor the decrease in<br />

radioactivity at skin surface (Malkinson, 1956, 1958, 1964; Ainsworth,<br />

1960; Wahlberg, 1965).<br />

3.2.2.5. Histology<br />

Histological techniques have elucidated absorption profiles and<br />

penetration routes for these few compounds which produce colored end<br />

- 37 -


products after chemical reaction for example, certain drugs change<br />

epidermal sulfhydryl groups in an easily detectable way (Bradshaw,<br />

1961; Chayen et al., 1970) few compounds fluoresce, and their behavior<br />

in skin may revealed by microscopy such as vitamin A, tetracycline, and<br />

benzpyrene.<br />

4. Theoretical advantages of transdermal routes for systemic therapy:<br />

Transdermal administration of drugs possesses several advantages<br />

in therapy compared with oral or parenteral adminsteration (Barry, 1991).<br />

These include:<br />

The avoidance of hepatic (first pass) metabolism by which the liver<br />

enzymes may reduce the amount of medicament passing into the<br />

system circulation.<br />

Transdermal input of a drug would avoid several variables which make<br />

gastrointestinal absorption a problem like dramatic change in pH,<br />

stomach emptying, intestinal motility, and the action of human and<br />

bacterial enzymes and the effect of food on drug absorption<br />

The percutaneous delivery may control the administration of highly<br />

potent drug, produce a relatively constant plasma level of drugs,<br />

concurrent decrease in side effects and improves patient compliance.<br />

The percutaneous administration can be valuable for drugs with low<br />

therapeutic indices and for which significant variation in plasma<br />

concentration are dangerous.<br />

Substitution for oral or potential administration in certain clinical<br />

situations (pediatrics, geriatrics and nausea).<br />

Ease of self administration.<br />

- 38 -


Types of skin preparations<br />

There are a large number of different types of external medicines,<br />

ranging from dry powders through semi-solid to liquids. (Figure 3)<br />

illustrates the formulation of the main types of preparation used on the<br />

skin.<br />

1. Solids:<br />

Dusting powders are applied to the skin for a surface effect such as<br />

drying or lubricating, or an antibacterial action. They are made of fine<br />

particle size powders together with any medicament (Winfield, 1998).<br />

2. Liquids:<br />

Soaks have an active ingredient dissolved in aqueous solvent and are<br />

often used as astringents, for cooling or to leave a film of solid on the<br />

skin. Oily vehicles can be used in bath additives to leave an emollient<br />

film on the skin surface.<br />

Liniments are alcoholic or oily solutions or emulsions designed to be<br />

rubbed into the skin. The medicament is usually a rubefacient.<br />

Lotions are aqueous solutions, suspensions or emulsions that cooled<br />

inflamed skin and deposit a protective layer of solid.<br />

Paints and tinctures are concentrated aqueous or alcoholic<br />

antimicrobial solutions.<br />

Collodions are organic solvents containing a polymer and keratolytic<br />

agent for treating corns and calluses.<br />

Emulsion is a dispersion in which the dispersed phase is composed of<br />

small globules of a liquid distributed through a vehicle in which it is<br />

immiscible by the aid of surfactant but it is thermodynamically unstable.<br />

<br />

pomades and foot washes (Winfield, 1998).<br />

- 39 -


- 40 -


3. Semi-solids:<br />

3.1. Ointments:<br />

Ointments are usually oily vehicles that may contain a surfactant to<br />

allow them to be washed off easily (barrier creams). They are used as<br />

emollients, or for drug delivery either to the surface or for deeper<br />

penetration (Winfield, 1998).<br />

Ointment bases are classified into:<br />

3.1.1. Hydrocarbon bases (oleaginous bases):<br />

These bases are immiscible with water and are not absorbed by the<br />

skin. They are almost inert and absorb very little water from a<br />

formulation or from skin exudates. However, they inhibit water loss from<br />

the skin by forming a waterproof film and by improving hydration, may<br />

encourage absorption of the medicaments through the skin.<br />

The constituents of hydrocarbon bases includes<br />

* Soft paraffin: There are two varieties, one is yellow and the other<br />

(bleached) form is white.<br />

* Hard paraffin which is used to stiffen ointment bases.<br />

* Liquid paraffin: It is used to soften ointment bases and to reduce the<br />

viscosity of creams.<br />

Hydrocarbon bases may contain ingredients additional to<br />

petrolatum, for example, paraffin ointment B.P. is a blend of white<br />

beeswax, hard paraffin, cetostearyl alcohol and soft paraffin (Collett,<br />

1991).<br />

3.1.2. Absorption bases:<br />

Absorption bases are less occlusive than the hydrocarbon bases and are<br />

easier to spread. They are good emollients. These bases absorb water and<br />

aqueous solutions to produce water-in-oil (W/O) emulsions. They consist<br />

- 41 -


of a mixture of sterol- type emulgent with one or more paraffins (Collett,<br />

1991).<br />

3.1.2.1. Non-emulsified:<br />

These constituents include:<br />

* Wool fat (anhydrous lanolin): It can absorb about 50% of its weight<br />

water.<br />

* Wool alcohols: This is the emulsifying fraction of wool fat.<br />

* Beeswax and cholesterol : They are included in some ointment bases to<br />

increase water-absorbing power.<br />

3.1.2.2. Water-in-oil emulsions:<br />

These are similar in properties to the previous group and are<br />

capable of absorbing more water. The constituents of emulsified<br />

absorption base include Hydrous Wool Fat BP (Lanolin) and Oily cream<br />

BP.<br />

3.1.3. Water-miscible bases (Emulsifying bases):<br />

Despite their hydrophilic nature, absorption base are difficult to<br />

wash from the skin. Although they can emulsify a large quantity of water<br />

they are immiscible with an excess. Ointments made from water-miscible<br />

bases are easily removed after use. The three emulsifying ointments from<br />

water-miscible bases, i.e. Emulsifying Ointment BP (anionic), Cetrimide<br />

Emulsifying Ointment BP (cationic) and Cetomacrogol Emulsifying<br />

Ointment BP (non-ionic). These contains paraffins and O/W emulgent<br />

and have the general formula:<br />

Anionic, cationic or non-ionic emulsifying wax 30%<br />

White Soft Paraffin 50%<br />

Liquid Paraffin 20%<br />

They are used for preparing O/W creams and as ointment bases<br />

when easy removal from the skin is advantageous. Other advantages of<br />

- 42 -


this type of base include, miscibility with exudates, good contact with the<br />

skin, high cosmetic acceptability and easy removal from the hair (Collett,<br />

1991).<br />

3.1.4. Water -soluble bases:<br />

Completely water-soluble bases have been developed the<br />

macrogols (polyethylene glycols). The macrogols vary in consistency<br />

from viscous liquids to waxy solids. They are non-toxic and non-irritating<br />

to the skin unless it is badly inflamed. Products with ointment-like<br />

consistency can be obtained by mixing liquid and waxy forms in suitable<br />

proportions. The water-soluble bases have the advantages of being non-<br />

occlusive, miscible with exudates, non-staining and easily removed by<br />

washing.<br />

The macrogol bases, being water-soluble, have the disadvantage of<br />

having a very limited capacity to take up water without a physical change,<br />

They are less bland than the paraffins and reduce the activity of a number<br />

of antimicrobial substances. They may also react with plastic closures<br />

(Collett, 1991).<br />

3.2. Pastes:<br />

Pastes are vehicles (aqueous or oily) with a high concentration of<br />

added solid. This makes them thick so they don not spread and so<br />

localizes drug delivery. They can also be used for sun-blocks (Winfield,<br />

1998).<br />

.<br />

3.3. Gels:<br />

Gels are transparent or translucent, non greasy, aqueous<br />

preparations (Collett, 1991). They are usually used for lubrication or<br />

applying a drug to the skin. Oily gels are also available where occlusion<br />

is required (Winfield, 1998).<br />

- 43 -


Bases for gels formulations:<br />

3.3.1. Tragacanth:<br />

Tagacanth gels are susceptible to microbial degradation and to<br />

changes in PH outside the range pH 4.5-7. Concentrations of tragacanth<br />

from 2% to 5 % produce gels of increasing viscosity.<br />

3.3.2. Sodium alginate:<br />

The viscosity of alginate gels is more standardized than that of<br />

tragacanth. A concentration of 1.5 % produces fluid gels and 5-10 % gels<br />

are suitable as dermatological vehicles.<br />

3.3.3. Pectin:<br />

Pectin gels are suitable for acid products. They are prone to<br />

microbial contamination and to water loss by evaporation and may<br />

require the inclusion of humectants.<br />

3.3.4. Starch gels:<br />

Starch gels are little used dermatological bases. Mucilages<br />

prepared with water alone lose by evaporation and are prone to microbial<br />

contamination. Glycerol concentrations of 50% or greater combine<br />

humectant and preservative functions.<br />

3.3.5. Gelatin:<br />

Gelatin forms gels at concentrations of 2-15 %. Gelatin gels are<br />

rarely used alone as a dermatological base but may be combined with<br />

other ingredients such as pectin.<br />

3.3.6. Polyvinyl alcohols:<br />

Polyvinyl alcohols (PVAs) have been used to prepare gels that dry<br />

very quickly. The residual film is strong and plastic, giving good contact<br />

between the skin and the medicament. The required concentration is<br />

usually between 10 %and 20 % depending on the grade of PVA and the<br />

desired viscosity.<br />

3.3.7. Clays:<br />

- 44 -


Gels containing 7-20 % of bentonite are used as dermatological<br />

bases. They are opalescent and lack the attractive clear appearance of<br />

many other types of gels.<br />

3.3.8. Carbomers :<br />

Neutralized carbomer gels are also used as bases for lubricants<br />

(0.3-1 %) and in dermatological preparations (0.5-5%). These gels are<br />

clear provided that an excessive amount of air is not incorporated during<br />

preparation.<br />

3.3.9. Cellulose derivatives:<br />

These are widely used because they produce neutral gels of stable<br />

viscosity, good resistance to microbial attack, high clarity and good film<br />

strength when dried on the skin. Methylcellulose 450 at a concentration<br />

of 3-5% produces satisfactory gels. Carmellose sodium (sodium<br />

carboxymethylcellulose) is easier to dissolve and the medium viscosity<br />

grade produces lubricant gels at a concentration of 1.5-5 % and<br />

dermatological gels at greater concentrations. Hypromellose<br />

(hydroxypropyl methylcellulose) form exceptionally clear gels which are<br />

used in ophthalamic products.<br />

Hypromellose, short for hydroxypropyl methylcellulose (HPMC),<br />

is a semisynthetic, inert, viscoelastic polymer used as an ophthalmic<br />

lubricant, as well as an excepient and controlled-delivery component in<br />

oral medicaments, found in a variety of commercial products (Collett,<br />

1991).<br />

3.4. Emulgels:<br />

Emulgel is a system consists of hydrophilic surfactant(s), oil, water,<br />

and gelling agent. Emulgel bases offer many advantages over other<br />

preparations:<br />

- 45 -


(i) They permit incorporation of aqueous and oleaginous ingredients, and<br />

their rheological properties can be controlled easily.<br />

(ii) They are easy to remove from a container in the desired quantity<br />

without waste.<br />

(iii) Upon application these preparations exhibit good spreadability; they<br />

can easily be applied to the desired part of the body without running or<br />

dripping and they are not tacky.<br />

The selection of oil phase, emulsifier and gelling agent is one of the most<br />

important factors in the preparation of emulgel bases.<br />

* Choice of oil phase:<br />

Many emulsions for external use contain oil which is present solely as a<br />

carrier for the active agent. It must be realized, however, that the type of<br />

oil used may also have an effect on the transport of the drug into the skin.<br />

One of the most widely used oil for this type of preparation is liquid<br />

paraffin. A variety of fixed oils of vegetable origin are also available, the<br />

most widely used being arachis, sesame, cotton seed and maize oils.<br />

* Choice of emulsifying agent:<br />

The inclusion of an emulsifying agent or agents is necessary to facilitate<br />

actual emulsification during manufacture and also to ensure emulsion<br />

stability during the shelf life of the product.<br />

* Choice of gelling agent:<br />

They are different gelling agents as mentioned before. They differ<br />

in their characteristics that affect the consistency of the emulgel (Balata,<br />

1999).<br />

- 46 -


4. Others:<br />

4.1. Submicron emulsions (SME):<br />

SME is liquid dispersion system formed by processing a medium-<br />

chain triglycerides emulsion with high-pressure homogenizer. SME has<br />

microparticles with diameter ranging from 3 to 10 <br />

layers of stratum corneum , increase its fluidity, disrupt barrier continuity,<br />

this result in slow, continuous, and controlled systemic delivery of the<br />

drug (Gupta and Garg, 2002).<br />

4.2. Microemulsion:<br />

Microemulsion is a liquid dispersion of water and oil with<br />

surfactant and co-surfactant. It is transparent, homogeneous, and<br />

thermodynamically stable, which provides sustained release effect after<br />

application on the skin over 24 hours (El-Nokaly and Cornell, 1990).<br />

4.3. Microsponges :<br />

Microsponges are polymeric delivery system consisting of porous<br />

microspheres that can entrap several active substances such as anti-fungal,<br />

anti-infective, and anti-inflammatory. Those systems can be incorporated<br />

into creams, lotions, powders, soaps from which the entrapped substances<br />

are released to the skin in controlled- release manner (Report, 1992).<br />

4.4. Transfersomes:<br />

Transfersomes are vesicles made from phosphatidylcholine and<br />

contained at least one component that controllably destabilizes lipid<br />

bilayers and makes the vesicles very deformable. Such additives are bile<br />

salts, polysorbate, glycolipids, and alkyl or acyl-polyethoxylenes.<br />

Transfersomes are applied to the skin to achieve sustained drug release,<br />

- 47 -


and in this way skin surface acting as reservoir for drug as well as carrier<br />

(Cevc, 2003).<br />

4.5. Niosomes:<br />

Niosomes are unilameller or multilamellar vesicles where in<br />

aqueous solutions are enclosed in highly ordered bilayers made up of non<br />

ionic surfactants with or without cholesterol and diacetyl phosphate<br />

(Namdeo and Jain, 1996). Niosomes are supposed to give desirable<br />

interaction with human skin when applied in topical preparations by<br />

reducing transepidermal water loss and by increasing smoothness via<br />

replenishing lost skin lipids.<br />

4.6. Liposomes :<br />

Liposomes are concentric bilayered structures made of amphipathic<br />

phospholipids and depending on the number of bilayers; liposomes are<br />

classified as multilamaller vesicles (MLVs), small unilamaller vesicles<br />

(SUVs), or large unilamaller vesicles (LUVs). They range in size<br />

from .025 – <br />

regulated by the method of preparation and composition (Kshirsagr,<br />

2000).<br />

4.7. Solid lipid nanoparticles (SLNs):<br />

SLNs consist of physiological and biocompatible lipids, prepared<br />

by several techniques such as hot and cold dispersion of lipids and high<br />

pressure homogenization of melted lipids (Schwartz et al., 1992; Domb,<br />

1995; Westesen et al., 1998; Yang et al., 1999). SLNs posses the<br />

advantages of better drug penetration because their small particle size<br />

ensure close contact to stratum corneum and increase the amount of<br />

encapsulated drug penetrating skin. SLNs provide both burst and<br />

- 48 -


sustained drug release and they can be incorporated into aqueous gel or<br />

creams in which stability is maintained (Gupta and Garg, 2002).<br />

4.8. Microneedles:<br />

Microneedle concept employs an array of micron-scale needles that<br />

inserted into skin sufficiently far that it can deliver drug into the body, but<br />

not so far that it hits nerves there by avoids causing pain (Prausnitz et al.,<br />

2003).<br />

4.9. Metred- dose transdemal spray (MDTS):<br />

MDTS is a topical solution made up of a volatile:non-volatile<br />

vehicle containing drug dissolved as a single phase solution. Upon<br />

application to the skin, evaporation of the volatile component of the<br />

vehicle occurs leaving the remaining non-volatile penetration enhancer<br />

and drug to partition into the stratum corneum during the first minute<br />

after application, resulting in stratum corneum reservoir of drug and<br />

enhancer which releases the drug in sustained pattern (Morgan et al.,<br />

1998).<br />

4.10. Macroflux tehnology:<br />

Macroflux system incorporates a titanium microprojection array<br />

that creates superficial pathways through the skin barrier layers to allow<br />

transportation of therapeutic proteins and vaccines that currently require<br />

parentral administration (Cormier and Daddona, 2003).<br />

4.11. Transdemal drug delivery devices (TDDS) :<br />

1987):<br />

TDDS are broadly classified into the following types (Chien,<br />

- 49 -


4.11.1. Reservoir systems:<br />

In these systems, the drug reservoir is embedded between an<br />

impervious backing layer and a rate controlling membrane. The drug<br />

release only through the rate controlling membrane, can be microporous<br />

or non-porous. In the drug reservoir compartment, the drug can be in the<br />

form of solution, suspension, gel, or embedded in a solid polymer matrix.<br />

On the outer surface of the polymeric membrane a thin layer of drug-<br />

compatible, hypoallergenic adhesive polymer can be applied.<br />

4.11.2. Matrix systems. Drug in adhesive system:<br />

The drug reservoir is formed by dispersing the drug in an adhesive<br />

polymer and then spreading the medicated polymer adhesive by solvent<br />

casting or by melting the adhesive onto an impervious backing layer. On<br />

top of the reservoir, layers of unmedicated adhesive polymer are applied.<br />

4.11.3. Matrix dispersion system:<br />

The drug is dispersed homogeneously in a hydrophilic or lipophilic<br />

polymer matrix. This drug containing polymer disc then is fixed onto on<br />

occlusive base plate in a compartment fabricated from a drug-<br />

impermeable backing layer. Instead of applying the adhesive on the face<br />

of the drug reservoir, it is spread along the circumference to form a strip<br />

of adhesive rim.<br />

4.11.4. Microreservoir system:<br />

This drug delivery system is a combination of reservoir and matrix<br />

dispersion system. The drug reservoir is formed by first suspending the<br />

drug in an aqueous solution of water-soluble polymer and then dispersing<br />

- 50 -


the solution homogeneously in a lipophilic polymer to form thousands of<br />

unleachable, microscopic spheres of drug reservoirs. The<br />

thermodynamically unstable dispersion is stabilized quickly by<br />

immediately cross-linking the polymer insitu.<br />

- 51 -


Diabetes mellitus<br />

Diabetes mellitus is a group of disorders of carbohydrate<br />

metabolism in which the action of insulin is diminished or absent through<br />

altered secretion, decreased insulin activity, or a combination of both<br />

factors. It is characterised by hyperglycaemia. As the disease progresses<br />

tissue or vascular damage ensues leading to severe complications such as<br />

retinopathy, nephropathy, neuropathy, cardiovascular disease, and foot<br />

ulceration.<br />

Diabetes mellitus may be categorised into several types but the two<br />

major types are type 1 (insulin-dependent diabetes mellitus; IDDM) and<br />

type 2 (non-insulin-dependent diabetes mellitus; NIDDM). The term<br />

juvenile-onset diabetes has sometimes been used for type 1 and maturity-<br />

onset diabetes for type 2 (Martindale, 1996) .<br />

Oral Antidiabetics<br />

If patients with type 2 diabetes have not achieved suitable control<br />

after about 3 months of dietary modification and increased physical<br />

activity, then oral antidiabetics (oral hypoglycaemics) may be tried. The<br />

two major classes are the sulfonylureas and the biguanides. Sulfonylureas<br />

act mainly by increasing endogenous insulin secretion, while biguanides<br />

act chiefly by decreasing hepatic gluconeogenesis and increasing<br />

peripheral utilisation of glucose. Both types function only in the presence<br />

of some endogenous insulin production. More recently developed classes<br />

of oral antidiabetics include the alpha-glucosidase inhibitors, the<br />

meglitinides, and the thiazolidinediones. Alpha-glucosidase inhibitors act<br />

by delaying the absorption of glucose from the gastrointestinal tract;<br />

meglitinides increase endogenous insulin secretion; and<br />

- 52 -


thiazolidinediones appear to increase insulin sensitivity (Martindale,<br />

1996).<br />

1.Mode of action:<br />

Sulphonylurea<br />

Sulfonylureas appear to have several modes of action, apparently<br />

mediated by inhibition of ATP-sensitive potassium channels. Initially,<br />

secretion of insulin by functioning islet beta cells is increased. However,<br />

insulin secretion subsequently falls again but the hypoglycaemic effect<br />

persists and may be due to inhibition of hepatic glucose production and<br />

increased sensitivity to any available insulin; this may explain the<br />

observed clinical improvement in glycaemic control (Martindale, 1996).<br />

2.<br />

Uses and Administration:<br />

The sulfonylurea antidiabetics are a class of oral antidiabetic drugs<br />

used in the treatment of type 2 diabetes mellitus. They are given to<br />

supplement treatment by diet modification when such modification has<br />

not proved effective on its own, although metformin is preferred in<br />

patients who are obese (Martindale, 1996) .<br />

3. Adverse effects:<br />

Gastrointestinal disturbances such as nausea, vomiting, heartburn,<br />

anorexia, diarrhoea, and a metallic taste may occur with sulfonylureas<br />

and are usually mild and dose-dependent; increased appetite and weight<br />

gain may occur. Skin rashes and pruritus may occur and photosensitivity<br />

has been reported. Rashes are usually hypersensitivity reactions and may<br />

progress to more serious disorders . Facial flushing may develop in<br />

patients receiving sulfonylureas, particularly chlorpropamide, when<br />

alcohol is consumed .<br />

Mild hypoglycaemia may occur; severe hypoglycaemia is usually<br />

an indication of overdosage and is relatively uncommon. Hypoglycaemia<br />

- 53 -


is more likely with long-acting sulfonylureas such as chlorpropamide and<br />

glibenclamide, which have been associated with severe, prolonged, and<br />

sometimes fatal hypoglycaemia.<br />

Other severe effects may be manifestations of a hypersensitivity<br />

reaction. They include altered liver enzyme values, hepatitis and<br />

cholestatic jaundice, leucopenia, thrombocytopenia, aplastic anaemia,<br />

agranulocytosis, haemolytic anaemia, erythema multiforme or the<br />

Stevens-Johnson syndrome, exfoliative dermatitis, and erythema<br />

nodosum.<br />

The sulfonylureas, particularly chlorpropamide, occasionally<br />

induce a syndrome of inappropriate secretion of antidiuretic hormone<br />

(SIADH) characterised by water retention, hyponatraemia, and CNS<br />

effects. However, some sulfonylureas, such as glibenclamide, glipizide,<br />

and tolazamide are also stated to have mild diuretic actions (Martindale,<br />

1996) .<br />

4. Precautions:<br />

Sulfonylureas should not be used in type 1 diabetes mellitus. Use in<br />

type 2 diabetes mellitus is contra-indicated in patients with ketoacidosis<br />

and in those with severe infection, trauma, or other severe conditions<br />

where the sulfonylurea is unlikely to control the hyperglycaemia; insulin<br />

should be used in such situations.<br />

Insulin is also preferred for therapy during pregnancy.<br />

Sulfonylureas with a long half-life such as chlorpropamide or<br />

glibenclamide are associated with an increased risk of hypoglycaemia.<br />

They should therefore be avoided in patients with impairment of renal or<br />

hepatic function, and a similar precaution would tend to apply in other<br />

groups with an increased susceptibility to this effect, such as the elderly,<br />

debilitated or malnourished patients, and those with adrenal or pituitary<br />

- 54 -


insufficiency. Irregular mealtimes, missed meals, changes in diet, or<br />

prolonged exercise may also provoke hypoglycaemia. Where a<br />

sulfonylurea needs to be used in patients at increased risk of<br />

hypoglycaemia, a short-acting drug such as tolbutamide, gliquidone, or<br />

gliclazide may be preferred; these three sulfonylureas, being principally<br />

inactivated in the liver, are perhaps particularly suitable in renal<br />

impairment, although careful monitoring of blood-glucose concentration<br />

is essential (Martindale, 1996) .<br />

- 55 -


- 56 -


Scope of Work<br />

Sulfonylureas are widely used as oral hypoglycemic drugs in the<br />

treatment of non insulin dependent diabetes mellitus (NIDDM). Since<br />

sulfonylureas are usually taken for a long period, the compliance of the<br />

patients is very important. Therefore, for the improvement of the<br />

compliance of the patients, the development of a transdermal dosage form<br />

of sulfonylureas was attempted in this study (Takahashi et al., 1997). In<br />

addition, Sulfonylureas have associated with severe and sometimes fetal<br />

hypoglycemia and gastric disturbances like nausea, vomiting, heartburn,<br />

anorexia and increased appetite after oral therapy. The feasibility of<br />

application of transdermal delivery for some sulfonylureas was also<br />

previously reported (Srinivas and Nayanabhirama, 2005).<br />

The present work is concerned with the pharmaceutical<br />

formulation of certain sulfonylureas namely, gliclazide and glibenclamide<br />

in different bases for topical application. The bases include water soluble,<br />

emulsion, oleaginous, absorption, gel and emulgel bases. The in vitro<br />

release of these drugs from the above mentioned bases was also studied<br />

Gliclazide is practically insoluble in water; therefore, the<br />

improvement of its dissolution is an important issue for enhancing its<br />

bioavailability and therapeutic efficacy. Accordingly, solid dispersions of<br />

gliclazide in different carrier systems (PEG 4000, PEG 6000, glucose and<br />

urea) were prepared. The solid phases obtained were characterized by<br />

Fourier transform infrared spectroscopy, differential scanning calorimetry<br />

and X-ray powder diffraction. Solubility diagrams and dissolution studies<br />

were also carried out.<br />

- 57 -


The role of improvement of gliclazide dissolution on the release<br />

rate of gliclazide from different topical preparations mentioned above was<br />

also studied.<br />

Studies have been carried out to find suitable enhancers to<br />

promote the percutaneous absorption of glibenclamide; therefore, the<br />

effect of certain penetration enhancers with different concentrations on<br />

the release of the glibenclamide from water soluble base was<br />

demonstrated.<br />

In vivo experiments were carried out in order to demonstrate<br />

the blood glucose reducing hypoglycemic activity of gliclazide and<br />

glibenclamide systems in both normal and diabetic rats. Drugs were<br />

applied topically and compared to an orally administrated doses.<br />

- 58 -


- 59 -


1. Description:<br />

Introduction<br />

Gliclazide<br />

1.1.Name, formula, molecular weight:<br />

1-(3-Azabicyclo [3.3.0] oct-3-yl)-3-tosyl urea<br />

Figure 10: Structure of Gliclazide.<br />

Molecular weight: 323.4 C15 H21 N3 O3 S<br />

1.2. Appearance, odour and colour:<br />

without taste.<br />

Glz is a white, crystalline, odourless powder and practically<br />

2. Physical properties:<br />

2.1. Melting point:<br />

181°C.<br />

2.2. Solubility:<br />

Practically insoluble in water; slightly soluble in alcohol;<br />

sparingly soluble in acetone; freely soluble in dichloromethane.<br />

3. Pharmacokinetics:<br />

Glz is readily absorbed from the gastrointestinal tract. It is<br />

extensively bound to plasma proteins. The half-life is about 10 to 12<br />

- 60 -


hours. Glz is extensively metabolized in the liver to metabolites that have<br />

no significant hypoglycaemic activity. Metabolites and a small amount of<br />

unchanged drug are excreted in the urine (Martindale, 1996).<br />

4. Mode of action:<br />

5. Dosage and adminstration:<br />

As mentioned before under sulfonylureas.<br />

It is given by mouth in the treatment of type 2 diabetes mellitus and<br />

has duration of action of 12 to 24 hours. Because its effects are less<br />

prolonged than those of chlorpropamide or glibenclamide it may be more<br />

suitable for elderly patients, who are prone to hypoglycaemia with longer-<br />

acting sulfonylURs. The usual initial dose is 40 to 80 mg daily, gradually<br />

increased, if necessary, up to 320 mg daily. Doses of more than 160 mg<br />

daily are given in 2 divided doses. A modified-release tablet is also<br />

available: the usual initial dose is 30 mg once daily, increased if<br />

necessary up to a maximum of 120 mg daily (Martindale, 1996).<br />

.<br />

6. Precautions:<br />

7. Adverse Effects:<br />

8. Interactions:<br />

As mentioned before under sulfonylureas.<br />

As mentioned before under sulfonylureas.<br />

An increased hypoglycaemic effect has occurred or might be<br />

expected with ACE inhibitors, alcohol, allopurinol, some analgesics<br />

(notably azapropazone, phenylbutazone, and the salicylates), azole<br />

- 61 -


antifungals (fluconazole, ketoconazole, and miconazole),<br />

chloramphenicol, cimetidine, clofibrate and related compounds, coumarin<br />

anticoagulants, fluoroquinolones, heparin, MAOIs, octreotide (although<br />

this may also produce hyperglycaemia), ranitidine, sulfinpyrazone,<br />

sulfonamides (including co-trimoxazole), tetracyclines, and tricyclic<br />

antidepressants.<br />

Beta blockers have been reported both to increase hypoglycaemia<br />

and to mask the typical sympathetic warning signs. There are sporadic<br />

and conflicting reports of a possible interaction with calcium-channel<br />

blockers, but overall any effect seems to be of little clinical significance.<br />

In addition to producing hypoglycaemia alcohol can interact with<br />

chlorpropamide to produce an unpleasant flushing reaction. Such an<br />

effect is rare with other sulfonylureas and alcohol (Martindale, 1996).<br />

- 62 -


- 63 -


Introduction<br />

Therapeutic effectiveness of a drug depends upon the bioavailability<br />

and ultimately upon the solubility of drug molecules. Solubility is one of<br />

the important parameter to achieve desired concentration of drug in<br />

systemic circulation for pharmacological response to be shown. Currently<br />

only 8% of new drug candidates have both high solubility and<br />

permeability .<br />

The solubility of a solute is the maximum quantity of solute that can<br />

dissolve in a certain quantity of solvent or quantity of solution at a<br />

specified temperature .<br />

In the other words the solubility can also define as the ability of one<br />

substance to form a solution with another substance.<br />

The substance to be dissolved is called as solute and the dissolving<br />

fluid in which the solute dissolve is called as solvent, which together<br />

form a solution (Anil et al., 2007).<br />

1. Process of solubilisation:<br />

As shown in (Figure 4), the process of solubilisation involves the<br />

breaking of inter-ionic or intermolecular bonds in the solute, the<br />

separation of the molecules of the solvent to provide space in the solvent<br />

for the solute, interaction between the solvent and the solute molecule or<br />

ion (Anil et al., 2007).<br />

.<br />

Step 1: Holes opens in the solvent.<br />

- 64 -


Step2: Molecules of the solid breaks away from the bulk.<br />

Step 3: The freed solid molecule is intergrated into the hole in the<br />

solvent.<br />

Figure 4: Diagramatic representation of process of solubilization.<br />

2. Factors affecting solubility:<br />

The solubility depends on the physical form of the solid, the nature<br />

and composition of solvent medium as well as temperature and pressure<br />

of system (Anil et al., 2007).<br />

2.1. Particle Size:<br />

The size of the solid particle influences the solubility because as a<br />

particle becomes smaller, the surface area to volume ratio increases. The<br />

larger surface area allows a greater interaction with the solvent.<br />

- 65 -


2.2. Temperature:<br />

Temperature will affect solubility. If the solution process absorbs<br />

energy then the solubility will be increased as the temperature is<br />

increased. If the solution process releases energy then the solubility will<br />

decrease with increasing temperature. Generally, an increase in the<br />

temperature of the solution increases the solubility of a solid solute. A<br />

few solid solutes are less soluble in warm solutions. For all gases,<br />

solubility decreases as the temperature of the solution increases (Anil et<br />

al., 2007).<br />

2.3. Pressure:<br />

For gaseous solutes, an increase in pressure increases solubility and<br />

a decrease in pressure decrease the solubility. For solids and liquid<br />

solutes, changes in pressure have practically no effect on solubility.<br />

2.4. Nature of the solute and solvent:<br />

While only 1 gram of lead (II) chloride can be dissolved in 100<br />

grams of water at room temperature, 200 grams of zinc chloride can be<br />

dissolved. The great difference in the solubilities of these two substances<br />

is the result of differences in their natures.<br />

2.5. Molecular size:<br />

Molecular size will affect the solubility. The larger the molecule or<br />

the higher its molecular weight the less soluble the substance. Larger<br />

molecules are more difficult to surround with solvent molecules in order<br />

to solvate the substance. In the case of organic compounds the amount of<br />

carbon branching will increase the solubility since more branching will<br />

reduce the size (or volume) of the molecule and make it easier to solvate<br />

the molecules with solvent.<br />

- 66 -


2.6. Polarity:<br />

Polarity of the solute and solvent molecules will affect the solubility.<br />

Generally non-polar solute molecules will dissolve in non-polar solvents<br />

and polar solute molecules will dissolve in polar solvents .<br />

2.7. Polymorphs:<br />

Polymorphs can vary in melting point. Since the melting point of the<br />

solid is related to solubility, so polymorphs will have different solubilities<br />

(Anil et al., 2007). Generally the range of solubility differences between<br />

different polymorphs is only 2-3 folds due to relatively small differences<br />

in free energy (Singhal and Curatolo, 2004)<br />

3. Techniques of solubility enhancement:<br />

There are various techniques available to improve the solubility of<br />

poorly soluble drugs. Some of the approaches to improve the solubility<br />

are (Pinnamaneni et al., 2002):<br />

3.1.<br />

Particle size reduction:<br />

Particle size reduction can be achieved by micronisation and<br />

nanosuspension. Each technique utilizes different equipments for<br />

reduction of the particle size.<br />

3.1.1 Micronization:<br />

Micronisation increases the dissolution rate of drugs through increased<br />

surface area, it does not increase equilibrium solubility (Chaumeil, 1998).<br />

Micronization of drugs is done by milling techniques using jet mill, rotor<br />

stator colloid mills etc. Micronization is not suitable for drugs having a<br />

high dose number because it does not change the saturation solubility of<br />

the drug.<br />

- 67 -


3.1.2. Nanosuspension:<br />

Nanosuspensions are sub-micron colloidal dispersion of pure<br />

particles of drug, which are stabilised by surfactants ( Anil et al., 2007).<br />

The advantages offered by nanosuspension to increase dissolution rate is<br />

due to larger surface area exposed, while absence of Ostwald ripening is<br />

due to the uniform and narrow particle size range obtained, which<br />

eliminates the concentration gradient factor.<br />

3.2. Modification of the crystal habit:<br />

Polymorphism is the ability of an element or compound to<br />

crystallize in more than one crystalline form. Different polymorphs of<br />

drugs are chemically identical, but they exhibit different physicochemical<br />

properties including solubility, melting point, density, texture, stability<br />

etc.<br />

Some drugs can exist in amorphous form (i.e. having no internal<br />

crystal structure). Such drugs represent the highest energy state and can<br />

be considered as super cooled liquids. They have greater aqueous<br />

solubility than the crystalline forms because they require less energy to<br />

transfer a molecule into solvent.<br />

3.3. Complexation:<br />

Complexation is the association between two or more molecules to<br />

form a nonbonded entity with a well defined stichiometry. Complexation<br />

relies on relatively weak forces such as London forces, hydrogen bonding<br />

and hydrophobic interactions.<br />

3.4. Solubilization by surfactants:<br />

Surfactants are molecules with distinct polar and nonpolar regions.<br />

Most surfactants consist of a hydrocarbon segment connected to a polar<br />

- 68 -


group. The polar group can be anionic, cationic, zwitterionic or nonionic<br />

(Swarbrick and Boylan, 2002). When small apolar molecules are added<br />

they can accumulate in the hydrophobic core of the micelles. This process<br />

of solubilization is very important in industrial and biological processes.<br />

The presence of surfactants may lower the surface tension and increase<br />

the solubility of the drug within an organic solvent (Anil et al., 2007).<br />

3.5. Cosolvency:<br />

The solubilisation of drugs in co-solvents is an another technique for<br />

improving the solubility of poorly soluble drug (Amin et al., 2004). It is<br />

well-known that the addition of an organic cosolvent to water can<br />

dramatically change the solubility of drugs (Yalkowsky and Roseman,<br />

1981).<br />

Weak electrolytes and nonpolar molecules have poor water<br />

solubility and it can be improved by altering polarity of the solvent. This<br />

can be achieved by<br />

addition of another solvent. This process is known as cosolvency. Solvent<br />

used to increase solubility known as cosolvent. Cosolvent system works<br />

by reducing the interfacial tension between the aqueous solution and<br />

hydrophobic solute. It is also commonly referred to as solvent blending<br />

(Joseph, 2002).<br />

3.6. Chemical Modifications:<br />

For organic solutes that are ionizable, changing the pH of the system<br />

may be simplest and most effective means of increasing aqueous<br />

solubility. Under the proper conditions, the solubility of an ionizable drug<br />

can increase exponentially by adjusting the pH of the solution. A drug<br />

that can be efficiently solubilized by pH control should be either weak<br />

acid with a low pKa or a weak base with a high pKa (Anil et al., 2007) .<br />

- 69 -


The use of salt forms is a well known technique to enhanced<br />

dissolution profiles (Agharkar et al., 1976). Salt formation is the most<br />

common and effective method of increasing solubility and dissolution<br />

rates of acidic and basic drugs (Serajuddin, 2007). An alkaloid base is,<br />

generally, slightly soluble in water, but if the pH of medium is reduced by<br />

addition of acid, the solubility of the base is increased as the pH continues<br />

to be reduced. The reason for this increase in solubility is that the base is<br />

converted to a salt, which is relatively soluble in water (e.g. Tribasic<br />

calcium phosphate)<br />

3.7. Solid dispersions:<br />

A solid dispersion may be defined as a dispersion of one or more<br />

active ingredients in an inert carrier or matrix in the solid state prepared<br />

by the melting, solvent, or melting-solvent method (Chiou and<br />

Riegelman, 1971).<br />

3.7.1. Advantages of solid dispersions over other strategies to<br />

improve bioavailability of poorly water soluble drugs:<br />

Improving drug bioavailability by changing their water solubility<br />

has been possible by chemical or formulation approaches (Majerik et al.,<br />

2007; Yoshihashi et al., 2006; Cutler et al., 2006).<br />

Chemical approaches to improving bioavailability without changing<br />

the active target can be achieved by salt formation or by incorporating<br />

polar or ionizable groups in the main drug structure, resulting in the<br />

formation of a pro-drug. Solid dispersions appear to be a better approach<br />

to improve drug solubility than these techniques, because they are easier<br />

to produce and more applicable. For instance, salt formation can only be<br />

used for weakly acidic or basic drugs and not for neutral. Furthermore, it<br />

- 70 -


is common that salt formation does not achieve better bioavailability<br />

because of its in vivo conversion into acidic or basic forms (Serajuddin,<br />

1999; Karavas et al., 2006).<br />

Formulation approaches include solubilization and particle size<br />

reduction techniques, and solid dispersions, among others. Solid<br />

dispersions are more acceptable to patients than solubilization products,<br />

since they give rise to solid oral dosage forms instead of liquid as<br />

solubilization products usually do (Serajuddin, 1999; Karavas et al.,<br />

2006). Milling or micronization for particle size reduction are commonly<br />

performed as approaches to improve solubility, on the basis of the<br />

increase in surface area ( Pouton, 2006; Craig, 2002). Solid dispersions<br />

are more efficient than these particle size reduction techniques, since the<br />

latter have a particle size reduction limit around 2–5 <br />

is not enough to improve considerably the drug solubility or drug release<br />

in the small intestine (Pouton, 2006; Karavas et al, 2006; Muhrer et al.,<br />

2006) and, consequently, to improve the bioavailability (Serajuddin,<br />

1999; Karavas et al., 2006; Rasenack and Muller, 2004). Moreover,<br />

solid powders with such a low particle size have poor mechanical<br />

properties, such as low flow and high adhesion, and are extremely<br />

difficult to handle (Pouton, 2006; Karavas et al, 2006; Muhrer et al.,<br />

2006) .<br />

3.7.2. Solid dispersions disadvantages:<br />

Despite extensive expertise with solid dispersions, they are not<br />

broadly used in commercial products, mainly because there is the<br />

possibility that during processing (mechanical stress) or storage<br />

(temperature and humidity stress) the amorphous state may undergo<br />

crystallization (Pokharkar et al., 2006; Van den Mooter et al., 2006;<br />

Chauhan et al., 2005; Vasanthavada et al., 2004). The effect of<br />

- 71 -


moisture on the storage stability of amorphous pharmaceuticals is also a<br />

significant concern, because it may increase drug mobility and promote<br />

drug crystallization (Vasanthavada et al., 2004; Johari et al., 2005).<br />

Moreover, most of the polymers used in solid dispersions can absorb<br />

moisture, which may result in phase separation, crystal growth or<br />

conversion from the amorphous to the crystalline state or from a<br />

metastable crystalline form to a more stable structure during storage. This<br />

may result in decreased solubility and dissolution rate (Van den Mooter<br />

et al., 2006; Wang et al., 2005). Therefore, exploitation of the full<br />

potential of amorphous solids requires their stabilization in solid state, as<br />

well as during in vivo performance (Pokharkar et al., 2006).<br />

3.7.3. The advantageous properties of solid dispersions:<br />

Management of the drug release profile using solid dispersions is<br />

achieved by manipulation of the carrier and solid dispersion particles<br />

properties. Parameters, such as carrier molecular weight and composition,<br />

drug crystallinity and particle porosity and wettability, when successfully<br />

controlled, can produce improvements in bioavailability (Ghaderi et al.,<br />

1999).<br />

3.7.3.1. Particles with reduced particle size:<br />

Molecular dispersions, as solid dispersions, represent the last state<br />

on particle size reduction, and after carrier dissolution the drug is<br />

molecularly dispersed in the dissolution medium. Solid dispersions apply<br />

this principle to drug release by creating a mixture of a poorly water<br />

soluble drug and highly soluble carriers (Leuner and Dressman, 2000).<br />

A high surface area is formed, resulting in an increased dissolution rate<br />

and, consequently, improved bioavailability (Leuner and Dressman,<br />

2000; Bikiaris et al., 2005).<br />

- 72 -


3.7.3.2. Particles with improved wettability:<br />

A strong contribution to the enhancement of drug solubility is<br />

related to the drug wettability improvement verified in solid dispersions<br />

(Karavas et al., 2006). It was observed that even carriers without any<br />

surface activity, such as urea (Sekiguchi and Obi, 1964) improved drug<br />

wettability. Carriers with surface activity, such as cholic acid and bile<br />

salts, when used, can significantly increase the wettability properties of<br />

drugs. Moreover, carriers can influence the drug dissolution profile by<br />

direct dissolution or co-solvent effects (Pouton, 2006; Leuner and<br />

Dressman, 2000; Kang et al., 2004). Recently, the inclusion of<br />

surfactants (Van den Mooter et al., 2006; Ghebremeskel et al., 2007) in<br />

the third generation solid dispersions reinforced the importance of this<br />

property.<br />

3.7.3.3. Particles with higher porosity:<br />

Particles in solid dispersions have been found to have a higher<br />

degree of porosity (Vasconcelos, and Costa, 2007). The increase in<br />

porosity also depends on the carrier properties, for instance, solid<br />

dispersions containing linear polymers produce larger and more porous<br />

particles than those containing reticular polymers and, therefore, result in<br />

a higher dissolution rate. The increased porosity of solid dispersion<br />

particles also hastens the drug release profile (Ghaderi et al.,1999;<br />

Vasconcelos, and Costa, 2007).<br />

3.7.3.4. Drugs in amorphous state:<br />

Poorly water soluble crystalline drugs, when in the amorphous state<br />

tend to have higher solubility (Pokharkar et al., 2006; Lloyd et al.,<br />

1999). The enhancement of drug release can usually be achieved using<br />

the drug in its amorphous state, because no energy is required to break up<br />

- 73 -


the crystal lattice during the dissolution process (Taylor and Zografi,<br />

1997) . In solid dispersions, drugs are presented as supersaturated<br />

solutions after system dissolution, and it is speculated that, if drugs<br />

precipitate, it is as a metastable polymorphic form with higher solubility<br />

than the most stable crystal form ( Leuner and Dressman, 2000;<br />

Van den Mooter et al., 2006; Karavas et al., 2006).<br />

For drugs with low crystal energy (low melting temperature or heat<br />

of fusion), the amorphous composition is primarily dictated by the<br />

difference in melting temperature between drug and carrier. For drugs<br />

with high crystal energy, higher amorphous compositions can be obtained<br />

by choosing carriers, which exhibit specific interactions with them<br />

(Vippagunta et al., 2006).<br />

3.7.4.<br />

Method for preparation of solid dispersions:<br />

3.7.4.1.<br />

Melting method:<br />

The physical mixture of a drug in a water soluble carrier is heated<br />

directly until it melts. The melted mixture is then cooled and solidified<br />

rapidly while vigorously stirred. The final solid mass is crushed,<br />

pulverized and sieved. A disadvantage is that many substances either<br />

drugs or carriers may decompose or evaporate during the fusion process<br />

at high temperatures. However, this evaporation problem may be avoided<br />

if the physical mixture is heated in a sealed container. Melting under a<br />

vacuum or blanket of an inert gas such as nitrogen may be employed to<br />

prevent oxidation of the drug or carrier. Another disadvantage is the<br />

drug/carrier immiscibility and the consequent irregular crystallization<br />

may lead to only moderate increases in dissolution rate and difficulties in<br />

formulation (Wrenn and Simeon, 1998). Currently, the melting method<br />

is known as “hot melt technology” and provides pharmaceutical<br />

technologists with new possibilities.<br />

- 74 -


3.7.4.1.1. Direct melt filling:<br />

In 1978, Francois and Jones, further developed the solid dispersion<br />

method by directly filling hard gelatin capsules with semisolid materials<br />

as a melt, which solidified at room temperature. Catham 1987 reported<br />

the possibility of preparing PEG-based solid dispersions by filling drug-<br />

PEG melts into hard gelatin capsules.<br />

3.7.4.1.2<br />

Melt extrusion:<br />

Melt extrusion is a new method for producing solid dispersions.<br />

Special equipment is needed to develop the dosage form solid dispersions,<br />

which limits the use of the extrusion method. Forster at al., 2002,<br />

reported the use of melt extrusion to prepare glass solutions of poorly<br />

water-soluble drugs with hydrophilic excipients. It is claimed that the<br />

method is an improvement to existing formulation methods such as spray-<br />

drying and co-melting because it uses smaller quantities of drug reduces<br />

particle size and speeds up the formulation process (Breitenbach 2002).<br />

3.7.4.1.3.<br />

Hot spin melting<br />

method:<br />

A further alternative for processing thermolabile substances is by hot<br />

spin melting. Here, the drug and carrier are melted together over an<br />

extremely short time in a high-speed mixer and, in the same apparatus,<br />

dispersed in air or an inert gas in a cooling tower. Some drugs have been<br />

processed into solid dispersions using hot-spin-meIting include<br />

progesterone (Fricke et al., 1995) and dienogest (Kaufmann et al.,<br />

1995).<br />

3.7.4.2.<br />

Solvent method:<br />

Prepared by dissolving a physical mixture of two solid components<br />

in a common solvent, followed by evaporation of the solvent. The choice<br />

of solvent and its removal rate are critical to the quality of the dispersion.<br />

- 75 -


The main advantage of the solvent method is that thermal decomposition<br />

of drugs or carriers may be prevented because of the low temperature<br />

required for the evaporation of organic solvents. However , some<br />

disadvantages associated with this method are the high cost of preparation,<br />

the difficulty in completely removing liquid solvent , the possible adverse<br />

effect of its supposedly negligible amount of the solvent on the chemical<br />

stability of the drug (Wrenn and Simeon , 1998).<br />

Mallick et al., 2003, prepared albendazole solid dispersions by<br />

solvent evaporation technique using water soluble carriers such as<br />

polyethylene glycol and polyvinyl pyrrolidone.<br />

3.7.4.3.<br />

Melting-solvent<br />

method:<br />

Prepared by first dissolving a drug in a suitable liquid solvent and<br />

then incorporating the solution directly into a melt of carrier. The fluid is<br />

then cooled to room temperature. Such a unique method possesses the<br />

advantages of both the melting and solvent methods (Craig 1990).<br />

3.7.4.4. Other methods:<br />

Other methods for preparation of solid dispersions including co-<br />

grinding ( Babu et al ., 2002 ) , kneading ( Singh and Udupa 1997 ) ,<br />

spray drying ( Palmieri et al ., 2002 ) , freeze-drying (Emara et al .,<br />

2002 ) and supercritical fluid technique e.g.supercritical C02 ( Moneghini<br />

et al ., 2001).<br />

Sethia and Squillante 2004, compared the physicochemical and<br />

dissolution properties of carbamazepine solid dispersions prepared by<br />

either a conventional solvent evaporation versus supercritical fluid<br />

process. They found that, the supercritical based process produced solid<br />

dispersions with intrinsic dissolution rate better than conventional solid<br />

dispersions.<br />

- 76 -


3.7.5. Proposed Structures of solid dispersions:<br />

The physicochemical structures of solid dispersions play an<br />

important role in controlling their drug release. Four representative<br />

structures have been outlined as representative of interactions between<br />

carrier and drug.<br />

3.7.5.1.<br />

Simple eutectic mixtures:<br />

No review of solid dispersions would be complete without a brief<br />

description of eutectic mixtures, which are the cornerstone of this<br />

approach to improving bioavailability of poorly soluble compounds. A<br />

simple eutectic mixture consists of two compounds that are completely<br />

miscible in the liquid state but only to a very limited extent in the solid<br />

state. Solid eutectic mixtures are usually prepared by rapid cooling of a<br />

co-melt of the two compounds in order to obtain a physical mixture of<br />

very fine crystals of the two components. As shown in (Figure 5), when a<br />

mixture with composition E, consisting of a slightly soluble drug and an<br />

inert, highly water soluble carrier, is dissolved in an aqueous medium, the<br />

carrier will dissolve rapidly, releasing very fine crystals of the drug<br />

(Sekiguchi and Obi, 1961; Goldberg et al., 1966). The large surface<br />

area of the resulting suspension should result in an enhanced dissolution<br />

rate and thereby improved bioavailability.<br />

- 77 -


Figure 5: Phase diagram for eutectic system<br />

(reproduced from Castellan, 1983).<br />

3.7.5.2.<br />

Solid solutions:<br />

Solid solutions are comparable to liquid solutions, consisting of just<br />

one phase irrespective of the number of components. Solid solutions of a<br />

poorly water-soluble drug dissolved in a carrier with relatively good<br />

aqueous solubility are of particular interest as a means of improving oral<br />

bioavailability (Schachter et al., 2004). In the case of solid solutions, the<br />

drug’s particle size has been reduced to its absolute minimum viz. the<br />

molecular dimensions (Goldberg et al., 1965). Furthermore, the<br />

dissolution rate is determined by the dissolution rate of the carrier. By<br />

judicious selection of a carrier, the dissolution rate of the drug can he<br />

increased by up to several orders of magnitude. Solid solutions can be<br />

classified according to two methods. First, they can be classified<br />

according to their miscibility (continuous versus discontinuous solid<br />

solutions) or second, according to the way in which the solvate molecules<br />

are distributed in the Solvendum (substitutional, interstitial or<br />

amorphous).<br />

- 78 -


3.7.5.2.1. Continuous solid solutions<br />

The components are miscible in all proportions. Theoretically, this<br />

means that the bonding strength between the two components is stronger<br />

than the bonding strength between the molecules of each of the individual<br />

components. Leuner and Dressman (2000) stated that solid solutions of<br />

this type have not been reported in most literatures.<br />

3.7.5.2.2.<br />

Discontinuous solid solutions:<br />

The solubility of each of the components in the other is limited. A<br />

typical phase diagram is shown in (Figure 6), <br />

regions of true solid solutions. In these regions, one of the solid<br />

components is completely dissolved in the other solid component. Note<br />

that below a certain temperature, the mutual solubilities of the two<br />

components start to decrease.<br />

Figure 6: Phase diagram for Discontinuous solid solutions<br />

(reproduced from Castellan, 1983).<br />

3.7.5.2.3.<br />

Substitutional crystalline solid solutions:<br />

Classical solid solutions have a crystalline structure, in which the<br />

solute molecules can either substitute for solvent molecules in the crystal<br />

- 79 -


lattice or fit into the interstices between the solvent molecules. A<br />

substitutional crystalline solid dispersion is depicted in (Figure 7).<br />

Substitution is only possible when the size of the solute molecules differs<br />

by less than 15% or so from that of the solvent molecules (Leuner and<br />

Dressman, 2000).<br />

Figure 7: Substitutional crystalline solid solutions.<br />

(reproduced from Chiou and Riegelman, 1971)<br />

3.7.5.2.4.<br />

Interstitial crystalline solid solutions:<br />

In interstitial solid solutions, the dissolved molecules occupy the<br />

interstitial spaces between the solvent molecules in the crystal lattice<br />

(Figure 8) The relative molecular size is a crucial criterion for classifying<br />

the solid solution type. In the case of interstitial crystalline solid solutions,<br />

the solute molecules should have a molecular diameter that is no greater<br />

than 0.59 of the solvent molecule’s molecular diameter (Leuner and<br />

Dressman, 2000). Furthermore, the volume of the solute molecules<br />

should be less than 20% of the solvent.<br />

Figure 8: Interstitial crystalline solid solutions.<br />

- 80 -


(reproduced from Chiou and Riegelman, 1971)<br />

3.7.5.2.5. Amorphous crystalline solid solutions:<br />

In an amorphous solid solution, the solute molecules are dispersed<br />

molecularly but irregularly within the amorphous solvent (Figure<br />

9) .Using griseofulvin in citric acid, Chiou and Riegelman (1969) were<br />

the First to report the formation of an amorphous solid solution to<br />

improve a drug’s dissolution properties. Polymer carriers are particularly<br />

likely to form amorphous solid solutions, as the polymer itself is often<br />

present in the form of an amorphous polymer chain network. In addition,<br />

the solute molecules may serve to plasticize the polymer, leading to a<br />

reduction in its glass transition temperature.<br />

Figure 9: Amorphous crystalline solid solution<br />

(reproduced from Kreuter, 1999)<br />

3.7.5.3.<br />

Glass solution and glass suspensions:<br />

A glass solution is a homogenous glassy system in which a solute<br />

dissolves in a glassy solvents e.g. sugars, citric acid. It is often<br />

characterized by transparency and brittleness below the glass transition<br />

temperature. The lattice energy in glass solution is less than in solid<br />

solutions because of its similarity with liquid solutions. Consequently,<br />

faster dissolution rates of drugs from their glass solutions are expected<br />

compared to those from solid solutions (Mummaneni and Vasavada,<br />

1990).<br />

- 81 -


3.7.5.4. Combination of systems:<br />

The possibility exists that some or indeed all systems may show<br />

characteristics of more than one of the above structures. For example, the<br />

formation of eutectic mixtures must involve solid-state complexation to<br />

some extent (Juppo et al., 2003).<br />

3.7.6.<br />

Carriers for solid dispersions:<br />

The carrier used for solid dispersion formulation has been a water-<br />

soluble or water miscible polymer such as polyethylene glycol (PEG) or<br />

polyvinylpyrrolidone (PVP) or low molecular weight materials such as<br />

sugars. However, the proliferation of publications in the area since the<br />

first solid dispersions were described (Sekiguchi and Obi, 1961) has led<br />

to a broadening of these definitions to include water insoluble matrices<br />

such as Gelucires and Eudragits that may yield either slow or rapid<br />

release. Consequently, the properties of the carrier have a great influence<br />

on the dissolution characteristics of the dispersed drug. A carrier, as<br />

suggested by Kerc et al. (1998), should be: i) freely water soluble with<br />

intrinsic rapid dissolution properties; ii) non-toxic and pharmacologically<br />

inert; iii) chemically compatible with the drug and in the solid-state<br />

should not form strongly bonded complexes that could reduce the<br />

dissolution rates; iv) preferably, can increase the aqueous solubility of the<br />

drug; v) soluble in a variety of organic solvents (for carriers intended for<br />

solvent processes); and vi) chemically, physically and thermally stable<br />

with a low melting point to avoid the use of excessive heat during<br />

dispersion preparation(for carriers intended for fusion processes). With<br />

references to these criteria there now follows brief review of the carriers<br />

described in the literature with particular emphasis on their potentials and<br />

limitations.<br />

- 82 -


3.7.6.1.<br />

Polyethylene glycol:<br />

Polyethylene glycols are polymers of ethylene oxide, with a<br />

molecular weight usually falling in the range 200: 300000. For the<br />

manufacture of solid dispersions and solutions, PEGs with molecular<br />

weights of 1500:35000 are usually employed. As the molecular weight<br />

increases, so does the viscosity of the PEG. At molecular weight of up to<br />

600, PEGs are fluid, in the range 800: 1500 they have a consistency that<br />

is best described as vaseline like, from 2000 to 6000 they are waxy and<br />

those of 20 000 and above form hard, brittle crystals at room temperature.<br />

Their solubility in water is generally good. Furthermore a particular<br />

advantage of PEGs for the formation of solid dispersions is that they also<br />

have good solubility in many organic solvents. The melting point of the<br />

PEGs of interest lies under 65°C in every case (e.g. the m.p. of PEG 1000<br />

is 30: 40°C, the m.p. of PEG 4000 is 50: 58°C and the m.p. of PEG 20000<br />

is 60:63°C) (Price, 1994). These relatively low melting points are<br />

advantageous for the manufacture of solid dispersions by the melting<br />

method. Additional attractive features of the PEGs include their ability to<br />

solubilize some compounds (Fini et al., 2005) as well as to improve<br />

compound wettability (Ambike et al., 2004). Even the dissolution rate of<br />

a relatively soluble drug like aspirin can be improved by formulating it as<br />

a solid dispersion in PEG 6000 (Corrigan et al., 1979).<br />

3.7.6.2.<br />

Polyvinylpyrrolidone:<br />

Polymerization of vinylpyrrolidone leads to polyvinylpyrrolidone<br />

(PVP) of molecular weights ranging from 2500 to 3000000 (Walking,<br />

1994).<br />

Similarly to the PEGs, PVPs have good water solubility and can<br />

improve the wettability of the dispersed compound in many cases<br />

(Mendyk and Jachowicz, 2005). Improved wetting and thereby an<br />

- 83 -


improved dissolution rate from a solid dispersion in PVP has been<br />

demonstrated for tolbutamide. The chain length of the PVP has a very<br />

significant influence on the dissolution rate of the dispersed drug from the<br />

solid dispersion. The aqueous solubility of the PVPs becomes poorer with<br />

increasing chain length and a further disadvantage of the high MW PVPs<br />

is their much higher viscosity at a given concentration (Takeuchi et al.,<br />

2004).<br />

3.7.6.3.<br />

Urea:<br />

Urea is the final product of human protein metabolism. Its solubility<br />

in water is greater than 1 in 1 and it exhibits good solubility in many<br />

common organic solvents. It has a relatively low melting point of 131 °C.<br />

Consequently, both solvent and fusion processes could be used to prepare<br />

urea dispersions. In one of the first bioavailability studies of solid<br />

dispersions, it was shown that sulphathiazole was better absorbed in<br />

rabbits when given as eutectic mixture with urea (Sekiguchi and Obi,<br />

1961). Although urea is not often used as a carrier these days, it has been<br />

shown that the dissolution rate of the poorly soluble compound ofloxacin<br />

can be improved by more than three fold by incorporating it in a<br />

coevaporate with urea (Okonogi et al., 1997). Similarly, urea was used in<br />

combination with PEG to increase the dissolution rate of piroxicam (Pan<br />

et al., 2000).<br />

3.7.6.4.<br />

Sugars:<br />

Although sugars and related compounds are highly water-soluble<br />

and have few, if any, toxicity issues, they are less suitable than other<br />

carriers for the manufacture of solid dispersions. The melting point of<br />

most sugars is high, making preparation by the hot melt method<br />

- 84 -


problematic, and their solubility in most organic solvents is poor, making<br />

it difficult to prepare co-evaporates.<br />

3.7.6.5.<br />

Chitosan:<br />

Chitosan, a derivative of the polysaccharide chitin that is formed by<br />

deacetylation at the N position, has also been used as a carrier in solid<br />

dispersions. It exhibits good biocompatibility and safety after oral and<br />

parenteral administration. Low molecular weight chitosan is a good<br />

candidate as a carrier for enhancing the dissolution and bioavailability of<br />

a number of poorly water soluble drugs (Asada et al., 2004; Takahashi<br />

et al., 2005).<br />

3.7.6.6.<br />

Emulsifiers:<br />

The release behaviour of many drugs can also be improved by the<br />

use of emulsifying agents. Two mechanisms are possible here:<br />

improvement of wetting characteristics and solubilisation of the drug.<br />

Owing to their potential toxicity problems, such as damage to mucosal<br />

surfaces, they are usually used in combination with another carrier. For<br />

example, the release of naproxen from solid dispersions in PEG 4000,<br />

6000 and 20000 could be further enhanced when either sodium lauryl<br />

sulphate or Tween® 80 was added to the system (Mura et al., 1999).<br />

Inclusion of alkali dodecylsulphate surfactants in carrier systems can lead<br />

to conversion of a solid dispersion to a solid solution. Melts of<br />

griseofulvin and PEG 6000 normally contain crystalline areas but in the<br />

presence of sodium lauryl sulphate, a solid solution is formed (Wulff et<br />

al., 1996).<br />

3.7.6.7.<br />

Other carriers:<br />

- 85 -


Many other substances have been tested as carriers for solid<br />

dispersions. A hydrolysis product of collagen, Gelita® Collagel, was<br />

reported to improve the release rate of oxazepam by a factor of six when<br />

prepared as a solid dispersion by spray drying (Jachowicz et al., 1993).<br />

Even after tabletting, the solid dispersion displayed better release<br />

characteristics than the physical mixture or the drug powder alone<br />

(Jachowicz and Nurnberg, I 997). Other materials tested include<br />

phospholipids (Sammour et al., 2001), inulin (Visser et al., 2004), silica<br />

(Watanabe et al., 2003) ......etc.<br />

3.7.7. Characterization of solid dispersions<br />

The methods that have been used to characterize solid dispersions<br />

are summarized in (Table1). In addition to characterizing the solid<br />

dispersion, these methods can be used to differentiate between solid<br />

solutions (molecularly dispersed drug), solid dispersions and physical<br />

mixtures of drug and carrier. It is usually assumed that dispersions in<br />

which no crystallinity can be detected are molecularly dispersed. The<br />

absence of crystallinity is used as a criterion to differentiate between solid<br />

solutions and solid dispersions (Leuner and Dressman, 2000).<br />

Table 1: Methods for the characterization of solid dispersion.<br />

1- Dissolution testing<br />

2- Thermoanalytical methods: * Differential thermoanalysis (DTA).<br />

* Differential scanning caloimetry (DSC).<br />

* Hot stage microscopy.<br />

3- X-Ray diffraction (XRD).<br />

4- Spectroscopic methods, e.g. FTIR spectroscopy.<br />

5- Microscopic methods: * Polarization microscopy.<br />

* Scanning electron microscopy (SEM).<br />

- 86 -


3.7.7.1. Dissolution testing:<br />

When the goal of preparing a solid dispersion is to improve the<br />

dissolution characteristics of the drug in question, the results of the<br />

release rate experiments are obviously of prime importance in assessing<br />

the success of the approach. A well-designed release experiment will<br />

show whether the solubility of the drug and its dissolution rate has been<br />

enhanced, as well as whether the resulting supersaturated solution is<br />

stable or tends to precipitate quickly. Comparison of results with those for<br />

pure drug powder and physical mixtures of the drug and carrier can help<br />

to indicate the mechanism by which the carrier improves dissolution<br />

(Leuner and Dressman, 2000).<br />

3.7.7.2.<br />

Thermoanalytical methods:<br />

It includes all methods that examine a characteristic of the system as<br />

a function of temperature. Differential scanning calorimetry (DSC) is the<br />

most highly regarded method. DSC enables the quantitative detection of<br />

all processes in which energy is required or produced (i.e. endothermic<br />

and exothermic phase transformations). The usual method of<br />

measurement is to heat the reference and test samples in such a way that<br />

the temperature of the two is kept identical. The additional heat required<br />

is recorded and used to quantitate the energy of the phase transition.<br />

Exothermic transitions, such as conversion of one polymorph to a more<br />

stable polymorph, can also be detected. Lack of a melting peak in the<br />

DSC of a solid dispersion indicates that the drug is present in an<br />

amorphous rather than a crystalline form. Since the method is quantitative<br />

in nature, the degree of crystallinity can also be calculated for systems in<br />

which the drug is partly amorphous and partly crystalline. However,<br />

crystallinities of fewer than 2% cannot generally be detected with DSC<br />

(Kreuter, 1999).<br />

- 87 -


3.7.7.3.<br />

X-ray<br />

diffraction ( XRD):<br />

The principle behind X-ray diffraction is that when an X-ray beam is<br />

applied to the sample, interference bands can he detected. The angle at<br />

which the interference bands can be detected depends on the wavelength<br />

applied and the geometry of the sample with respect to periodicities in the<br />

structure. Crystallinity in the sample is reflected by a characteristic<br />

fingerprint region in the diffraction pattern. Owing to the specificity of<br />

the fingerprint, crystallinity in the drug can be separately identified from<br />

crystallinity in the carrier. Therefore, it is possible with X-ray diffraction<br />

to differentiate between solid solutions, in which the drug is amorphous,<br />

and solid dispersions, in which it is at least partly present in the<br />

crystalline form, regardless of whether the carrier is amorphous or<br />

crystalline. However, crystallinities of less than 5- 10% cannot generally<br />

be detected with X- ray diffraction (Villiers et al., 1998).<br />

3.7.7.4.<br />

Infrared spectroscopy:<br />

Structural changes and lack of a crystal structure can lead to changes<br />

in bonding between functional groups that can be detected by infrared<br />

spectroscopy. Since not all peaks in the IR spectrum are sensitive to<br />

crystalline changes, it is possible to differentiate between those that are<br />

sensitive to changes in crystallinity and those that are not (Taylor and<br />

Zografi, 1997).<br />

- 88 -


1- Materials and supplies:<br />

Experiment and methodology<br />

* Gliclazide was kindly supplied by Egyptian International<br />

Pharmaceutical<br />

Industries Company (EIPICO)<br />

* Chloroform, glucose, methanol and urea (El-Gomhouria Co.,<br />

Egypt).<br />

* Polyethylene glycol 4000, 6000 (Hoechest Chemikalien, Werk Gendort,<br />

Germany).<br />

2- Equipment:<br />

* UV/VIS spectrophotometer (Schimadzu U.V.-1201, Cat NO.<br />

206-62409, Schimadzu Corporation, Japan).<br />

* Thermostatic shaker water bath (Julpo SW 20C, Japan).<br />

* Vacuum oven (Lab-line instruments, Inc., USA).<br />

* Dissolution tester, rotating paddle (Erweka RT6- Frankfurt,<br />

Germany).<br />

* Perkin-Elmer FTIR spectrophotometer (1600 series, Perkin-Elmer<br />

Corporation, Norwalk, USA).<br />

* Differential scanning calorimeter (model 50, Schimadzu,<br />

Japan).<br />

* D-5000 x- ray diffractometer (Kristallofex D-5000 Powder<br />

Diffractometer, Siemens, Germany).<br />

Set of sieves (Mettler, Germany).<br />

Electronic Digital Balance (Mettrt-Toledo, Ag,CH 8606,<br />

Greifensee,Switzerland).<br />

* Buchi rotavapor R-3000, (Switzerland).<br />

- 89 -


3- Software:<br />

Microsoft Office XP, Microsoft Corporation, USA.<br />

4. Methods:<br />

4.1. UV scanning of Glz:<br />

Ten mg of Glz were dissolved in 100 ml methanol to obtain a<br />

solution; 1 ml is diluted to 10 ml with methanol to produce a solution<br />

containing 10 μg /ml of Glz in methanol. The obtained solution was<br />

scanned spectrophotometerically from 200 to 400 nm using methanol as<br />

blank.<br />

4.2. Construction of calibration curve of Glz in methanol:<br />

0.1 gram of Glz was dissolved in 100 ml methanol to obtain a<br />

solution, 2.5 ml is diluted to 25 ml with methanol to produce a solution<br />

containing 100 μg /ml of Glz. Aliquots of 1, 1.5, 2, 2.5, and 3 ml were<br />

further diluted to 10 ml with methanol. After dilution, the solution<br />

contained 10, 15, 20, 25, and 30 μg/ml of Glz respectively.<br />

The calibration equation was constructed by regressing the relative<br />

absorbances against the corresponding Glz solutionsconcentrations at<br />

227 nm using methanol as blank.<br />

4.3. Construction of calibration curve of Glz in S<br />

buffer of pH 7.4:<br />

0.1 gram of Glz was dissolved in 5 ml methanol, then completed to<br />

100 ml with sörensen’ buffer. 2.5 ml is diluted to 25 ml with methanol to<br />

produce a solution containing 100 μg /ml of Glz . Aliquots of 0.5, 0.75, 1,<br />

1.5, 2, and 2.5 ml were furtherly diluted to 10 ml with sörensen’ buffer.<br />

After dilution, the solution contained 5, 7.5, 10, 12.5, 15, 20, and 25<br />

μg/ml of Glz respectively. The calibration equation was constructed by<br />

- 90 -


egressing the relative absorbances against the corresponding Glz<br />

solutionsconcentrations at 227 nm using sörensen’phosphate buffer as<br />

blank.<br />

4.4. Preparation of solid dispersions:<br />

Gines’ et al., 1996, stated that, the technology employed to prepare<br />

the solid dispersion, the proportion and properties of the carrier used<br />

present an important influence on the properties of the resulting (SD). So,<br />

in this study different types and proportions of carriers were examined<br />

(Table 2).<br />

Table 2: Types of carriers and their ratios in Glz solid dispersions<br />

and physical mixtures.<br />

Carrier Drug: Carrier weight ratio Solvent used <br />

PEG 6000 10:90 8:92 5:95 1:99 Chloroform<br />

PEG 4000 10:90 8:92 5:95 1:99 Chloroform<br />

Glucose 1:1 1:2 1:3 1:5 1:10 Methanol<br />

Urea 1:1 1:2 1:3 1:5 1:10 Methanol<br />

Types of solvent used in solvent evaporation method.<br />

4.4.1. Preparation of urea solid dispersions:<br />

The calculated amounts of Glz with UR were dissolved in methanol<br />

with continuous stirring in a dish followed by evaporation of the solution<br />

under vacuum at 40°C. Dispersions were dried in a vacuum oven at room<br />

temperature for 24 hr. The dry products were removed from the<br />

containers and ground in laboratory mortar (Etman, 2000).<br />

- 91 -


4.4.2. Preparation of glucose solid dispersions:<br />

The co-precipitates were prepared using solvent evaporation<br />

method. The calculated amounts of Glz and glucose were dispersed<br />

homogeneously in the least amount of methanol at 40°C, and the solvent<br />

was evaporated at 40°C under vacuum. The obtained co-precipitates were<br />

dried in a vacuum oven at room temperature for 24 hr, and then the dried<br />

mass was pulverized (Greenhalgh et al., 1999)<br />

4.4.3. Preparation of PEG 4000 and PEG 6000 solid dispersions:<br />

The required amounts of Glz and PEG 4000 or PEG 6000 were<br />

accurately weighted and dissolved in chloroform. Mixtures were<br />

evaporated using a rotary evaporator at 45°C and further drying was<br />

performed using a vacuum dessicator for 48 hours at room temperature<br />

.Subsequently, the dispersions were pulverized in a mortar (Law et al.,<br />

1992).<br />

4.5. Preparation of physical mixtures:<br />

(PMs) were prepared simply by triturating appropriate quantities of<br />

Glz and carriers using a porcelain mortar and a pestle, then transferring to<br />

a vacuum dessicator until ready for use.<br />

*** All samples were sieved. Powdered samples below 420 um (40<br />

mesh) were stored in closed containers away from the light and humidity<br />

until use.<br />

4.6. Solubility measurements:<br />

4.6.1. Effect of different carriers on the solubility of Glz:<br />

- 92 -


Solubility studies were carried out according to the method of<br />

Higuchi and Connors, (1965). An excess of the Glz (10 mg) was placed<br />

into 25-ml glass vial containing various concentrations of each carrier,<br />

ranging from 1 to 7 %, in 10 ml distilled water. All glass vials were<br />

closed with stopper and cover-sealed with cellophane membrane to avoid<br />

solvent loss .The content of the suspension was equilibrated by shaking in<br />

a thermostatically controlled water bath at 25°C for 72 hr.<br />

After attainment of equilibrium, the content of each vial was then<br />

filtered through a double filter paper (Whatman 42). The filtrate was<br />

suitably diluted and assayed spectrophotometrically at 227 nm to measure<br />

the amount of dissolved drug. There was no interference from all the used<br />

carriers at this wavelength except urea interfered with analysis of Glz,<br />

thus the solutions containing urea were measured against a blank of urea.<br />

The average of triplicate measurements was reported. The solubility of<br />

Glz alone in water at the same temperature was also determined following<br />

the same procedure mentioned above.<br />

4.6.2. Effect of pH change on the solubility of Glz:<br />

The solubility of Glz in Sorensen' phosphate buffer with different<br />

pH ranging from 5 to 7.4 at the same temperature was also determined<br />

following the same procedure mentioned above.<br />

4.7. Dissolution studies:<br />

The dissolution of Glz from the prepared (SDs), and (PMs) was<br />

carried out according to the USP-25, NF 20 (2002), rotating paddle<br />

method. Dissolution medium consisting of 500 ml distilled water. The<br />

stirring rate was 100 rpm and the temperature was maintained at 37 ±<br />

0.5°C. A sample of 40 mg of Glz or its equivalent of the (SDs), or the<br />

- 93 -


(PMs) was placed on the surface of the dissolution medium. At a<br />

appropriate time intervals (5, 10, 20, 30, 45, 60, 90, and 120 min), 5 ml<br />

sample were withdrawn and replaced with an equivalent amount of the<br />

fresh dissolution medium kept at 37°C. The samples were filtered rapidly<br />

through a double layered filter paper (Whatman 42). The filtrates were<br />

suitably diluted and assayed spectrophotometrically at 227 nm without<br />

interference from the carriers. In case of (SDs) containing UR, the<br />

solutions measured against a blank of UR.<br />

The amount of Glz dissolved at different time intervals was calculated<br />

using a standard calibration curve .Each experiment was carried out in<br />

triplicates. The cumulative amount of the drug released was calculated as<br />

follows (AL-Suwayeh, 2003):<br />

Receptor compartment volume = VR<br />

Sample volume withdrawn =5 ml<br />

Sample #1(5min),# 2 (10 min), # 3 (20 min), # 4 (30 min), # 5 (45 min), #<br />

6 (60 min), # 7 (90min), # 8 (120min).<br />

Concentration C1 (5 min), C2 (10 min), C3 (20 min), C4 (30 min), C5<br />

(45 min), C6 (60 min), C7 (90 min), C8 (120 min).<br />

Cumulative amount at sample # 1 (5 min) = VR X C1<br />

Cumulative amount at sample # 2 (10 min) = VR X C2 +5 ml X (C1)<br />

Cumulative amount at sample # 3 (20 min) = VR X C3 + 5 ml X (C1+<br />

C2)<br />

Cumulative amount at sample # 4 (30min) = VR X C4 + 5 ml X<br />

(C1+C2+C3). And so on.<br />

- 94 -


4.8. Fourier transform infrared (FTIR) spectroscopy:<br />

FTIR spectra were obtained on a Prekin-Elmer 1600 FTIR<br />

spectrophotometer using KBr disc method. The scanning range was 400-<br />

4000 cm -1 .<br />

4.9. Differnntial scanning calorimetry (DSC):<br />

The DSC thermograms were recorded on a Schimadzu-DSC 50.<br />

Samples (1.3 mg) were heated in hermetically sealed aluminum pans over<br />

the temperature range 50-200°C at a constant rate of 10°C/min under a<br />

nitrogen purge 30 ml/min.<br />

4.10. X-ray diffraction:<br />

X-ray diffraction patterns were obtained using a Siemens<br />

Kristallofex D- <br />

<br />

of 0-80°.<br />

- 95 -


1. UV scanning of Glz:<br />

Results and discussion<br />

UV scanning of Glz in methanol was carried out (Figure 11). Two<br />

absorption maxima were observed at 227 nm and 273 nm The ratio of the<br />

maxmax 273 nm is<br />

0.825 to 0.034. So, measurements were done at 227 nm (Tadeusz et al.,<br />

2005).<br />

Wavelength<br />

Figure 11: UV spectra of Glz in methanol.<br />

2. Calibration curves of Glz in methanol and sörensen’s phosphate<br />

buffer pH 7.4:<br />

(Figures 12, 13) show a linear relationship between the absorbance<br />

and the concentration of Glz in either methanol or in sörensen’s<br />

max in the<br />

concentration range used.<br />

- 96 -


Absorbance<br />

1.2<br />

1<br />

0.8<br />

0.6<br />

0.4<br />

0.2<br />

0<br />

y = 0.0329x + 0.0233<br />

R 2 = 0.9973<br />

R= 0.9986<br />

0 5 10 15 20 25 30 35<br />

Concentration (ug/ml)<br />

Figure 12: Calibration curve of Glz in methanol at max 227 nm.<br />

Absorbance<br />

1<br />

0.9<br />

0.8<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

y = 0.0361x + 0.0296<br />

R 2 = 0.9887<br />

R= 0.9943<br />

0 5 10 15<br />

concentration (ug/ml)<br />

20 25 30<br />

Figure 13 : Calibration curve of Glz in phosphate buffer (7.4) at max<br />

227 nm.<br />

- 97 -


3. Solubility measurements:<br />

3.1. Effect of different carriers on the solubility of Glz :<br />

In the present study, the solubility of Glz in distilled water at 25°C<br />

was found to be 42.13 μg /ml.<br />

(Figure 14, 15) depict the effect of different carriers on Glz<br />

solubility in distilled water at 25°C. In case of PEG 4000, 6000, the<br />

solubility of Glz linearly increased as the carrier concentration increased,<br />

showing the feature of an AL-type solubility phase diagram (Higuchi and<br />

Corner, 1965) .This result illustrates that the complex formed was<br />

soluble and did not form a precipitate over the range of carrier<br />

concentration.<br />

As shown in (Table 3), the solubilizing power of PEGS slightly decreased<br />

with increasing PEG molecular weight. As the solubility factors were 1.9<br />

and 1.6 for PEG 4000 and PEG 6000, respectively. This is in accordance<br />

with Mura et al.,1999, who found that, the solubility of naproxen is<br />

affected by PEG molecular weight as the solubilizing power of PEG 4000<br />

> PEG 6000 > PEG 20,000.<br />

On other hand the solubility plot of glucose, and urea showed a Bs-type<br />

curve (Higuchi and Corner, 1965). The initial rising portion was<br />

followed by plateau region and finally a decrease in total concentration of<br />

Glz. As shown in (Table 3), the solubility factor were 1.28 and 1.4 for<br />

glucose and urea, respectively. Consequently, these carriers can be ranked<br />

according to its effect on increasing the solubility of Glz as PEG 4000 <br />

PEG 6000 glucose urea. The increased solubility of Glz in carrier's<br />

solution may be attributed to both complex formation and reduction in<br />

interfacial tension of water and hence intermolecular forces and polarity<br />

caused by the presence of these carriers (Al-Angary et al., 1996).<br />

- 98 -


Table 3: Solubility enhancement data of Glz in various carrier<br />

solutions at 25°C.<br />

Item Glz PEG-<br />

Phase solubility<br />

diagram type<br />

Optimum<br />

carrier<br />

concentration<br />

%(w/v)<br />

Solubility<br />

(ug/ml)<br />

Solubility factor<br />

a<br />

4000<br />

- 99 -<br />

PEG-<br />

6000<br />

Glu UR<br />

----- AL AL BS BS<br />

------ 7% 7% 3% 5%<br />

42.13 80.23 67.63 54.3 48.19<br />

------- 1.9 1.6 1.28 1.14<br />

Solubility factor a = Total solubility / intrinsic solubility.


Solubility (ug/ml)<br />

Solubility (ug/ml)<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

0 1 2 3 4<br />

Carrier % (w/v)<br />

5 6 7 8<br />

- 100 -<br />

PEG 4000<br />

PEG 6000<br />

Figure 14: Phase solubility diagram of Glz in water<br />

at 25°C in presence of PEG 4000 and PEG 6000.<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

0<br />

Urea Glucose<br />

0 1 2 3 4 5 6 7 8<br />

Carrier % (w/v)<br />

Figure 15: Phase solubility diagram of Glz in water<br />

at 25°C in presence of glucose and urea.


3.2. Effect of pH change on the solubility of Glz:<br />

Table 4 demonstrate the solubility of Glz in different pH's. Glz<br />

contains an -hydroxyl secondary amine, with a pKa of 7.8. It exhibits<br />

pH dependent solubility. It can be noted that the solubility increased with<br />

increasing pH (higher in alkaline rather than acidic one). This can be<br />

attributed to the effect of pH on the degree of the ionization and hence the<br />

solubility of the drug.<br />

Table 4: Effect of change in pH on the solubility of Glz.<br />

4. Dissolution studies:<br />

pH Solubility (g/ml)<br />

5 40.11 ± 0.26<br />

5.6 52.34 ± 0.22<br />

6 156.77 ± 2.07<br />

6.4 227.4 ± 5.02<br />

7.4 745.5 ± 13.45<br />

The dissolution profiles of pure Glz, its physical mixtures and solid<br />

dispersions with different carriers are shown in (Figures 16-19) Data are<br />

average of three measurements. The shapes of the dissolution profiles<br />

were examined using the following parameters:<br />

I) The initial dissolution rate (IDR) calculated as percent dissolved of the<br />

- 101 -


drug over the first twenty minutes per minutes.<br />

II) The percentage of the drug dissolved after 20 and 60 minutes (PD20<br />

and<br />

PD60).<br />

III) The dissolution efficiency (DE %) parameter after sixty minutes<br />

(Arias<br />

et al., 1995).<br />

The dissolution efficiency can be defined as the area under the curve<br />

up to a certain time. It is measured using the trapezoidal method and is<br />

expressed as a percentage of the area of the rectangle described by 100%<br />

dissolution in the same time (Torrado et al., 1996).<br />

The calculated dissolution parameters revealed that, pure Glz<br />

yielded the slowest dissolution rate with only about 28.4 % of the drug is<br />

dissolved in 120 min. The hydrophobic property of Glz prevented its<br />

contact with the dissolution medium which led to a slow dissolution rate<br />

(Tantishaiyakul et., al 1996).<br />

As shown in Tables 5-8, all (PMs) released the Glz at the faster rate than<br />

the drug alone as reflected by higher (IDR) and greater extent of<br />

dissolution after 120 min. These results can be explained on the basis that<br />

the dry mixing brings the drug in close contact with the hydrophilic<br />

polymer (Van den Mooter et al., 1998). Also may be due to; a possible<br />

solubilization effect by the carrier operating the microenvironment<br />

(diffusion layer) immediately surrounds the drug particle in the early<br />

stage of solubilization (Arias et al., 1996). Indeed, during dissolution<br />

experiments, it was noticed that (PMs) immediately sink to the bottom of<br />

the dissolution vessels as (SDs) do.<br />

- 102 -


Table 5: Dissolution parameters (±SD) of gliclazide in distilled water from different gliclazide - PEG 6000<br />

systems.<br />

DE*100<br />

(%)<br />

PD60<br />

(%)<br />

PD20<br />

(%)<br />

Composition (w/w) IDR<br />

(%dissolved/min)<br />

Gliclazide powder 0 ± 0 0 ± 0 13.05 ± 1.2 3.83 ± 0.06<br />

Gliclazide-to-PEG 6000<br />

PM 10:90<br />

1.12 ± 0.12 22.58 ± 0.4 38.08 ± 0.19 24.44 ± 0.64<br />

SD 10:90<br />

3.27 ± 0.07 65.53 ± 1.4 72.89 ± 1.5<br />

63.54 ± 1.1<br />

PM 8:92<br />

1.12 ± 0.09 22.59 ± 1.9 36.89 ± 0.18 25.30 ± 0.96<br />

SD 8:92<br />

3.2 ± 0.04 64.04 ± 0.87 69.29 ± 1.76 60.83 ± 0.91<br />

PM 5:95<br />

1.08 ± 0.08 21.63 ± 1.6 38.1 ± 1.2 24.82 ± 1.1<br />

SD 5:95<br />

3.66 ± 0.04 73.25 ± 0.9 93.23 ± 0.42 76.45 ± 0.30<br />

34.76 ± 0.65<br />

89.78 ± 3.48<br />

41.19 ± 0.49<br />

100.49 ± 1.00<br />

35.79 ± 0.48<br />

92.36 ± 1.5<br />

1.79 ± 0.02<br />

4.6 ± 0.07<br />

PM 1:99<br />

SD 1:99<br />

IDR = Initial dissolution rate.<br />

PD20 = Extent of dissolution after 20 min.<br />

PD60 = Extent of dissolution after 60 min.<br />

DE% = Dissolution efficiency after 60 min.<br />

103


plain drug 10:90 PM 10:90 SD 8:92 PM 8:92 SD 5:95 PM<br />

5:95 SD 1:99 PM 1:99 SD<br />

120<br />

100<br />

80<br />

60<br />

40<br />

% Drug dissolved<br />

20<br />

0<br />

0 20 40 60 80 100 120<br />

Time (min)<br />

Figure 16: Dissolution profile of gliclazide-PEG 6000 systems.<br />

104


Table 6: Dissolution parameters (±SD) of gliclazide in distilled water from different gliclazide - PEG 4000<br />

systems.<br />

Composition (w/w) IDR<br />

PD20<br />

PD60<br />

DE*100<br />

(%dissolved/min) (%)<br />

(%)<br />

(%)<br />

Gliclazide powder 0 ± 0 0 ± 0 13.05 ± 1.2 3.83 ± 0.06<br />

Gliclazide-to-PEG 4000<br />

PM 10:90 1.08 ± 0.032 21.69 ± 0.65 38.39 ± 0.19 25.38 ± 1.09<br />

SD 10:90 1.62 ± .047 32.53 ± 0.45 42.32 ± 1.5<br />

34.58 ± 0.85<br />

PM 8:92<br />

1.2 ± 0.013 24.13 ± 1.69 36.49 ± 0.18 26.99 ± 1.7<br />

SD 8:92<br />

2.07 ± 0.028 64.04 ± 0.57 50.85 ± 1.76 42.23 ± 0.28<br />

PM 5:95<br />

1.02 ± 0.11 20.75 ± 1.3 33.67 ± 1.2 23.26 ± 1.6<br />

SD 5:95<br />

3.17 ± 0.04 63.49 ± 0.89 64.29 ± 0.42 61.81 ± 0.44<br />

PM 1:99<br />

1.77 ± 0.004 35.41 ± 0.08 43.98 ± 0.49 36.07 ± 1.9<br />

SD 1:99 3.34 ± 0.075 66.94 ± 1.5 68.58 ± 1.56 62.8 ± 1.9<br />

IDR = Initial dissolution rate.<br />

PD20 = Extent of dissolution after 20 min.<br />

PD60 = Extent of dissolution after 60 min.<br />

DE% = Dissolution efficiency after 60 min.<br />

105


plain drug 10:90 PM 10:90 SD 8:92 PM 8:92 SD 5:95 PM<br />

5:95 SD 1:99 PM 1:99 SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Drug dissolved<br />

30<br />

20<br />

10<br />

0<br />

0 20 40 60 80 100 120<br />

Time (min)<br />

Figure 17: Dissolution profile of gliclazide-PEG 4000 systems.<br />

106


It is also apparent that, the rate and the extent of dissolution of Glz from<br />

(SDs) exceeded those of pure Glz or the corresponding (PMs). The DE%<br />

of (8:92) Glz-PEG 6000 co-precipitate (Table 5), for example, was<br />

60.83%.While, the DE% of the corresponding physical mixture was only<br />

25.3%.<br />

The observed higher dissolution of the prepared (SDs) could<br />

possibly due to the solubilizing effect of the carriers that may be operate<br />

in the diffusion layer immediately surrounding the drug particles. Also,<br />

each single crystallite of the drug was very intimately encircled by the<br />

soluble carrier particles which can readily dissolve and cause the aqueous<br />

medium to contact and wet the drug particles easily (Etman, 2000).<br />

Moreover, it can be generally assumed that the increased dissolution via<br />

(SDs) could be explained on the basis of alterations in the solid-state<br />

structures of the carriers and the drug particles. These structural changes<br />

include the formation of solid solution, eutectic mixtures or soluble<br />

complex between the drug and the carriers and formation of amorphous<br />

drug particles or loss of crystallinity of the drug. For most (SDs), more<br />

than one of these factors may probably be responsible for the dissolution<br />

enhancement (Trapani et al., 1999; Mura et al., 1999). Therefore, the<br />

IR spectra, differential scanning calorimetry and x-ray diffraction patterns<br />

of the pure drug, carriers and their (PMs) and (SDs) were performed.<br />

4.1. Effect of different carriers on the dissolution of Glz from<br />

(SDs):<br />

PEG 6000 had the most influential effect on the rate and the extent<br />

of dissolution of Glz, followed by PEG-4000, glucose and finally urea.<br />

107


Table 7: Dissolution parameters (±SD) of gliclazide in distilled water from different gliclazide – glucose<br />

systems.<br />

Composition (w/w) IDR<br />

PD20<br />

PD60<br />

DE*100<br />

(%dissolved/min) (%)<br />

(%)<br />

(%)<br />

Gliclazide powder 0 ± 0 0 ± 0 13.05 ± 1.2 3.83 ± 0.06<br />

Gliclazide-to-Glucose<br />

PM 1:1<br />

1.12 ± 0.038 22.51 ± 0.65 32.13 ± 1.03 22.84 ± 0.75<br />

SD 1:1<br />

1.16 ± .045 23.23 ± 0.45 40.1 ± 1.18<br />

25.52 ± 0.05<br />

PM 1:2<br />

1.03 ± 0.067 20.66 ± 1.69 34.18 ± 01.15 24.08 ± 1.4<br />

SD 1:2<br />

1.36 ± 0.028 27.27 ± 0.57 43.51 ± 1.30 29.51 ± 0.51<br />

PM 1:3<br />

1.06 ± 0.025 21.35 ± 1.3 38.53 ± 0.4 25.74 ± 0.54<br />

SD 1:3<br />

1.37 ± 0.027 27.58 ± 0.89 42.16 ± 0.8 32.45 ± 0.25<br />

PM 1:5<br />

1.42 ± 0.022 28.42 ± 0.08 42.76 ± 0.81 30.05 ± 0.43<br />

SD 1:5<br />

1.61 ± 0.075 32.38 ± 1.5 46.58 ± 1.00 35.02 ± 0.79<br />

PM 1:10<br />

1.3 ± 0.006 26.07 ± 0.13 41.68 ± 1.51 31.56 ± 1.29<br />

SD 1:10<br />

1.8 ± 0.004 36.04 ± 0.09 45.56 ± 1.06 37.05 ±0.87<br />

IDR = Initial dissolution rate<br />

PD20 = Extent of dissolution after 20 min.<br />

PD60 = Extent of dissolution after 60 min.<br />

DE% = Dissolution efficiency after 60 min.<br />

108


Plain drug 1:1PM 1:1 SD 1:2PM 1:2 SD 1:3PM<br />

1:3 SD 1:5PM 1:5 SD 1:10PM 1:10 SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Drug Released<br />

30<br />

20<br />

10<br />

0<br />

0 20 40 60 80 100 120<br />

Time (min)<br />

Figure 18: Dissolution profile of gliclazide-glucose systems.<br />

109


Table 8: Dissolution parameters (±SD) of gliclazide in distilled water from different gliclazide –urea<br />

systems.<br />

Composition (w/w) IDR<br />

PD20<br />

PD60<br />

DE*100<br />

(% dissolved/min) (%)<br />

(%)<br />

(%)<br />

Gliclazide powder 0 ± 0 0 ± 0 13.05 ± 1.2 3.83 ± 0.06<br />

Gliclazide-to-Glucose<br />

PM 1:1 0.28 ± 0.08 5.6 ± 0.60 24.53 ± 1.05 11.85 ± 1.3<br />

SD 1:1 0.68 ± 0.05 13.56 ± 1.005 24.81 ± 1.15<br />

16.49<br />

± 1.03<br />

PM 1:2 0.67 ± 0.016 13.65 ± 0.32 25.8 ± 0.69 16.25 ± 0.02<br />

SD 1:2<br />

0.95 ± 0.13 19.18 ± 1.6 34.31 ± 1.9 22.48 ± 1.6<br />

PM 1:3<br />

0.74 ± 0.033 14.88 ± 0.67 29.01 ± 0.45 17.93 ± 0.64<br />

SD 1:3<br />

1.29 ± 0.07 25.99 ± 1.44 36.45 ± 1.4 27.11 ± 1.003<br />

PM 1:5<br />

0.78 ± 0.01 15.78 ± 0.38 29.3 ± 0.29 18.66 ± 0.09<br />

SD 1:5<br />

1.28 ± 0.07 25.70 ± 1.5 35.06 ± 1.00 26.81 ± 1.79<br />

PM 1:10 1.09 ± 0.05 21.96 ± 1.055 34.9 ± 0.35 25.15 ± 0.52<br />

SD 1:10 1.42 ± 0.03 28.51 ± 0.74 39.18 ± 0.24 31.22 ±1.47<br />

IDR = Initial dissolution rate.<br />

PD20 = Extent of dissolution after 20 min.<br />

PD60 = Extent of dissolution after 60 min.<br />

DE% = Dissolution efficiency after 60 min.<br />

110


Plain drug 1:1PM 1:1 SD 1:2PM 1:2 SD 1:3PM<br />

1:3 SD 1:5PM 1:5 SD 1:10PM 1:10 SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Drug Released<br />

30<br />

20<br />

10<br />

0<br />

0 20 40 60 80 100 120<br />

Time (min)<br />

Figure 19: Dissolution profile of gliclazide-urea systems.<br />

111


The DE% after 60 minutes was found to be 89.78%, 62.8%, 37.05%<br />

and 31.22% from (1:99) PEG 6000, (1:99) PEG 4000, (1:10) glucose, and<br />

(1:10) urea solid dispersions respectively.<br />

This is in agreement with the results of the phase solubility diagram,<br />

as it was observed that, the solubility of Glz in PEGs solutions was more<br />

than that of glucose and urea. Although the solubility factor of PEG 4000<br />

was more than PEG 6000, it was found that PEG 6000 is a better carrier<br />

than PEG 4000. This is in an agreement with (Mura et al., 1999), who<br />

found that the dissolution capacity of PEG 20000 <br />

4000 although the solubilizing power of PEG 4000 <br />

20000. This may be due to the higher viscosity of dissolution medium<br />

provided by the PEG 6000 than PEG 4000 retards aggregation and<br />

agglomeration of drug particles (Doshi, 1997).<br />

4.2. Effect of carrier concentration on the dissolution of Glz from<br />

(SDs):<br />

The dissolution data of Glz from its different systems suggested that,<br />

drug-to-carrier ratio had a great influence on the drug dissolution<br />

enhancement. For example, the dissolution profile of (SDs) containing<br />

PEG 4000 (Figure 17) show different dissolution rates for dispersions<br />

containing 90%, 92%, 95%, and 99% of PEG 4000. Dispersions<br />

containing 99% of PEG 4000 appeared to be the best preparation showing<br />

a DP60 value of 68.58% which is about 5.25-fold increase compared with<br />

Glz alone.<br />

In case of all carriers (Figure 16-19), the dissolution of Glz was<br />

enhanced as the proportion of the polymer increased. This is consistent<br />

with that reported by Gul and Zhu 1998, who stated that, the<br />

dissolution rate of ibuprofen increased with increasing PEG 10000<br />

loading, and this may be attributed to the finer subdivision of the drug<br />

particles in dispersions containing higher carrier loading. On the other<br />

112


hand, Moneghini et al., 1998 and Chutimaworapan et al., 2000a stated<br />

that, when the proportion of PEG increased, the dissolution was<br />

suppressed. This result could be ascribed to the formation of a viscous<br />

hydrophilic layer around the particles of the drug that slowed the drug<br />

release into the dissolution medium.<br />

It was important to find the optimal drug – carrier ratio in order to<br />

achieve the optimal dissolution profile. When the weight ratio of carrier<br />

decreased below its critical concentration, the concentration being too<br />

small was probably insufficient to enhance dissolution to the maximum<br />

extent hence, as the proportion of carrier increased, the dissolution rate<br />

also increased. Above this critical concentration, as the proportion of<br />

carrier increased, the longer time required for diffusion of the drug from<br />

the matrix probably resulted in a decreased dissolution rate<br />

(Tantishaiyakul et al., 1996).<br />

All data are summarized in Table 9 and Figure 20<br />

Table 9: Collective data for dissolution of Glz obtained from<br />

different carriers used.<br />

System a<br />

% Released b<br />

113<br />

% Increase c<br />

Glz 13.05 -<br />

Drug : carrier<br />

(1:99) PEG 6000 SD 100.49 670.038<br />

(1:99) PEG 4000 SD 68.58 425.51<br />

(1:10) Glu SD 45.56 249.11<br />

(1:10) UR SD 39.18 200.22<br />

a 40 mg of the drug or its equivalent were used.<br />

b After 60 min.<br />

c In relation to drug alone.


10<br />

1:99 PEG 6000<br />

9<br />

8<br />

1:99 PEG 4000<br />

7<br />

6<br />

1:10 glucose<br />

5<br />

1:10 urea<br />

A / B<br />

4<br />

3<br />

2<br />

1<br />

0<br />

Figure 20: Ratio between % of gliclazide dissolved from (A) drug in different solid dispersions and (B) drug<br />

114


In order to shed light on the mechanism of dissolution enhancement<br />

from solid dispersions, further studies were performed on the investigated<br />

solid dispersions, physical mixtures and individual components. In case<br />

of urea and glucose solid dispersions and their respective physical<br />

mixtures the studies were performed at drug to carrier ratios (1:5), while<br />

in case of PEG 4000 and PEG 6000, the studies were performed at (1:9)<br />

drug to carrier ratio, as higher drug content is more suitable for practical<br />

use (Okonogi et al., 1997).<br />

5. Fourier-transform infrared spectroscopy:<br />

FTIR spectra were performed to investigate the possible type of<br />

interaction between Glz and different carriers (Figures 21-24).<br />

(Table 10) showed that the characteristic shoulders of Glz were<br />

traced at 3274.2 cm -1 (N – H stretching), 3192.9, 3113.2 cm -1 (C – H<br />

aromatic), 2950-2836 cm -1 (C-H aliphatic), 1350, 1164.3 cm -1 (S=O<br />

asymmetrical and symmetrical band) and 1596 (N-H deformation). The<br />

major peak of C=O was at 1709 cm -1 .<br />

In case of PEG 4000 and PEG 6000 systems, the carbonyl stretching<br />

band of Glz that appeared at 1710.3 cm -1 decreased in the intensity with<br />

the disappearance of the aromatic C-H stretching band and N-H<br />

stretching band and predominance of O-H band corresponding to PEGs.<br />

It was concluded from the chemical structures that an interaction of a<br />

significant magnitude could be present between the aromatic hydrogens<br />

of the drug and the hydroxyl groups of PEG, Mukne and Nagarsenker,<br />

2004 attributed the complete disappearance of the aromatic stretching<br />

vibrations of the phenyl group of triametrene by its complexation with ß-<br />

cyclodextrin to be due to the significant interaction between the phenyl<br />

group of triametrene and the cyclodextrin. On contrary, Glz – glucose<br />

systems showed peaks at 3410, 3280, 2943, 2872 and 1709 cm -1 which<br />

were the superimposed peaks of the two components. In spectra of<br />

1


Glz systems with UR, no differences in the positions of the absorption<br />

bands was observed, hence providing evidence for the absence of any<br />

chemical interactions in the solid state between Glz and these carriers. In<br />

the physical mixture and solid dispersion spectra, C=O and N-H peaks of<br />

UR were overlapped with C=O and N-H of Glz, which formed a two<br />

broad bands around 3300 cm -1 . If the drug and these carriers interact, then<br />

the functional groups in the FTIR spectra will show bands changes and<br />

broadening compared to the spectra of the plain carriers (Silverstein et<br />

al., 1991).<br />

2


Table 10: FTIR spectra of Glz solid dispersions and physical<br />

mixtures compared with individual components.<br />

System Assignment max (cm -1 )<br />

Glz<br />

PEG 6000<br />

Glz – PEG 6000<br />

(PM)(10:90)<br />

Glz – PEG 6000<br />

(SD)(10:90)<br />

N – H (stretching)<br />

C – H (aromatic)<br />

C – H (aliphatic)<br />

C=O<br />

N-H (deformation band)<br />

S=O (asymmetrical and<br />

symmetrical band)<br />

-O-H (stretching)<br />

C-H (stretching)<br />

C-O (ether)<br />

O-H (bending)<br />

-O-H (stretching)<br />

C-H (stretching)<br />

C=O<br />

C-O (ether)<br />

O-H (bending)<br />

-OH (stretching)<br />

C-H (stretching)<br />

C=O<br />

C-O (ether)<br />

O-H (bending)<br />

3<br />

3274.2<br />

3192.9 - 3113.2<br />

2950 - 2867 - 2836<br />

3447<br />

2886.8<br />

1710.2<br />

1112.3<br />

1345.7<br />

1709<br />

1596<br />

1350 -1164<br />

3445.8<br />

2887<br />

1110.7<br />

1344<br />

3446<br />

2888.2<br />

1710.3<br />

1110.9<br />

1345.5


Cont. Table 10: FTIR spectra of Glz solid dispersions and physical<br />

mixtures compared with individual components.<br />

System Assignment max (cm -1 )<br />

Glz<br />

PEG 4000<br />

Glz – PEG 4000<br />

(PM)(10:90)<br />

Glz – PEG 4000<br />

(SD)(10:90)<br />

N – H (stretching)<br />

C – H (aromatic)<br />

C – H (aliphatic)<br />

C=O<br />

N-H (deformation band)<br />

S=O (asymmetrical and<br />

symmetrical band)<br />

-OH (stretching)<br />

C-H (stretching)<br />

C-O (ether)<br />

O-H (bending)<br />

-OH (stretching)<br />

C-H (stretching)<br />

C=O<br />

C-O (ether)<br />

O-H (bending)<br />

-O-H (stretching)<br />

C-H (stretching)<br />

C=O<br />

C-O (ether)<br />

O-H (bending)<br />

4<br />

3274.2<br />

3192.9 - 3113.2<br />

2950 - 2867 - 2836<br />

1709<br />

1596<br />

1350 -1164<br />

3414.3<br />

2887.6<br />

1110.4<br />

1344.7<br />

3447.2<br />

2887.23<br />

1710.3<br />

1110.5<br />

1345.2<br />

3422.6<br />

2888.0<br />

1710.3<br />

1112.1<br />

1346.2


Cont. Table 10: FTIR spectra of Glz solid dispersions and physical<br />

mixtures compared with individual components.<br />

System Assignment max (cm -1 )<br />

Glz<br />

Glucose<br />

Glz – glu<br />

(PM)(1:10)<br />

Glz – glu<br />

(SD)(1:10)<br />

N – H (stretching)<br />

C – H (aromatic)<br />

C – H (aliphatic)<br />

C=O<br />

N-H (deformation band)<br />

S=O (asymmetrical and<br />

symmetrical band)<br />

-O-H (stretching)<br />

5<br />

(broad)<br />

C-H (stretching)<br />

O-H (bending)<br />

-OH (stretching)<br />

-NH (stretching)<br />

C-H (stretching)<br />

C=O<br />

O-H (bending)<br />

-O-H (stretching)<br />

-NH (stretching)<br />

C-H (stretching)<br />

C=O<br />

O-H (bending))<br />

3274.2<br />

3192.9 - 3113.2<br />

2950 - 2867 - 2836<br />

1709<br />

1596<br />

1350 -1164<br />

3411.6 -3316.0<br />

2944.1<br />

1342.0<br />

3410.4<br />

3280.7<br />

2943.4<br />

1709.9<br />

1346.7<br />

3408.8<br />

3276.3<br />

2941.5<br />

1709.9<br />

1348.0


Cont. Table 10: FTIR spectra of Glz solid dispersions and physical<br />

mixtures compared with individual components.<br />

System Assignment max (cm -1 )<br />

Glz<br />

UR<br />

Glz – UR<br />

(PM)(1:10)<br />

Glz – UR<br />

(SD)(1:10)<br />

N – H (stretching)<br />

C – H (aromatic)<br />

C – H (aliphatic)<br />

C=O<br />

N-H (deformation band)<br />

S=O (asymmetrical and<br />

symmetrical band)<br />

-N-H (stretching)<br />

C=O<br />

-C-N<br />

-N-H (stretching)<br />

C=O<br />

-C-N<br />

-N-H (stretching)<br />

C=O<br />

-C-N<br />

6<br />

3274.2<br />

3192.9 - 3113.2<br />

2950 - 2867 - 2836<br />

1709<br />

1596<br />

1350 -1164<br />

3445.7- 3347.4<br />

1678.8 – 1622.7<br />

1152.4<br />

3446.5-3347.6-<br />

3277.9<br />

1707.4-1682-<br />

1622.8<br />

1162.1<br />

3445.7-3347.6-<br />

3276.0<br />

1708.1-1682.4-<br />

1623.4<br />

1162.4


Wave number (cm -1 )<br />

Figure 21: FTIR spectra of Glz –PEG 6000 systems A) Glz ; B) pure<br />

PEG 6000; C) PM (1:9) and D) SD (1:9).<br />

7<br />

D<br />

C<br />

B<br />

A


Wave number (cm -1 )<br />

Figure 22: FTIR spectra of Glz –PEG 4000 systems A) Glz ; B) pure<br />

PEG 4000; C) PM (1:9) and D) SD (1:9).<br />

8<br />

D<br />

C<br />

B<br />

A


Figure 23: FTIR spectra of Glz –glucose systems A) Glz ; B) pure<br />

glu; C) PM (1:10) and D) SD (1:10).<br />

Wave number (cm -1 )<br />

9<br />

D<br />

C<br />

B<br />

A


Figure 24: FTIR spectra of Glz –UR systems A) Glz ; B) pure UR; C)<br />

PM (1:10) and D) SD (1:10).<br />

Wave number (cm -1 )<br />

6. Differential scanning calorimetry:<br />

10<br />

D<br />

C<br />

B<br />

A


It was the general aim to prepare dispersions in which the drug was<br />

dispersed in as near a molecular state as possible to provide a thermo<br />

energetic state of the drug of high aqueous solubility once the carrier<br />

dissolved. Thermal analysis, especially DSC, had a powerful tool<br />

evaluating the drug – carrier interactions (Nour, 1993). DSC is<br />

particularly useful in determining the solubility of the drug in a polymeric<br />

and is capable of detecting polymorphic modifications. Interactions in the<br />

samples are derived or deduced from DSC by changes in thermal events<br />

such as elimination of an endothermic or exothermic peak or appearance<br />

of a new peak (Ford and Timmins, 1989). In order to get evidence on<br />

the possible interaction between Glz and the investigated carriers, DSC<br />

studies were performed on the prepared physical mixtures, solid<br />

dispersions as well as various individual components. The DSC<br />

thermograms of Glz containing systems are shown in (Figures 25-28).<br />

The heat of fusion and fusion temperature values for the raw materials<br />

and binary systems are represented in (Table 11). The DSC curves of<br />

pure Glz exhibited a sharp endothermic peak at 166.2, which corresponds<br />

to its melting point.<br />

The DSC themograms of Glz-PEG 4000 and Glz-PEG 6000 solid<br />

dispersions and corresponding physical mixtures showed no Glz<br />

endothermic peak but did exhibit the endothermic peaks due to the fusion<br />

of the carriers. This result indicated that Glz might be in an amorphous<br />

state. Yakou et al., 1984, studied the physicochemical characteristics of<br />

phenytoin – PEG 4000 solid dispersion; they observed the disappearance<br />

of sharp endothermic peak corresponds to phenytoin melting point with<br />

predominance of that corresponds to PEG 4000 melting point. They<br />

concluded that phenytoin was uniformly dispersed in an amorphous state<br />

in a solid matrix of PEG 4000. The absence of a drug melting<br />

endothermic peak could also have been due to its dissolution in the<br />

11


melted carrier. Mura et al., 1999, studied the DSC scans of solid<br />

dispersion of naproxen in binary systems with different molecular<br />

weights, they observed the disappearance of the drug melting peak which<br />

indicated the dissolution of the naproxen in the melted carrier. A slight<br />

change occurs in the shape of PEGs endothermic peaks which appeared<br />

broadend in solid dispersions.<br />

In case of UR, no differences were apparent between DSC scans of<br />

the (PM) and the (SD) (Figure 28). In fact, the two systems displayed<br />

two endothermic peaks corresponding to the carrier fusion, whereas drug<br />

endothermic effect was not detected and this may be due to its dissolution<br />

in the melted carrier.<br />

(Figure 27) illustrates the DSC thermograms of Glz –glucose<br />

systems. The absence of of Glz peak and the predominance of glucose<br />

peaks. This suggests that Glz is completely soluble in liquid phase of<br />

glucose (Domian et al., 2000).<br />

12


Table 11: Fusion temperatures (Tc) and heat of fusion (Glz<br />

solid dispersions and physical mixtures compared with individual<br />

components.<br />

System Fusion temperature<br />

(Tc) (ºC)<br />

13<br />

Heat of fusion<br />

(<br />

Glz 166.2 135.38<br />

PEG 6000 60.52 184.49<br />

Glz – PEG 6000<br />

(PM)(10:90) 60.02 169.52<br />

Glz – PEG 6000<br />

(SD)(10:90) 59.42 180.55<br />

PEG 4000 60.52 192.58<br />

Glz – PEG 4000<br />

(PM)(10:90) 60.38 162.52<br />

Glz – PEG 4000<br />

(SD)(10:90) 59.97 167.82<br />

Glu 156.13 223.18<br />

Glz – glu (PM)<br />

(1:10)<br />

Glz – glu (SD)<br />

(1:10)<br />

154<br />

182.46<br />

153.21<br />

183.65<br />

193.21<br />

42.72<br />

194.19<br />

58.26<br />

UR 132.97 225.17<br />

Glz – UR (PM)<br />

(1:10)<br />

Glz – UR (SD)<br />

(1:10)<br />

131.12<br />

192.52<br />

132.23<br />

181.92<br />

176<br />

110.56<br />

170.97<br />

73.12


Temp (c)<br />

Figure 25: DSC spectra of Glz –PEG 6000 systems A) Glz ; B) pure<br />

PEG 6000; C) PM (1:9) and D) SD (1:9).<br />

14<br />

D<br />

C<br />

B<br />

A


Temp (c)<br />

Figure 26: DSC spectra of Glz –PEG 4000 systems A) Glz ; B) pure<br />

PEG 4000; C) PM (1:9) and D) SD (1:9).<br />

15<br />

D<br />

C<br />

B<br />

A


Figure 27: DSC spectra of Glz –glucose systems A) Glz; B) pure glu;<br />

C) PM (1:5) and D) SD (1:5).<br />

Temp(c)<br />

16<br />

D<br />

C<br />

B<br />

A


Figure 28: DSC spectra of Glz –UR systems A) Glz ; B) pure UR; C)<br />

PM (1:5) and D) SD (1:5).<br />

Temp (c)<br />

17<br />

D<br />

C<br />

B<br />

A


7. X-ray diffraction:<br />

The x-ray diffractuion patterns of Glz, PEG 4000, PEG 6000, glu,<br />

UR, physical mixtures and solid dispersions were illustrated in (Figures<br />

29-32). Their characteristic peaks and intensities are presented in (Table<br />

12)<br />

Glz was a highly crystalline powder with characteristic diffraction<br />

<br />

addition there were some other peaks of lower intensity.<br />

In case of untreated PEG 6000, there were sharp peaks at 19° and<br />

23.12°, while in case of PEG 4000 the diffraction peaks were traced at<br />

19.016° and 23.217°. The diffraction patterns of PEG 6000 and PEG<br />

4000 solid dispersions and physical mixtures are nearly identical to that<br />

of untreated ones. The peaks of Glz were completely missed thus<br />

indicating that Glz was in amorphous form. This was in line with our<br />

findings from FTIR analysis where interactions might be present between<br />

the drug and either of these two carriers.<br />

Glucose and urea in pure form revealed high degree of crystallinity.<br />

X-ray patterns of glucose solid dispersions and physical mixtures showed<br />

the superimposed diffraction peaks of both drug and carrier with<br />

reduction in their intensities. On other hand , in case of urea solid<br />

dispersion and physical mixture, the diffraction peaks of Glz was not<br />

observed whereas the diffraction peaks of urea was noted. This indicated<br />

that Glz was in amorphous state (Okonogi et al., 1997).<br />

18


Table 12: Intensities at characteristic differº) for some gliclazide solid dispersions and physical<br />

mixtures compared with individual components.<br />

System intensity intensity intensity intensity intensity intensity<br />

Gliclazide 14.58 33.1 19.68 43.1 20.12 36.9 20.58 100 22.72 24.9 28.4 35.5<br />

PEG 6000 19 86.5 23.12 100<br />

Glic-PEG 6000<br />

(10:90) (PM) 19.03 89.1 23.12 100<br />

Glic-PEG 6000<br />

(10:90) (SD) 19.03 100 23.21 97.6<br />

PEG 4000 19.016 100 23.217 90.6<br />

Glic-PEG 6000<br />

(10:90) (PM) 19.1 99.7 23.26 100<br />

Glic-PEG 6000<br />

(10:90) (SD) 19.02 92.4 23.2 100<br />

19


Cont.Table 12:º) for some gliclazide solid dispersions and physical<br />

mixtures compared with individual components.<br />

System intensity intensity intensity intensity intensity intensity<br />

Gliclazide 14.58 33.1 19.68 43.1 20.12 36.9 20.58 100 22.72 24.9 28.4 35.5<br />

Glucose 20.52 100 25.38 30.9 17.01 44 28.39 64.2<br />

Glic-glucose<br />

(1:10) (PM) 20.74 37.2 14.39 92.2 17.055 57.7 10.74 100 18.15 70.3 22.014 30.1<br />

Glic- glucose<br />

(1:10) (SD) 20.57 100 14.62 25.2 17.067 25.5 28.4 35 19.72 7.2 22.017 17.1<br />

Urea 22.044 100 28.4 35<br />

Glic-urea<br />

(1:10) (PM) 22.22 96.3 22 45.7 31.47 100 35.47 36.9<br />

Glic-urea<br />

(1:10) (SD) 22.15 100 35.17 57<br />

20


2-Theta-Scale<br />

Figure 29: X-ray spectra of Glz –PEG 6000 systems A) Glz ; B) pure<br />

PEG 6000; C) PM (1:9) and D) SD (1:9).<br />

21<br />

D<br />

C<br />

B<br />

A


2-Theta-Scale<br />

Figure 30: X-ray spectra of Glz –PEG 4000 systems A) Glz ; B) pure<br />

PEG 4000; C) PM (1:9) and D) SD (1:9).<br />

22<br />

D<br />

C<br />

B<br />

A


Figure 31: X-ray spectra of Glz –glucose systems A) Glz ; B) pure<br />

glu; C) PM (1:5) and D) SD (1:5).<br />

2-Theta-Scale<br />

23<br />

D<br />

C<br />

B<br />

A


Figure 32: X-ray spectra of Glz –UR systems A) Glz ; B) pure UR; C)<br />

PM (1:5) and D) SD (1:5).<br />

2-Theta-Scale<br />

24<br />

D<br />

C<br />

B<br />

A


Conclusion:<br />

1- The preparation of Glz solid dispersions was examined with<br />

different carriers.<br />

2- The proportion and properties of the carrier used present an important<br />

influence on the properties of the resulting soild dispersions<br />

3- PEG 4000, PEG 6000, glucose and UR were used as carriers, led to an<br />

increase in the dissolution rate of Glz .<br />

4- FTIR, DSC and XRD diffraction revealed an interaction between<br />

Glz and PEG 4000 and PEG 6000, with possibility of a<br />

polymorphic change in Glz for all systems used.<br />

25


Introduction<br />

Percutaneous penetration involves drug dissolution in the vehicle,<br />

diffusion of the solubilized drug from the vehicle to the surface of the<br />

skin and drug penetration through skin layers. Selection of the<br />

appropriate vehicle and modification of drug characteristics may improve<br />

penetration (Mario et al., 2005).<br />

Permeation of the drug from prepared systems in donor<br />

compartment through a semipermeable membrane involves three<br />

consecutive processes: first, dissolution of the solid dispersed particles,<br />

then diffusion of the drug across the dissolution media, and finally its<br />

permeation through the membrane. All three processes make a<br />

contribution to the overall diffusion rate (Mario et al., 2005).<br />

To improve the release rate of the drug, solid dispersions were<br />

incorporated into the topical bases. The effectiveness of incorporation of<br />

solid dispersions in topical formulations on the release of the Glz was<br />

determined by comparing the percent of the drug released after six hours<br />

in presence and absence of solid dispersions.<br />

27


1- Materials and supplies:<br />

Experiment and methodology<br />

* Hydroxy propylmethyl cellulose 50 cp (HPMC) (Sigma<br />

Chemical, St.Louis, MO, USA)<br />

* White soft paraffin, wool fat, cetyl alcohol, propylene glycol,<br />

sodium lauryl sulfate (SLS), polyethylene glycol 400, liquid<br />

paraffin, hard paraffin and borax (El-Nasr CO. Cairo, Egypt).<br />

* Octanol , span 80 (Merk Sharp and Dohmn, Germany)<br />

* White beeswax, gum acacia (El-Gomhouria Co.,Egypt).<br />

* Glucose- LS, GOD-PAP, Modern Laboratory Chemicals, Egypt.<br />

* Streptozotocin (Sigma Chemical Company,USA).<br />

* Other materials were mentioned previously in chapter one.<br />

2- Equipment:<br />

* Diffusion glass cell, this is composed of an open ends glass<br />

tube with 2.9 cm as external diameter, 2.6 cm as internal<br />

diameter and length of 30 cm. Semipermeable cellophane<br />

membrane was stretched over one open end of glass tube and<br />

made watertight by a rubber band.<br />

* Viscometer (Fungi lab S.A, Spain).<br />

* Eppendorf Centrifuge 5415 C (maximum speed 14000 min -1 ), West<br />

Germany.<br />

* UV/VIS Spectrophotometer (Jenway, 6105).<br />

* PH meter (Cole-Parmer Instrument Co USA).<br />

* On Call EZ Blood Glucose Meter (San Diego, CA 92121, USA).<br />

* Other equipments were mentioned previously in chapter one.<br />

3- Software:<br />

Microsoft Office XP, Microsoft Corporation, USA.<br />

28


SPSS statistics Package, SPSS Institute Inc., Cary, USA.<br />

4. Methods:<br />

4.1. Determination of partition coefficient of Glz:<br />

*** Preparation of saturated solution of the drug:<br />

An excess of the Glz (10 mg) was placed into 25-ml glass vial<br />

containing 10 ml distilled water. The glass vials was closed with stopper<br />

and cover-sealed with cellophane membrane to avoid solvent loss .The<br />

content of the suspension was equilibrated by shaking in a<br />

thermostatically controlled water bath at 25°C for 7 days. After<br />

attainment of equilibrium, the content of the vial was then filtered<br />

through a double filter paper (Whatman 42).The filtrate was assayed<br />

spectrophotometrically at 227 nm to measure the amount of the drug.<br />

*** Method:<br />

In glass vials 5 ml of saturated solution of the drug were added to 5<br />

ml of n-octanol. The vials were placed in a thermostatically controlled<br />

water bath at 25°C for 24 hrs. The aqueous phase was separated from the<br />

oily phase by the separating funnel and the amount of the drug in aqueous<br />

phase was assayed spectrophotometerically at 227 nm using distilled<br />

water as blank. The concentration of the drug was obtained from a<br />

previously constructed calibration curve. Partition coefficient of Glz in<br />

octanol/water system was determined according to the following equation<br />

(El-Nahas, 2001):<br />

Conc. of Glz in oily phase<br />

Partition coefficient = --------------------------------------------<br />

Conc. of Glz in aqueous phase<br />

29


4.2. Preparation of solid dispersions:<br />

Solid dispersions of Glz with each of PEG 6000, PEG 4000, urea<br />

and glucose were prepared at weight ratios of 8:92 (drug:carrier) for<br />

PEGs (SDs) and 1:10 (drug:carrier) for urea and glucose SDs. The<br />

amount of SDs introduced was adjusted to maintain the drug<br />

concentration at 1% in the formulations.<br />

4.3. The methods of preparation of topical preparations:<br />

The following formulae were selected in which 10 mg of Glz, or its<br />

equivalent of (SDs) was incorporated in each one gram of the topical<br />

formula. In case of urea and glucose, (SDs) that demonstrated the best<br />

dissolution properties, (1:10) drug to carrier ratio, were used. However in<br />

case of PEG 4000 and PEG 6000, (SDs) of (8:92) drug to carrier ratio<br />

were used because the ratio of (1:99) that gave the highest dissolution<br />

was not practically suitable for incorporation into the base due to higher<br />

powder content.<br />

4.3.1. Water soluble base:<br />

Polyethylene glycol base :( U.S.P. XXII).<br />

- PEG 4000 40 gm<br />

- PEG 400 60 gm<br />

Preparation:<br />

PEG 4000 was melted at 60° C on a water bath. Then PEG 400<br />

containing the drug or the solid dispersion was added. The mixture was<br />

continuously stirred until congealed and packed in a plastic jar and stored<br />

at ambient temperature until used.<br />

4.3.2. Absorption base (B.P. 1963):<br />

- Wool fat 5 gm<br />

-Cetyl alcohol 5 gm<br />

-Hard paraffin 5 gm<br />

30


-White soft paraffin 85 gm.<br />

Preparation:<br />

Accurate amount of the drug or the solid dispersion was weighed,<br />

levigated and incorporated into the melted base with continuous stirring<br />

until congealed then packed into plastic jar until used.<br />

4.3.3. Emulsion bases:<br />

O/W emulsion base (Beeler’s base) (Ezzedeen et al., 1986).<br />

-White bees wax 1 gm<br />

-Cetyl alcohol 15 gm<br />

-Propylene glycol 10 gm<br />

-Sodium lauryl sulphate 2 gm.<br />

-Water 72 gm.<br />

W/O emulsion base: (Ezzedeen et al., 1986).<br />

-Liquid paraffin 45 gm<br />

-White bees wax 10 gm<br />

- Wool fat 2 gm<br />

- Borax 8 gm<br />

-Water 41 gm<br />

- Span 80 1 gm<br />

Preparation:<br />

The aqueous phase and the oil phase were placed in separate<br />

containers and heated at 70°C .The drug was dissolved in the oily phase.<br />

Then the aqueous phase was added to the oil phase at the same<br />

temperature with continuous stirring until cool and congealed<br />

31


4.3.4. Hydroxy propyl methylcellulose gel (Sobati, 1998):<br />

- HPMC 12 gm<br />

- Water 88 gm<br />

Preparation:<br />

The drug was dispersed in a quantity of water then the gelling agent<br />

was added with continuous stirring, set aside for complete swelling and<br />

the weight was adjusted by the addition of the water.<br />

All the formulations mentioned previously were summarized in<br />

(Table13)<br />

4.4. In vitro release of Glz from different topical formulations:<br />

The release study was determined using the simple dialysis<br />

technique. In this method, 1 gm of the tested formulation containing (10<br />

mg of the drug) was accurately weighed over the cellophane membrane<br />

which previously soaked in the phosphate buffer pH 7.4 for 30 minutes,<br />

the loaded membrane was stretched over the end of a glass tube of about<br />

2.9 cm as external diameter, and 2.6 cm as internal diameter as shown in<br />

(Figure 33) (Donor).<br />

The diffusion cell was placed at the center of 1000 ml dissolution cell<br />

containing 100 ml of phosphate buffer pH 7.4. The donor was suspended<br />

in the acceptor in such a manner that the membrane was located just<br />

below the surface of the sink condition. The stirring rate was 100 rpm and<br />

the temperature was maintained at 37 ± 0.5 °C. At suitable time intervals<br />

(30, 60, 90,120,150,180, 240,300 and 360 minutes), 2.5 ml sample was<br />

withdrawn from the sink solution and replaced with an equivalent amount<br />

of the fresh release medium kept at 37 °C, diluted with methanol and<br />

assayed spectrophotometerically at 227 nm using a suitable blank.<br />

32


Each experiment was done in triplicate, and the average was calculated.<br />

The cumulative amount of the drug released was calculated as mentioned<br />

before.<br />

33


Figure 33: Diagrammatic representation of the drug diffusion<br />

apparatus.<br />

34


4.5. Effect of incorporation of solid dispersions in different topical<br />

preparations:<br />

Previously prepared solid dispersions were incorporated in the<br />

topical formulations that demonstrated the best release results (water<br />

soluble base, HPMC gel and O/W cream).In vitro release of these<br />

preparation were done as mentioned above.<br />

4.6. Detrmination of viscosity of topical different bases:<br />

The viscosity of each of PEG bases, O/W cream and HPMC gel which<br />

contains Glz :PEG 4000 (8:92)SD and Glz : glu (1:10) SD was<br />

determined at room temperature, using spindle number 5 at 2 r.p.m (El-<br />

Megrab et al., 2006).<br />

4.7. Kinetic evaluation of the in vitro release data:<br />

The data obtained from the experiments were analyzed to know the<br />

mechanism of the release of the drug using the following kinetic<br />

equations:<br />

(I) Zero order kinetics:<br />

A=A-k<br />

Where A<br />

A = drug concentration at time (t).<br />

t = time interval.<br />

k<br />

When this linear equation is plotted with the percent of drug remained on<br />

the vertical axis and (t) on the horizontal axis, a straight line would be<br />

obtained with (R) correlation coefficient, a slope equal to (-k<br />

intercept equal to (A<br />

35


Half time: is the time required for a drug to decompose to one half of the<br />

original concentration or it is the time at which A is decreased to 1 /2 A<br />

(Martin, 1994 ).<br />

(II) First order kinetics:<br />

t1/2 = A<br />

ln A = ln A- kt<br />

log A = log A- kt/ 2.303<br />

Where A<br />

A = drug concentration at time (t).<br />

t = time interval.<br />

k<br />

When this linear equation is plotted with the logarithm amount of percent<br />

drug remained on the vertical axis and (t) on the horizontal axis, a straight<br />

line would be obtained with (R) correlation coefficient, a slope equal to (-<br />

kt/ 2.303) and an intercept equal to (log A(Martin, 1994 ).<br />

The half life for first order kinetics equal to<br />

(III) Higuchi diffusion model:<br />

t1/2 = 0.693 / k.<br />

i) The diffusion occurs in a direction opposite to that of increasing<br />

concentration. That is to say, diffusion occurs in the direction of<br />

decreasing concentration of diffusant, Fick <br />

diffusion (Martin, 1994). A simplified Higuchi diffusion<br />

equation for drug released from topical preparation is.<br />

M = Q = 2C ½<br />

36


Where:<br />

M = Q = amount of the drug released into the receptor phase at time t.<br />

C<br />

<br />

t = time of release.<br />

D = diffusion coefficient of the drug.<br />

This equation describes drug release as being linear with the square root<br />

of the time<br />

Q = k t ½<br />

4.8. In vitro permeation of Glz through abdominal rabbit skin:<br />

4.8.1. Preparation of rabbit skin:<br />

The abdomen of white male rabbits (Jia-You et al., 1996; Hosny et<br />

al., 1998), weighing 2-3 Kg, were shaved by an electric hair clipper. The<br />

rabbit was scarified; the skin then excised surgically, without injury. The<br />

dermal side of the skin was carefully cleared of adhering blood vessels,<br />

fats or subcutaneous tissues using fine-point forceps and surgical scissors<br />

and washed with distilled water (Ceschel et al., 1999). The skin was<br />

stored and frozen. The frozen skin was thawed prior to cutting into pieces<br />

for experimental studies. The pieces of the skin were equilibrated by<br />

soaking in sörensen’ phosphate buffer (pH 7.4) for about one hour before<br />

the beginning of each experiment (Larrucea et al., 2001).<br />

4.8.2. In vitro permeation studies:<br />

In vitro permeation studies of Glz and Glz solid dispersions from<br />

different topical formulations were carried out utilizing locally fabricated<br />

diffusion cell. The excised rabbit skin was mounted on one end of the<br />

37


vertical diffusion cell (internal diameter 2.6 cm) by a rubber band with<br />

the sratum corneum side facing the donner compartment and the dermal<br />

side facing the receptor compartment, and the total area available for<br />

penetration was 5.3 cm 2 . Several drug concentrations ranging from 10 to<br />

50 mg of Glz per one gram of the formulation were applied on the<br />

stratum corneum. The diffusion cell was hanged into the center of the<br />

glass beaker, containing 100 ml of sörensen’ phosphate buffer (pH 7.4)<br />

(Mura et al., 2000; Ghazy et al., 2004) in such way that, the dermal<br />

surface was just flushed to the permeation fluid (Fang et al., 2003).The<br />

permeation fluid was maintained at 37°C ± 0.5°C and stirred at 100 rpm<br />

in thermostatically controlled water bath (Tehrani and Mehramizi,<br />

2000).To avoid evaporation, the beaker was kept covered during the<br />

experiment. Four-millimeter samples were withdrawn from the receptor<br />

phase at specified time intervals and immediately replaced by an equal<br />

volume of fresh buffer solution (pH 7.4) at the same temperature (37°C ±<br />

0.5°C) to maintain the volume of the receptor phase constant during the<br />

experiment . The samples were analyzed spectrophotometrically at 227<br />

nm against sörensen’ phosphate buffer (pH 7.4) as a blank (Larrucea et<br />

al., 2001). Each experiment was performed three times and the average<br />

was calculated. The cumulative amount of the drug permeated was<br />

calculated as mentioned before.<br />

4.8.3. Statistical analysis:<br />

Data were expressed as mean of three experiments ± the standard<br />

error (S.E.). The obtained data were compared statistically using One-<br />

way analysis of variance (ANOVA) test of significance on a computer<br />

statistical SPSS analysis program. A p-value of 0.05 or less was<br />

considered to be significant (Suwanpidokkul et al., 2004).<br />

38


4.9. In vivo studies:<br />

4.9.1. Animals:<br />

The animals used for the anti-diabetic and hypoglycemic activity<br />

study were white adult albino rats weighing between 200-250 gm. The<br />

animals were housed under standard laboratory conditions.<br />

4.9.2. Hypoglycemic activity in normal rats:<br />

The hair on the backside of the rats was removed with an electric hair<br />

clipper on the previous day of the experiment. The oral doses were given<br />

using a round tipped stainless steel needle attached to 1 ml syringe.<br />

Following an overnight fast, rats were divided into 4 groups (n=5) and<br />

treated as follows:<br />

Group I (Control) – 1ml gum acacia suspension was given orally.<br />

Group II - 25 mg/kg Glz in mucilage of gum acacia was given orally<br />

(Stetinova et al., 2007).<br />

Group III –1 gm water soluble ointment base containing 25 mg of the<br />

Glz was applied on 4cm 2 of the skin. Many trials on rats were done to<br />

find a suitable topical formula. The dose of 25 mg drug was selected by<br />

conducting a series of experiments with graded doses ranging between 10<br />

to 50 mg. The application site was covered with a non-occlusive dressing<br />

and wrapped with a semi-occlusive bandage.<br />

Group IV –1 gm water soluble ointment base containing certain amount<br />

of Glz - PEG 6000 solid dispersion (10:90) equivalent to 25 mg Glz was<br />

applied on 4cm 2 of the skin.<br />

Blood samples were collected in eppendorf predose (0 hr) and 2, 4, 6, 8<br />

and 24 hours post dose from the orbital sinuses; the serum glucose<br />

concentrations were assayed based on the standard glucose oxidase<br />

39


method (El-Sayed et al., 1989) using a commercial kit according to the<br />

supplied instructions as follows:<br />

Principle:<br />

reaction:<br />

Glucose present in the sample is determined according the following<br />

Glucose + O2 + H2O2 glucose oxidase enzyme gluconic acid + H2O2<br />

------------------><br />

2 H2O2 + phenol amino-4-antipyrine peroxidase enzyme quinoneimine+4<br />

H2O<br />

Sample preparation:<br />

-----------------><br />

The collected blood samples were left for 15 min in the refrigerator,<br />

then the serum was separated using a centrifuge operating at 4000 rpm for<br />

20 min, then the serum was taken by a syringe in a test tube.<br />

Procedure of measurement:<br />

The amounts of samples and standard used are summarized in (Table 14).<br />

Table 14: Amounts of sample and standard used.<br />

Blank Standard Sample<br />

Reagent 1.0 ml 1.0 ml 1.0 ml<br />

Standard<br />

reagent<br />

(100mg<br />

glu/dl)<br />

------- -------<br />

Sample ------- ------- <br />

40


Samples and reagent were mixed and incubated for 10 min at 37°C.<br />

The absorbance of sample (Asample) and standard (Astandard) against reagent<br />

blank were measured. The intensity of the developed pink colour was<br />

measured spectrometrically at 500 nm against a blank solution.<br />

Calculation:<br />

(Asample)<br />

Glucose concentration (mg/dl) = ----------- x 100<br />

(Astandard)<br />

4.9.3. Anti-diabetic activity in diabetic rats:<br />

4.9.3.1. Induction of diabetes mellitus:<br />

The overnight fasted rats were made diabetic by a single<br />

intraperitoneal injection of streptozotocin (STZ) (50 mg/kg; i.p) dissolved<br />

in citrate buffer (pH 4.5). The blood glucose was measured after 24 hrs<br />

and animals with blood glucose levels >250 mg/dL were selected<br />

(Sridevi et al., 2000).<br />

4.9.3.2. Anti-diabetic activity in diabetic rats:<br />

The anti diabetic activity of the prepared topical preparation was<br />

evaluated in overnight fasted diabetic rats.<br />

Diabetic rats were divided into 3 groups (n=5). The rats were treated as<br />

following:<br />

Group I (Control) – 1ml gum acacia suspension was given orally.<br />

Group II - Glz 25 mg/kg was given orally (Stetinova et al.,2007).<br />

Group III –1 gm water soluble ointment base containing certain amount<br />

of Glz - PEG 6000 solid dispersion (10:90) equivalent to 25 mg Glz was<br />

applied on 4cm 2 of the skin.<br />

41


At time intervals between 2-24 h after treatment, blood was collected<br />

from orbital sinuses; blood glucose levels were determined using the<br />

glucometer.<br />

The results obtained from the measurement of blood glucose level by<br />

both glucometer and standard glucose oxidase method were nearly the<br />

same.<br />

4.9.4. Statistical analysis:<br />

The obtained data were compared statistically using One-way<br />

analysis of variance (ANOVA) test of significance on a computer<br />

statistical SPSS analysis program. A p-value of 0.05 or less was<br />

considered to be significant (Suwanpidokkul et al., 2004).<br />

42


1. Partition coefficient of Glz:<br />

Results and Discussion<br />

In the present study, the partition coefficient of Glz was found<br />

to be 1.79 (log octanol/ water =0.25).<br />

2. In vitro release of Glz from different topical formulations:<br />

Glz was chosen to be formulated in topical bases, to demonstrate its<br />

expected action from different topical preparations, such as an ointment,<br />

cream and gel. It is important that the vehicle is able to release the active<br />

ingredient which it carries. Selection of different topical bases as vehicle<br />

for Glz depends on several factors such as polarity, viscosity, and<br />

homogeneity. For this purpose traditional classes of topical bases were<br />

investigated which include water soluble bases, emulsion bases,<br />

absorption bases and gel bases. The emulsion bases included O/W<br />

emulsion and W/O emulsion.<br />

The partition coefficient of the drug is considered as one of the<br />

important parameters for the estimation of the interaction of that drug<br />

with the vehicle and the receiving medium (Celebi et al., 1993).<br />

As general rule in ointment formulations is that, if the drug is held<br />

firmly by the vehicle the rate of the release of the drug is slow (Barr,<br />

1962)<br />

The release of the drug from ointments can be altered by modifying<br />

the composition of the vehicle (Idson, 1983). A greater release of the<br />

drug is expected when there is less affinity of the drug for the base.<br />

(Table 15) and (Figure 34) showed the release of Glz from different<br />

topical bases.<br />

From the data obtained it is clear that the percentage amount of drug<br />

released from water soluble base and gel base are greater than that<br />

released<br />

43


Table 15: In vitro release data of Glz from different topical<br />

Time (min)<br />

bases.<br />

Glz released % ± (sd)<br />

WSB HPMC gel O/W cream<br />

0 0 ± 0 0 ± 0 0 ± 0<br />

30 0 ± 0<br />

60<br />

44<br />

1.35 ± 0.12 0 ± 0<br />

5.70± 0.43 6.71 ± 0.70 0 ± 0<br />

90 14.90 ± 0 11.97 ± 0.74 1.70 ± 0.3<br />

120 22.15 ±0.38 16.80 ± 0.6 2.44 ± 0.3<br />

150 30.46± 0.37 21.41 ± 0.9 2.71 ± 0.4<br />

180 35.4 ± 0.41 25.79 ± 0.57 4.08 ± 0.021<br />

240 47.61± 0.39<br />

300 56.55± 0.07<br />

31.21 ± 0.48 4.90 ± 0.56<br />

37.86 ± 0.9 6.7 ± 0.9<br />

360 62.10 ± 0.42 43.38 ± 2.2 8.43 ± 0.38


WSB HPMC gel O/W cream<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Released<br />

30<br />

20<br />

10<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 34: In vitro release profile of gliclazide from different topical preparation.<br />

45


from other bases. The rate of drug release can be arranged in the<br />

following descending order:<br />

Water soluble base (62.1 %) > HPMC gel (43.38 %) > O/W emulsion<br />

base (8.43 %).<br />

There was no drug release from the absorption base. This may be<br />

attributed to composition of the absorption base which contains white soft<br />

paraffin with several additional lipoidal constituents which favor the<br />

retention of the drug in the base ( Furia, 1972).<br />

Also, there was no drug release from W/O emulsion base. This<br />

finding can be explained on the bases that in case of W/O emulsion bases,<br />

the presence of an oily vehicle as an external phase will result in<br />

formation of an occlusive film on the membrane surface, which will<br />

result in retardation of the permeation of the drug molecules across the<br />

membrane, into the sink solution (Ismail et al., 1990; Khitworth and<br />

Stephenson, 1976).<br />

On the other hand, the higher release of Glz from O/W emulsion<br />

base than from W/O emulsion base may be due to the formation of a<br />

continuous contact between the external phase of the O/W emulsion and<br />

the buffer (Nakano et al., 1971), however, the lower release of the drug<br />

from O/W emulsion base than from water soluble base and HPMC base<br />

may be due to the greater solubility of the drug in the internal oily phase<br />

which may cause a decrease in the rate of release of the drug.<br />

Due to the high lipid solubility of Glz, this may explain the slow<br />

release of the drug that is observed from these bases.<br />

The high diffusion rate of Glz from water soluble base that contains<br />

mainly polyethylene glycol may be due to diffusion of the buffer solution<br />

through the cellophane membrane and formation of water-PEG solution<br />

which increase the solubility and accordingly the rate and extent of Glz<br />

release.<br />

46


The high release of the Glz from HPMC gel is considered to be due<br />

to a high miscibility of this base with the release medium.<br />

3. Viscosity determination:<br />

As shown in (Table 16), water soluble base showed the highest<br />

viscosity followed by O/W cream and finally HPMC gel. It is noted that<br />

all bases included (8:92) PEG 4000 SD have higher viscosity than that<br />

included (1:10) glu SD.<br />

Table 16: Viscosity of different topical bases.<br />

Preparation Viscosity (poise)<br />

(8:92) PEG 4000 SD (1:10) glu SD<br />

WSB .9 2271.4<br />

O/W cream 2251.07 2071.02<br />

HPMC gel 985.90 816.90<br />

4. Effect of incorporation of solid dispersions in different topical<br />

preparations:<br />

(Tables 17-20) and (Figures 35-38) showed that all solid<br />

dispersions increased the overall Glz diffusion by increasing the amount<br />

of diffusible species in the donor phase by enhancing drug solubility.<br />

Therefore, Glz solid dispersions increased the Glz concentration gradient<br />

over the membrane, which resulted in increase in Glz diffusion (Mario et<br />

al., 2005). Incorporation of clotrimazole solid dispersion in O/W cream<br />

improved the antifungal activity of clotrimazol (Madhusudhan et al.,<br />

1999). It was found that formulations containing Rifampicin in the form<br />

of solid dispersion with PEG have shown the best release characteristics<br />

of the antibiotic from oleaginous bases containing Tween 80 (Youssef, et<br />

47


al., 1988). In this study PEG 6000 solid dispersion (8:92) drug to carrier<br />

ratio had the most influential effect on the rate and the extent of release of<br />

Glz , followed by glucose solid dispersion (1:10), PEG 4000 solid<br />

dispersion (8:92) and finally urea solid dispersion (1:10). This is in<br />

agreement with the dissolution results with exception that the dissolution<br />

efficiency of (8:92) PEG 4000 SD (42.23% ± 0.28) was greater than that<br />

of glucose solid dispersion (37.05 ± 0.87) and this may be due to the<br />

higher viscosity of topical bases containing PEG 4000 solid dispersion<br />

than that containing glucose solid dispersion as shown in Table 16. All<br />

data are summarized in Figure 39.<br />

48


Table 17: In vitro release of gliclazide and (8:92) gliclazide-PEG 6000 solid dispersion from different topical<br />

bases.<br />

Gliclazide released % ± (sd)<br />

O/W cream<br />

O/W cream<br />

HPMC gel<br />

HPMC gel<br />

WSB<br />

Time (min) WSB<br />

(SD)<br />

(gliclazide±)<br />

(SD)<br />

(gliclazide)<br />

(SD)<br />

(gliclazide)<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0 ± 0 3.03 ± 0.4 1.35 ± 0.12 6.70 ± 0.28 0 ± 0 1.90 ± 0<br />

60 5.70± 0.43 11.45 ± 1 6.71 ± 0.70 13.02 ± 0.35 0 ± 0 3.30 ± 0<br />

90 14.90 ± 0 19.75 ± 0.4 11.97 ± 0.74 19.48 ± 0.05 1.70 ± 0.3 4.35 ± 0.4<br />

120 22.15 ±0.38 27.30 ± 0.28 16.80 ± 0.6 25.41 ± 0.21 2.44 ± 0.3 7.16 ± 0.48<br />

150 30.46± 0.37 34.22 ± 0.11 21.41 ± 0.9 31.16 ± 0.98 2.71 ± 0.4 8.38 ± 0.44<br />

180 35.4 ± 0.41 39.20 ± 0.24 25.79 ± 0.57 39.62± 0.84 4.08 ± 0.021 8.80 ± 0.59<br />

240 47.61± 0.39 51.50 ± 1.4 31.21 ± 0.48 44.47 ± 1.2 4.90 ± 0.56 10.37 ± 0.63<br />

300 56.55± 0.07 60.90 ± 1.6 37.86 ± 0.9 51.47 ± 1.5 6.7 ± 0.9 14.4 ± 0.62<br />

360 62.10 ± 0.42 69.96 ± 2.5 43.38 ± 2.2 60.47 ± 1.5 8.43 ± 0.38 16.80 ± 0.37<br />

% Increase ------- 11.23 ------- 28.26 ------- 99.28<br />

49


WSB- glic WSB-SD HPMC gel-glic<br />

HPMC gel- SD O/W cream-glic O/W cream- SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Released<br />

30<br />

20<br />

10<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 35: In vitro release profile of gliclazide and (8:92) gliclazide –PEG 6000 solid dispersion from<br />

different topical bases.<br />

50


Table 18: In vitro release of gliclazide and (1:10) gliclazide-glucose solid dispersion from different topical<br />

Gliclazide released % ± (sd)<br />

O/W cream<br />

O/W cream<br />

HPMC gel<br />

HPMC gel<br />

WSB<br />

Time (min) WSB<br />

(SD)<br />

(gliclazide±)<br />

(SD)<br />

(gliclazide)<br />

(SD)<br />

(gliclazide)<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0 ± 0 1.14 ± 0.1 1.35 ± 0.12 6.3 ± 0.28 0 ± 0 1.60± 0.24<br />

60 5.70± 0.43 9.68 ± 0.7 6.71 ± 0.70 12.4 ± 0.35 0 ± 0 2.06 ± 0.4<br />

90 14.90 ± 0 17.88 ± 0.97 11.97 ± 0.74 18.72 ± 0.05 1.70 ± 0.3 3.16 ± 0.7<br />

120 22.15 ±0.38 25.96 ± 0.8 16.80 ± 0.6 21.90 ± 0.20 2.44 ± 0.3 5.8 ± 0.7<br />

150 30.46± 0.37 33.43 ± 1.2 21.41 ± 0.9 27.47 ± 0.98 2.71 ± 0.4 7.4 ± 1.1<br />

180 35.4 ± 0.41 37.90 ± 1.5 25.79 ± 0.57 34.04± 0.84 4.08 ± 0.021 8.65 ± 0.7<br />

240 47.61± 0.39 48.73 ± 1.5 31.21 ± 0.48 39.24 ± 1.2 4.90 ± 0.56 9.1± 0.37<br />

300 56.55± 0.07 58.5± 0.96 37.86 ± 0.9 45.89 ± 1.5 6.7 ± 0.9 12.12 ± 0.9<br />

360 62.10 ± 0.42 67.22 ± 1.1 43.38 ± 2.2 55.69 ± 1.5 8.43 ± 0.38 14.77 ± 1.0<br />

% Increase ------ 7.61 ----- 22.1 ----- 42.92<br />

51


WSB-glic WSB-SD HPMC gel- glic<br />

HPMC gel-SD O/W cream- glic O/W cream- SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Released<br />

30<br />

20<br />

10<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 36: In vitro release profile of gliclazide and (1:10) gliclazide –glucose solid dispersion from different topical<br />

bases.<br />

52


Table 19: In vitro release of gliclazide and (8:92) gliclazide-PEG 4000 solid dispersion from different topical<br />

bases.<br />

Gliclazide released % ± (sd)<br />

O/W cream<br />

O/W cream<br />

HPMC gel<br />

HPMC gel<br />

WSB<br />

Time (min) WSB<br />

(SD)<br />

(gliclazide±)<br />

(SD)<br />

(gliclazide)<br />

(SD)<br />

(gliclazide)<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0 ± 0 1.4 ± 0.17 1.35 ± 0.12 2.55 ± 0 0 ± 0 1.20± 0.2<br />

60 5.70± 0.43 8.15 ± 0.21 6.71 ± 0.70 8.98 ± 0.12 0 ± 0 1.64 ± 0.13<br />

90 14.90 ± 0 18.40 ± 0.9 11.97 ± 0.74 18.06 ± 0.35 1.70 ± 0.3 2.42 ± 0.22<br />

120 22.15 ±0.38 24.71 ± 0.7 16.80 ± 0.6 23.95 ± 0.43 2.44 ± 0.3 2.84 ± 0.2<br />

150 30.46± 0.37 32.71 ± 1.2 21.41 ± 0.9 27.7 ± 0.43 2.71 ± 0.4 4.44 ± 0.5<br />

180 35.4 ± 0.41 38.26 ± 0.08 25.79 ± 0.57 33.6± 0.58 4.08 ± 0.021 5.5 ± 0.35<br />

240 47.61± 0.39 48.48 ± 1.02 31.21 ± 0.48 38.1 ± 1.9 4.90 ± 0.56 8.16± 0.57<br />

300 56.55± 0.07 58.89 ± 1.5 37.86 ± 0.9 44.7 ± 1.6 6.7 ± 0.9 10.99 ± 1.03<br />

360 62.10 ± 0.42 65.1 ± 1.8 43.38 ± 2.2 53.7 ± 1.9 8.43 ± 0.38 11.71 ± 0.1<br />

% Increase -------- 4.60 ------- 19.21 ----- 28.01<br />

53


WSB- glic WSB-SD HPMC gel- glic<br />

HPMC gel- SD O/W cream-glic O/W cream-SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Released<br />

30<br />

20<br />

10<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 37: In<br />

vitro release profile of gliclazide and (8:92) gliclazide –PEG 4000 solid dispersion from<br />

different topical bases.<br />

54


Table 20: In vitro release of gliclazide and (1:10) gliclazide-urea solid dispersion from different topica<br />

bases.<br />

Gliclazide released % ± (sd)<br />

Time (min) WSB WSB HPMC gel HPMC gel O/W cream O/W cream<br />

(gliclazide) (SD) (gliclazide) (SD) (gliclazide±) (SD)<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0 ± 0 1.54 ± 0.13 1.35 ± 0.12 3.92 ± 0.18 0 ± 0 0± 0<br />

60 5.70± 0.43 8.07 ± 0.47 6.71 ± 0.70 10.30 ± 0.30 0 ± 0 0.99 ± 0<br />

90 14.90 ± 0 16.98 ± 0.37 11.97 ± 0.74 15.50 ± 0.27 1.70 ± 0.3 2.28± 0.59<br />

120 22.15 ±0.38 24.37 ± 0.48 16.80 ± 0.6 21.05 ± 0.07 2.44 ± 0.3 3.44 ± 0.63<br />

150 30.46± 0.37 31.14 ± 0.18 21.41 ± 0.9 25.1 ± 0.56 2.71 ± 0.4 4.21 ± 0.44<br />

180 35.4 ± 0.41 37.6± 1.8 25.79 ± 0.57 29.94± 0.06 4.08 ± 0.021 5.89 ± 0.62<br />

240 47.61± 0.39 47.25 ± 1.1 31.21 ± 0.48 35.15 ± 0.49 4.90 ± 0.56 7.20± 0.48<br />

300 56.55± 0.07 57.37± 1.03 37.86 ± 0.9 41.80 ± 0.71 6.7 ± 0.9 8.80 ± 0.37<br />

360 62.10 ± 0.42 64.15 ± 1.4 43.38 ± 2.2 52.41 ± 1.1 8.43 ± 0.38 10.74 ± 0.4<br />

% Increase ------ 3.19 ------- 17.22 ------- 21.5<br />

55


WSB- glic WSB- SD HPMC gel -glic<br />

HPMC gel - SD O/W cream- glic O/W cream -SD<br />

100<br />

90<br />

80<br />

70<br />

60<br />

50<br />

40<br />

% Released<br />

30<br />

20<br />

10<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure<br />

38: In vitro release profile of gliclazide and (1:10) gliclazide –urea solid dispersion from different topical bases.<br />

56


57<br />

1 W<br />

2 HPMC<br />

3 O/W cr


80<br />

PEG 6000<br />

glucose<br />

PEG 4000<br />

70<br />

urea<br />

PEG 6000<br />

glucose<br />

PEG 4000<br />

Drug<br />

60<br />

urea<br />

Drug alone<br />

(8:92) PEG 6000 SD<br />

(1:10) glucose SD<br />

(8:92) PEG 4000 SD<br />

(1:10) urea SD<br />

50<br />

Drug<br />

40<br />

% Released<br />

30<br />

PEG 6000<br />

glucose<br />

PEG 4000<br />

20<br />

urea<br />

Drug<br />

10<br />

0<br />

1 2 3<br />

Topical bases<br />

Figure 39: Release of gliclazide from different bases with different solid dispersions.<br />

58


5. Kinetic analysis of release data:<br />

As shown in (Table 21) the data of Glz and solid dispersions<br />

released from different topical formulations followed first order kinetics<br />

while that obtained from HPMC gel followed diffusion controlled<br />

mechanism or Higuchi model.<br />

Table 21: Kinetic data of the release of Glz and solid<br />

WSB<br />

HPMC gel<br />

O/W cream<br />

dispersions from different topical bases.<br />

Topical<br />

Correlation coefficient (R) Observed<br />

preparation Zero First Diffusion order<br />

Drug 0.9883 0.9987 0.9965 First<br />

(8:92) Glz- PEG<br />

6000 SD<br />

0.9933 0.9984 0.9982 First<br />

(8:92) Glz- PEG<br />

4000 SD<br />

0.9889 0.9993 0.9981 First<br />

(1:10) Glz-glu SD 0.9912 0.9990 0.9987 First<br />

(1:10) Glz-UR<br />

SD<br />

0.9906 0.9996 0.9980 First<br />

Drug 0.9908 0.9981 0.9987 D.M<br />

(8:92) Glz- PEG<br />

6000 SD<br />

0.9884 0.9959 0.9963 D.M<br />

(8:92) Glz- PEG<br />

4000 SD<br />

0.9904 0.9959 0.9967 D.M<br />

(1:10) Glz-glu SD 0.9863 0.9949 0.9965 D.M<br />

(1:10) Glz-UR<br />

SD<br />

0.9930 0.9941 0.9945 D.M<br />

Drug 0.9931 0.9933 0.9810 First<br />

(8:92) Glz- PEG<br />

6000 SD<br />

0.9912 0.9915 0.9837 First<br />

(8:92) Glz- PEG<br />

4000 SD<br />

0.9897 0.9899 0.9654 First<br />

(1:10) Glz-glu SD 0.9857 0.9870 0.9815 First<br />

(1:10) Glz-UR<br />

SD<br />

0.9957 0.9967 0.9935 First<br />

59


6. In vitro permeation of Glz through abdominal rabbit skin:<br />

Skin permeation studies indicated that Glz permeation through<br />

hairless rabbit skin was negligible. The possible reasons for this result<br />

may be i) Glz , a lipophilic drug, was retained within the stratum corneum<br />

with no partioning into the viable epidermis or ii) most of the drug was<br />

used up to saturate the binding sites in the skin and the remaining drug<br />

was probably insufficient to provide a significant concentration gradient<br />

(Srini et al., 1998).<br />

7. In vivo study:<br />

The result of hypoglycemic activity of the topically applied<br />

gliclazide and oral gliclazide (25 mg/kg; p.o.) in both normal and diabetic<br />

rats are shown in (Table 22-23) and (Figure 40-41).<br />

*** Studies in normal rats<br />

Gliclazide (oral) produced a significant decrease of 60.64 % ± 6.3<br />

(plevels at 2 hr and then the<br />

blood glucose levels decreased. The percentage reduction in the blood<br />

glucose levels at the end of 24 hr were only 24.83 ± 2.05. On other hand,<br />

the blood glucose reducing response of gliclazide (topical) was gradual<br />

and significant upto 24 h compared to control (p <br />

blood glucose reducing response was observed after 6 hr and thereafter<br />

remained stable up to 24 h. These results are in accordance with the<br />

results obtained by (Mutalik and Udupa, 2005).<br />

As shown in (Figure 40), the blood glucose reducing activity of<br />

ointment contained (10:90) gliclazide –PEG 6000 solid dispersions was<br />

significantly more when compared to ointment contained gliclazide alone.<br />

This is in agreement with the results of Madhusudhan et al., 1999 who<br />

60


found that Incorporation of clotrimazole solid dispersion in O/W cream<br />

improved the antifungal activity of clotrimazol.<br />

Topical route effectively maintained normoglycemic level in<br />

contrast to the oral group which produced remarkable hypoglycemia.<br />

61


Table 22 : Reduction in blood glucose level after oral and topical application of gliclazide and 10:90 gliclazide- PEG<br />

6000 solid dispersion in normal rats. All values are expressed as mean ± sd.<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Absolute<br />

blood glucose<br />

level (mg/dl)<br />

Group<br />

2hr 4hr 6hr 8hr 24hr<br />

73.36 ± 9.89<br />

(16.6 ± 2.09)<br />

75.06 ± 8.4<br />

(15.46 ±2.45)<br />

76.1 ± 9.2<br />

(13.44 ±1.44)<br />

76.45 ± 5.2<br />

(12.04 ± 0.99)<br />

88.72 ± 8.6 82.33 ± 7.7*<br />

(7.14±1.25)**<br />

Control<br />

(1ml gum acacia<br />

suspension)<br />

67.66 ± 4.1<br />

(24.83 ±2.05)<br />

48.5 ± 6.2<br />

(46.89 ±4.93)<br />

43.3 ± 4.64<br />

(51.85 ± 3.5)<br />

39.97 ± 5.3<br />

(55.99 ±4.41)<br />

89.85 ± 5.01 35.27 ± 6.07<br />

(60.64 ± 6.3)<br />

Oral gliclazide<br />

(25mg/kg)<br />

56.29 ± 1.4<br />

(32.71 ± 6.9)<br />

52.18 ± 3.8<br />

(34.34 ± 4.8)<br />

47.92 ± 5.1<br />

(40.05 ± 5.4)<br />

58.4 ± 2.2<br />

(30.14 ± 5.9)<br />

81.59 ± 8.9 58.22 ± 4.6<br />

(28.19 ± 4.5)<br />

WSB<br />

(Gliclazide)<br />

51.13 ± 3.5<br />

(37.45 ± 3.6)<br />

45.38 ± 3.5<br />

(44.66 ±4.51)<br />

38.46 ± 4.6<br />

(52.62 ± 6.7)<br />

47.05 ± 2.26<br />

(43.5 ± 3.05)<br />

82.14 ± 3.8 62.00 ± 3.2<br />

(25.5 ± 4.03)<br />

WSB<br />

(10:90 gliclazide-<br />

PEG 6000 SD)<br />

* Reduction in blood glucose level (mg/dl). ** Percentage reduction in blood glucose levels.<br />

62


80<br />

Control oral gliclazide WSB ( gliclazide) WSB (gliclazide-PEG 6000 SD)<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

% Reduction in blood glucose level(mg/dl)<br />

10<br />

0<br />

0 2 4 6 8 10 12 14 16 18 20 22 24 26<br />

Time (hr)<br />

Figure 40: Percent reduction in blood glucose levels after oral and topical administration of gliclazide in normal rats.<br />

63


*** Studies in diabetic rats:<br />

Results obtained from the diabetic rats after application of ointment<br />

base containing certain amount of Glz - PEG 6000 solid dispersion<br />

(10:90) equivalent to 25 mg Glz and oral gliclazide administration are<br />

shown in (Figure 41) and (Table 23).<br />

Oral and topical groups showed significant hypoglycemic activity up<br />

to 24 h (p <br />

effect produced by the topical gliclazide was significantly less when<br />

compared to oral administration. The topical and the oral drug produced a<br />

decrease of 36.35 % ± 4.42 and 21.33 % ± 3.73 respectively, in the blood<br />

glucose level after 24 h.<br />

Studies in diabetic rats showed small difference in the duration of<br />

action between the oral and topical groups and this may be due to reduced<br />

insulin level in diabetic models which impairs the principal metabolic<br />

pathways of sulphonylurea which resulted in its prolonged action in<br />

orally treated group (Strove and Belkina, 1989).<br />

These results are in accordance with the results obtained by Sridevi<br />

et al., 2000 who stated that the hypoglycemic activity of oral and topical<br />

groups did not differ significantly in the two groups after 8 hrs. The<br />

TDDS and the oral drug produced decrease of 61.9 ± 9.5% and 63.4 ±<br />

3.3% respectively, in the blood glucose levels after 24 hrs.<br />

Finally, the slow and sustained release of the drug from the<br />

transdermal system might reduce manifestations like severe<br />

hypoglycemia, sulphonylurea receptor down regulation and the risk of<br />

chronic hyperinsulinemia (Faber et al., 1990 and Bitzen et al., 1992).<br />

64


Table 23 : Reduction in blood glucose level after oral and topical application of gliclazide and 10:90 gliclazide- PEG<br />

6000 solid dispersion in diabetic rats. All values are expressed as mean ± sd.<br />

.<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Absolute<br />

blood glucose<br />

level (mg/dl)<br />

Group<br />

2hr 4hr 6hr 8hr 24hr<br />

223.4 ± 40.8<br />

(5.22 ± 2.74)<br />

224.4 ± 38.8<br />

(4.76±0.89)<br />

230.6 ± 40.8<br />

(2.16 ±1.68)<br />

232.2 ± 38.46<br />

(1.35 ± 1.06)<br />

235.6 ± 40.3 228.4± 36.26*<br />

(2.89±1.65)**<br />

Control<br />

(1ml gum acacia<br />

suspension)<br />

365.6 ± 13.63<br />

(36.35 ±4.42)<br />

316.6 ± 32.5<br />

(44.84 ±7.03)<br />

293.8 ± 43.11<br />

(48.88 ± 4.9)<br />

327.2 ± 41.5<br />

(43.18 ±6.8)<br />

577 ± 48.16 355.1 ± 55.32<br />

(38.28 ± 5.57)<br />

Oral gliclazide<br />

(25mg/kg)<br />

254 ± 27.21<br />

(21.33 ± 3.73)<br />

263.2 ± 34.98<br />

(18.69 ±3.08)<br />

284.2 ± 37.7<br />

(12.53± 1.47)<br />

293 ± 43.73<br />

(9.7 ± 1.14)<br />

324.6 ± 47 302.4 ± 44.89<br />

(6.78 ± 2.2)<br />

WSB<br />

(10:90 gliclazide-<br />

PEG 6000 SD)<br />

* Reduction in blood glucose level (mg/dl). ** Percentage reduction in blood glucose levels.<br />

65


Control Oral Glz Topical Glz<br />

60<br />

50<br />

40<br />

30<br />

20<br />

10<br />

% Reduction in the blood glucose level (mg/dl)<br />

0<br />

0 2 4 6 8 10 12 14 16 18 20 22 24 26<br />

Time (hr)<br />

Figure 41: Percent reduction in blood glucose levels after oral and topical administration of gliclazide in diabetic<br />

rats.<br />

66


Conclusion:<br />

From the previously demonstrated data the following results can be<br />

concluded:<br />

1- Glz has a lipophilic property.<br />

2- The amount of Glz released from water soluble base (PEG base) and<br />

HPMC gel base was found to be higher than that from other bases.<br />

3- The amount of Glz released from O/W emulsion base was greater than<br />

that released from W/O emulsion base.<br />

4- No drug is released from the absorption base and W/O emulsion base.<br />

5- The investigation showed the effect of incorporation of Glz solid<br />

dispersions in different carriers such as PEG 4000, PEG 6000, glucose<br />

and urea on the amount of Glz released from different topical bases<br />

which can be summarized as follows in descending order:<br />

(8:92) Glz-PEG 6000 SD > (1:10) Glz-glu SD > (8:92) Glz – PEG<br />

4000 SD > Glz- UR SD.<br />

6- The present study showed that gliclazide was absorbed through the<br />

skin and lowered the blood glucose levels.<br />

Topical preparations of Glz or its solid dispersions exhibited better<br />

control of blood glucose level than oral Glz administration in rats as<br />

topical route effectively maintained normoglycemic level in contrast to<br />

the oral group which produced remarkable hypoglycemia.<br />

The blood glucose reducing activity of ointment contained (10:90)<br />

gliclazide –PEG 6000 solid dispersions was significantly more when<br />

compared to ointment contained gliclazide alone.<br />

Finally, the slow and sustained release of the drug from the<br />

transdermal system might reduce manifestations like severe<br />

hypoglycemia, sulphonylurea receptor down regulation and the risk of<br />

chronic hyperinsulinemia.<br />

67


1. Description<br />

Introduction<br />

Glibenclamide<br />

1.1 Name, formula, molecular weight<br />

Glib is 1-{4-[2-(5-chloro-2-methoxybenzamido) ethyl]<br />

benzenesulphonyl}-3-cyclohexylurea<br />

Figure 43: Glib structure.<br />

C23 H28 Cl N3 O5 S<br />

Molecular Weight = 494.0<br />

1.2 Appearance, odour, colour:<br />

Glib is a white, crystalline, odourless powder and practically without<br />

taste (Pamela, 1981).<br />

2. Physical properties<br />

2.1 Melting point<br />

172° to 174°<br />

2.2 Solubility<br />

Glib is virtually insoluble in water and ether; soluble in 330 parts of<br />

alcohol, in 36 parts of chloroform, and in 250 parts of methanol (Pamela,<br />

1981).<br />

69


3.Pharmacokinetics:<br />

Glib is readily absorbed from the gastrointestinal tract, peak plasma<br />

concentrations usually occurring within 2 to 4 hours, and is extensively<br />

bound to plasma proteins. Absorption may be slower in hyperglycaemic<br />

patients and may differ according to the particle size of the preparation<br />

used. It is metabolised, almost completely, in the liver, the principal<br />

metabolite being only very weakly active. About 50% of a dose is<br />

excreted in the urine and 50% via the bile into the faeces (Martindale,<br />

1996) .<br />

4.Mode of action:<br />

As mentioned before under sulfonylureas.<br />

5. Uses and Administration:<br />

Glib is a sulfonylurea antidiabetic. It is given by mouth in the<br />

treatment of type 2 diabetes mellitus and has a duration of action of up to<br />

24 hours.<br />

The usual initial dose of conventional formulations in type 2<br />

diabetes mellitus is 2.5 to 5 mg daily with breakfast, adjusted every 7<br />

days by increments of 2.5 or 5 mg daily up to 15 mg daily. Although<br />

increasing the dose above 15 mg is unlikely to produce further benefit,<br />

doses of up to 20 mg daily have been given. Doses greater than 10 mg<br />

daily may be given in 2 divided doses. Because of the relatively long<br />

duration of action of Glib, it is best avoided in the elderly (Martindale,<br />

1996) .<br />

70


6. Precautions:<br />

As mentioned before under sulfonylureas.<br />

7. Adverse Effects:<br />

As mentioned before under sulfonylureas.<br />

8. Interactions:<br />

As mentioned before under sulfonylureas.<br />

9. Adverse Effects and Precautions<br />

As mentioned before under sulfonylureas.<br />

10. Methods of analysis:<br />

10.1. Polarography:<br />

Procedures have been described for quantitative work, an automated<br />

system, having a flow through micro cell used with silver- silver chloride<br />

reference electrode, has been stated to give good reproducibility (Pamela,<br />

1981).<br />

10.2. Non-aqueous titration:<br />

Tetramethylurea has been used as solvent for the titration of Glib<br />

with 0.1 normal lithium methoxide in benzene-methanol. The end point<br />

was determined potentiometrically or by using 0.2% azoviolet in toluene<br />

as visual indicator (Pamela, 1981).<br />

10.3. Chromatography:<br />

Several procedures have been proposed for the identification of Glib<br />

by thin-layer chromatography. Among the solvent systems described are<br />

butanol-methanol-chloroform-25% ammonia, propanol-cyclohexane and<br />

propanol-benzene-cyclohexane.<br />

71


High-perfprmance liquid chromatography has been recommended for<br />

quantitative determination of Glib in tablets. The column packing uesd<br />

was 1% ethylene propylene copolymer on DuPont Zipax, with 0.01 M<br />

sodium borate containing 27.5% v/v methanol as mobile phase.<br />

Testosterone serves as internal standard (Pamela, 1981).<br />

72


Introduction<br />

There is considerable interest in the skin as a site of drug application<br />

both for local and systemic effect. However, the skin, in particular the<br />

stratum corneum, poses a formidable barrier to drug penetration thereby<br />

limiting topical and transdermal bioavailability. Skin penetration<br />

enhancement techniques have been developed to improve bioavailability<br />

and increase the range of drugs for which topical and transdermal<br />

delivery is a viable option (Heather, 2005).<br />

Drug permeation across the stratum corneum obeys Fick’s first law<br />

(equation 1) where steady-state flux (J) is related to the diffusion<br />

coefficient (D) of the drug in the stratum corneum over a diffusional path<br />

length or membrane thickness (h), the partition coefficient (P) between<br />

the stratum corneum and the vehicle, and the applied drug concentration<br />

(C0) which is assumed to be constant:<br />

1)<br />

73<br />

dm/dt = J = D C0 P/ h<br />

(Equation<br />

Equation 1 aids in identifying the ideal parameters for drug diffusion<br />

across the skin. The influence of solubility and partition coefficient of a<br />

drug on diffusion across the stratum corneum has been extensively<br />

studied. Molecules showing intermediate partition coefficients (log P<br />

octanol/water of 1-3) have adequate solubility within the lipid domains of<br />

the stratum corneum to permit diffusion through this domain whilst still<br />

having sufficient hydrophilic nature to allow partitioning into the viable<br />

tissues of the epidermis. The maximum permeability measurement being<br />

attained at log P value 2.5, which is typical of these types of experiments.


Optimal permeability has been shown to be related to low molecular size<br />

(Potts and Guy, 1992) (ideally less than 500 Da (Bos and Meinardi,<br />

2000)) as this affects diffusion coefficient, and low melting point which is<br />

related to solubility. When a drug possesses these ideal characteristics (as<br />

in the case of nicotine and nitroglycerin), transdermal delivery is feasible.<br />

However, where a drug does not possess ideal physicochemical<br />

properties, manipulation of the drug or vehicle to enhance diffusion,<br />

becomes necessary. The approaches that have been investigated are<br />

summarised in (Figure 42) and discussed below.<br />

Figure 42: Techniques to optimize drug permeation across the skin.<br />

1. Penetration enhancement through optimization of drug and<br />

vehicle properties:<br />

1.1. Prodrugs and ion-pairs:<br />

74


The prodrug approach has been investigated to enhance dermal and<br />

transdermal delivery of drugs with unfavourable partition coefficients<br />

(Sloan, 1992; Sloan and Wasdo, 2003). The prodrug design strategy<br />

generally involves addition of a promoiety to increase partition<br />

coefficient and hence solubility and transport of the parent drug in the<br />

stratum corneum. Upon reaching the viable epidermis, esterases release<br />

the parent drug by hydrolysis thereby optimising solubility in the aqueous<br />

epidermis. The prodrug approach has been investigated for increasing<br />

skin permeability of non-steroidal anti-inflammatory drugs (Davaran et<br />

al., 2003; Thorsteinsson et al., 1999), naltrexone (Stinchcomb et al.,<br />

2002)<br />

Charged drug molecules do not readily partition into or permeate<br />

through human skin. Formation of lipophilic ionpairs has been<br />

investigated to increase stratum corneum penetration of charged species.<br />

This strategy involves adding an oppositely charged species to the<br />

charged drug, forming an ion-pair in which the charges are neutralised so<br />

that the complex can partition into and permeate through the stratum<br />

corneum. The ion-pair then dissociates in the aqueous viable epidermis<br />

releasing the parent charged drug which can diffuse within the epidermal<br />

and dermal tissues. (Megwa et al., 2000; Valenta et al., 2000).<br />

(Sarveiya et al., 2004) recently reported a 16-fold increase in the steady-<br />

state flux of ibuprofen ionpairs across a lipophilic membrane.<br />

1.2. Chemical potential of drug in vehicle – saturated and<br />

supersaturated solutions:<br />

The maximum skin penetration rate is obtained when a drug is at its<br />

highest thermodynamic activity as is the case in a supersaturated solution.<br />

Supersaturated solutions can occur due to evaporation of solvent or by<br />

mixing of cosolvents. These systems are inherently unstable and require<br />

75


the incorporation of antinucleating agents to improve stability (Heather,<br />

2005).<br />

1.3. Eutectic Systems:<br />

As previously described, the melting point of drug influences<br />

solubility and hence skin penetration. According to regular solution<br />

theory, the lower the melting point, the greater the solubility of a material<br />

in a given solvent, including skin lipids. The melting point of a drug<br />

delivery system can be lowered by formation of a eutectic mixture: a<br />

mixture of two components which, at a certain ratio, inhibit the<br />

crystalline process of each other, such that the melting point of the two<br />

components in the mixture is less than that of each component alone.<br />

EMLA cream, a formulation consisting of a eutectic mixture of<br />

lignocaine and prilocaine applied under an occlusive film, provides<br />

effective local anaesthesia for pain-free venepuncture and other<br />

procedures (Ehrenstrom and Reiz, 1982).<br />

1.4. Complexes:<br />

Complexation of drugs with cyclodextrins has been used to enhance<br />

aqueous solubility and drug stability. Cyclodextrin has a hydrophilic<br />

exterior and lipophilic core in which appropriately sized organic<br />

molecules can form non-covalent inclusion complexes resulting in<br />

increased aqueous solubility and chemical stability (Loftsson and<br />

Brewster, 1996). As flux is proportional to the free drug concentration,<br />

where the cyclodextrin concentration is sufficient to complex only the<br />

drug which is in excess of its solubility, an increase in flux might be<br />

expected. However, at higher cyclodextrin concentrations, the excess<br />

76


cyclodextrin would be expected to complex free drug and hence reduce<br />

flux. Skin penetration enhancement has also been attributed to extraction<br />

of stratum corneum lipids by cyclodextrins (Bentley et al., 1997).<br />

1.5. Liposomes and Vesicles:<br />

A variety of encapsulating systems have been evaluated including<br />

liposomes, deformable liposomes or transfersomes, ethosomes and<br />

niosomes.<br />

Liposomes are colloidal particles formed as concentric biomolecular<br />

layers that are capable of encapsulating drugs. The skin delivery of<br />

triamcinolone acetonide was four to five times greater from a liposomal<br />

lotion than an ointment containing the same drug concentration (Mezei<br />

and Gulasekharam, 1980). The mechanism of enhanced drug uptake<br />

into the stratum corneum is unclear. It is possible that the liposomes<br />

either penetrate the stratum corneum to some extent then interact with the<br />

skin lipids to release their drug or that only their components enter the<br />

stratum corneum. It is interesting that the most effective liposomes are<br />

reported to be those composed of lipids similar to stratum corneum lipids<br />

(Egbaria et al., 1990), which are likely to most readily enter stratum<br />

corneum lipid lamellae and fuse with endogenous lipids.<br />

Transfersomes are vesicles composed of phospholipids as their<br />

main ingredient with 10-25% surfactant (such as sodium cholate) and 3-<br />

10% ethanol. The surfactant molecules act as “edge activators”,<br />

conferring ultradeformability on the transfersomes, which reportedly<br />

allows them to squeeze through channels in the stratum corneum that are<br />

less than one-tenth the diameter of the transfersome (Cevc, 1996).<br />

77


Ethosomes are liposomes with a high alcohol content capable of<br />

enhancing penetration to deep tissues and the systemic circulation (Biana<br />

and Touitou, 2003; Touitou et al., 2000).<br />

Niosomes are vesicles composed of nonionic surfactants that have<br />

been evaluated as carriers for a number of drug and cosmetic applications<br />

(Shahiwala and Misra, 2002; Sentjurc et al., 1999). This area continues<br />

to develop with further evaluation of current formulations and reports of<br />

other vesicle forming materials.<br />

1.6. Solid lipid Nanoparticles:<br />

Solid lipid nanoparticles (SLN) have recently been investigated as<br />

carriers for enhanced skin delivery of sunscreens, vitamins A and E,<br />

triptolide and glucocorticoids (Santos Maia et al., 2002; Mei et al.,<br />

2003). It is thought their enhanced skin penetration is primarily due to an<br />

increase in skin hydration caused by the occlusive film formed on the<br />

skin surface by the SLN.<br />

2. Penetration enhancement by stratum cornium modification:<br />

2.1. Hydration:<br />

Water is the most widely used and safest method to increase skin<br />

penetration of both hydrophilic (Behl et al., 1980) and lipophilic<br />

permeants (McKenzie and Stoughton, 1962). The water content of the<br />

stratum corneum is around 15 to 20% of the dry weight Additional water<br />

within the stratum corneum could alter permeant solubility and thereby<br />

modify partitioning from the vehicle into the membrane. In addition,<br />

increased skin hydration may swell and open the structure of the stratum<br />

corneum leading to an increase in penetration. Hydration can be increased<br />

by occlusion with plastic films; paraffins, oils, waxes as components of<br />

ointments and water-in-oil emulsions that prevent transepidermal water<br />

78


loss; and oil-in-water emulsions that donate water. A commercial<br />

example of this is the use of an occlusive dressing to enhance skin<br />

penetration of lignocaine and prilocane from EMLA cream in order to<br />

provide sufficient local anaesthesia within about 1 hour.<br />

2.2. Penetration enhancers:<br />

They are chemicals that interact with skin constituents to promote<br />

drug flux. To-date, a vast array of chemicals has been evaluated as<br />

penetration enhancers (or absorption promoters). Properties for<br />

penetration enhancers acting within skin have been given by Barry, 1983<br />

as follows:<br />

• They should be non-toxic, non-irritating and non-allergenic.<br />

• They would ideally work rapidly, and the activity and duration of effect<br />

should be both predictable and reproducible.<br />

• They should have no pharmacological activity within the body—i.e.<br />

should not bind to receptor sites.<br />

• The penetration enhancers should work unidirectionally, i.e. should<br />

allow therapeutic agents into the body whilst preventing the loss of<br />

endogenous material from the body.<br />

• When removed from the skin, barrier properties should return both<br />

rapidly and fully.<br />

• The penetration enhancers should be appropriate for formulation into<br />

diverse topical preparations, thus should be compatible with both<br />

excipients and drugs.<br />

• They should be cosmetically acceptable with an appropriate skin ‘feel’.<br />

79


2.2.1. Sulphoxides and similar chemicals:<br />

Dimethylsulphoxide (DMSO) is one of the earliest and most widely<br />

studied penetration enhancers. It is a powerful aprotic solvent which<br />

hydrogen bonds with itself rather than with water. it has been shown to<br />

promote the permeation of, for example, antiviral agents, steroids and<br />

antibiotics (Wiiliam and Barry, 2004).<br />

Although DMSO is an excellent accelerant it does create problems.<br />

The effects of the enhancer are concentration dependent and generally co-<br />

solvents containing >60% DMSO are needed for optimum enhancement<br />

efficacy. However, at these relatively high concentrations DMSO can<br />

cause erythema and wheals of the stratum corneum and may denature<br />

some proteins. Studies performed over 40 years ago on healthy volunteers<br />

painted with 90% DMSO twice daily for 3 weeks resulted in erythema,<br />

scaling, contact urticaria, stinging and burning sensations and several<br />

volunteers developed systemic symptoms (Kligman, 1965). A further<br />

problem with DMSO use as a penetration enhancer is the metabolite<br />

dimethylsulphide produced from the solvent; dimethylsulphide produces<br />

a foul odour on the breath.<br />

Since DMSO is problematic for use as a penetration enhancer,<br />

researchers have investigated similar, chemically related materials as<br />

accelerants. Dimethylacetamide (DMAC) and dimethylformamide (DMF)<br />

are similarly powerful aprotic solvents with structures akin to that of<br />

DMSO. Also in common with DMSO, both solvents have a broad range<br />

of penetration enhancing activities.<br />

The mechanisms of the sulphoxide penetration enhancers and<br />

DMSO in particular, are complex. DMSO is widely used to denature<br />

proteins and on application to human skin has been shown to change the<br />

intercellular keratin confirmation. DMSO has also been shown to interact<br />

80


with the intercellular lipid domains of human stratum corneum. Further,<br />

DMSO within skin membranes may facilitate drug partitioning from a<br />

formulation into this “universal solvent” within the tissue.<br />

2.2.2. Azone:<br />

Azone was the first molecule specifically designed as a skin<br />

penetration enhancer. The chemical has low irritancy, very low toxicity<br />

(oral LD50 in rat of 9 g/kg) and little pharmacological activity although<br />

some evidence exists for an antiviral effect. Thus, judging from the<br />

above, Azone appears to possess many of the desirable qualities listed for<br />

a penetration enhancer.<br />

Azone enhances the skin transport of a wide variety of drugs<br />

including steroids, antibiotics and antiviral agents. As with many<br />

penetration enhancers, the efficacy of azone appears strongly<br />

concentration dependent and is also influenced by the choice of vehicle<br />

from which it is applied. Surprisingly, Azone is most effective at low<br />

concentrations, being employed typically between 0.1% and 5%, often<br />

between 1% and 3%.<br />

Azone probably exerts its penetration enhancing effects through<br />

interactions with the lipid domains of the stratum corneum.<br />

Singh et al., 1993 reported that ephedrine patches containing azone<br />

showed an increased flux of ephedrine through rat skin and epidermis<br />

with a reduced time lag.<br />

2.2.3. Pyrrolidones:<br />

A range of pyrrolidones and structurally related compounds have<br />

been investigated as potential penetration enhancers in human skin. They<br />

apparently have greater effects on hydrophilic permeants than for<br />

lipophilic materials. N-methyl-2-pyrrolidone (NMP) and 2-pyrrolidone<br />

(2P) are the most widely studied enhancers of this group.<br />

81


Pyrrolidones have been used as permeation promoters for numerous<br />

molecules including hydrophilic (e.g. mannitol, 5-fluorouracil and<br />

sulphaguanidine) and lipophilic (betamethasone-17-benzoate,<br />

hydrocortisone and progesterone) permeants. As with many studies,<br />

higher flux enhancements have been reported for the hydrophilic<br />

molecules. Recently NMP was employed with limited success as a<br />

penetration enhancer for captopril when formulated into a matrix type<br />

transdermal patch (Park et al., 2001).<br />

In terms of mechanisms of action, the pyrrolidones partition well<br />

into human corneum stratum. Within the tissue they may act by altering<br />

the solvent nature of the membrane and pyrrolidones have been used to<br />

generate ‘reservoirs’ within skin membranes. Such a reservoir effect<br />

offers potential for sustained release of a permeant from the stratum<br />

corneum over extended time periods (Wiiliam and Barry, 2004).<br />

2.2.4. Fatty acids:<br />

Percutaneous drug absorption has been increased by a wide variety<br />

of long chain fatty acids, the most popular of which is oleic acid. It<br />

appears that saturated alkyl chain lengths of around C10–C12 attached to a<br />

polar head group yields a potent enhancer. In contrast, for penetration<br />

enhancers containing unsaturated alkyl chains, then C18 appears near<br />

optimum. For such unsaturated compounds, the bent cis configuration is<br />

expected to disturb intercellular lipid packing more so than the trans<br />

arrangement, which differs little from the saturated analogue. Santoyo<br />

and Ygartua, employed the mono-unsaturated oleic acid, polyunsaturated,<br />

linoleic and linolenic acids and the saturated lauric acid enhancers for<br />

promoting piroxicam flux (Santoyo and Ygartua, 2000). As with Azone,<br />

oleic acid is effected at relatively low concentrations (typically less than<br />

10%) and can work synergistically when delivered from vehicles such as<br />

PG or ternary systems with dimethyl isosorbide (Aboofazeli et al., 2002)<br />

82


Considerable efforts have been directed at investigating the mechanisms<br />

of action of oleic acid as a penetration enhancer in human skin. It is clear<br />

from numerous literature reports that the enhancer interacts with and<br />

modifies the lipid domains of the stratum corneum, as would be expected<br />

for a long chain fatty acid with a cis configuration.<br />

2.2.5. Alcohols:<br />

Ethanol is the most commonly used alcohol as transdrmal<br />

penetration enhancer, it enhances permeation by extracting large amounts<br />

of stratum corneum lipids, it also increases the number of free sulphydryl<br />

groups of keratin in the stratum corneum proteins (Sinha and Maninder,<br />

2000). It increases permeation of ketoprofen from gel-spray formulation<br />

(Porzio et al., 1998).<br />

2.2.6. Propylene glycol (PG):<br />

PG is widely used alone or as cosolvent for other enhancers. PG<br />

increased the flux of heparin sodium (Bonina and Montenegro, 1992)<br />

and ketoprofen, but at higher concentration it inhibited the flux of<br />

ketoprofen. In combination with azone, PG increased the flux of<br />

methotrexate (Chatterjee et al., 1997), cyclosporine A (Duncan et al.,<br />

1990), and 5-fluouracil (Goodman and Berry, 1988). PG works by<br />

solvating keratin of stratum corneum , occupying hydrogen bonding sites<br />

and, thus reducing drug- tissue binding .<br />

2.2.7. Urea (UR):<br />

Urea is a hydrating agent (a hydrotrope) used in the treatment of<br />

scaling conditions such as psoriasis, ichthyosis and other hyper-keratotic<br />

skin conditions. Applied in a water in oil vehicle, urea alone or in<br />

combination with ammonium lactate produced significant stratum<br />

cornum hydration and improved barrier function when compared to the<br />

vehicle alone in human volunteers in vivo (Gloor et al., 2001). Urea also<br />

has keratolytic properties, usually when used in combination with<br />

83


salicylic acid for keratolysis. The somewhat modest penetration<br />

enhancing activity of urea probably results from a combination of<br />

increasing stratum cornum water content (water is a valuable penetration<br />

enhancer) and through the keratolytic activity.<br />

2.2.8. Surfactant:<br />

As with some of the materials described previously (for example<br />

ethanol and PG) surfactants are found in many existing therapeutic,<br />

cosmetic and agro-chemical preparations. Usually, surfactants are added<br />

to formulations in order to solubilise lipophilic active ingredients, and so<br />

they have potential to solubilise lipids within the stratum corneum.<br />

Typically composed of a lipophilic alkyl or aryl fatty chain, together with<br />

a hydrophilic head group, surfactants are often described in terms of the<br />

nature of the hydrophilic moiety. Anionic surfactants include sodium<br />

lauryl sulphate (SLS), cationic surfactants include cetyltrimethyl<br />

ammonium bromide, the nonoxynol surfactants are non-ionic surfactants<br />

and zwitterionic surfactants include dodecyl betaine. Anionic and cationic<br />

surfactants have potential to damage human skin; SLS is a powerful<br />

irritant and increased the trans epidemeral water loss in human volunteers<br />

in vivo (Tupker et al., 1990) and both anionic and cationic surfactants<br />

swell the stratum corneum and interact with intercellular keratin. Non-<br />

ionic surfactants tend to be widely regarded as safe. Surfactants generally<br />

have low chronic toxicity and most have been shown to enhance the flux<br />

of materials permeating through biological membranes.<br />

Surfactant facilitated permeation of many materials through skin<br />

membranes has been researched, with reports of significant enhancement<br />

of materials such as chloramphenicol through hairless mouse skin by<br />

SLS, and acceleration of hydrocortisone and lidocaine permeating across<br />

84


hairless mouse skin by the non-ionic surfactant Tween 80 (Sarpotdar<br />

and Zatz,1986a, 1986b).<br />

2.2.9. Gramicidin:<br />

Gramicidin is a linear peptide –type cataionic ionophore that has no<br />

charged or hydrophilic chains and its aqueous solubility is low.<br />

Gramicidin increased the flux of benzoic acid through rat abdominal skin<br />

by rearranging lipid barrier and increasing hydration of stratum corneum<br />

(Chi and Choi, 2000).<br />

2.2.10. Phospholipids:<br />

Phosphatidyl glycerol derivative increased the accumulation of<br />

bifonazole in skin and percutaneous penetration of tenoxicam;<br />

phosphatidyl choline derivatives promoted the percutaneous penetration<br />

of erythromycin (Yokomizo, 1996).<br />

2.2.11. Lipid synthesis inhibitors :<br />

The barrier layer consists of a mixture of cholesterol, free fatty<br />

acids, and ceramides, and these three classes of lipids are required for<br />

normal barrier function. Addition of inhibitors of lipid synthesis enhances<br />

the delivery of some drugs like lidocaine and caffeine .Fatty acid<br />

synthesis inhibitors like 5-(tetradecyloxy)-2-furancarboxilic acid (T<strong>OF</strong>A)<br />

and the cholesterol synthesis inhibitors like fluvastatin (FLU) or<br />

cholesterol sulfate (CS) delay the recovery of barrier damage produced by<br />

prior application of penetration enhancers like DMSO, acetone, and like<br />

causes a further boost in the transdermal permeation (Tsia et al., 1996).<br />

85


2.2.12. Amino acid derivatives :<br />

Various amino acid derivatives have been investigated for their<br />

potential in improving percutaneous permeation of drugs. Application of<br />

n-dodecyl-L-amino acid methyl ester and n-pentyl-N-acetyl prolinate on<br />

excised hairless mouse skin 1 hour prior to drug treatment produced<br />

greater penetration of hydrocortisone from its suspension (Fincher et al.,<br />

1996).<br />

2.2.13. Clofibric acid :<br />

Esters and amides of clofibric acid were studied for their<br />

permeation-enhancing property using nude mice skin. The best<br />

enhancement of hydrocortisone-21-acetate and betamethasone-17-<br />

valerate was observed with clofibric acid octyl amide when applied 1<br />

hour prior to each steroid. Amide analogues are generally more effective<br />

than ester derivatives of the same carbon chain length (Michniak et al.,<br />

1993).<br />

2.2.14. Dodecyl-N,N-dimethylamino acetate (DDAA):<br />

DDAA increasesd the transdermal permeation of a number of<br />

drugs,like propranolol HCl and timolol maleate.The permeability<br />

enhancing effect was due to changes in lipid structure of stratum corneum<br />

, like azone and oleic acid (Ruland et al ., 1994) and hydrating effect on<br />

the skin (Fleeker et al., 1989).Its duration of action is shorter than that of<br />

azone and dodecyl alcohol because of presence of hydrophilic groups<br />

(Hirvonen et al., 1994), so there is faster recovery of the skin structure<br />

and hence less irritation potential.<br />

2.2.15. Enzymes :<br />

86


Due to the importance of the phosphatidyl choline metabolism<br />

during maturation of the barrier lipids, the topical application of the<br />

phosphatidyl choline-depentent enzyme phospholipase c produced an<br />

increase in the transdermal flux of benzoic acid,mannitol, and<br />

testosterone . Triglycero hydrolase (TGH) increased the permeation of<br />

mannitol, while phospholipase A2 increased the flux of both benzoic acid<br />

and mannitol (Patil et al., 1996).<br />

87


1. Materials and supplies:<br />

Experiment and methodology<br />

* Glibenclamide was kindly supplied by Egyptian International<br />

Pharmaceutical Industries Company (EIPICO).<br />

* Sodium alginate (El-Gomhouria Company, Eygypt).<br />

* Tween 80 (Merk Sharp and Dohmn, Germany).<br />

* Cetrimide (Searle Company, England).<br />

* Transcutol, labrafil, oleic acid, linoleic acid, isopropylmyristate and<br />

isopropylpalmitate (Sigma Chemical Co.St.Louis, USA).<br />

* Other materials were mentioned previously in chapter two.<br />

2. Equipment:<br />

These were mentioned previously in chapter two.<br />

3. Software:<br />

These were mentioned previously in chapter two.<br />

4. Methods:<br />

4.1. UV scanning of Glib:<br />

About 20 and 100 μg /ml of Glib in methanol were scanned<br />

spectrophotometerically from 200-400 nm using methanol as blank.<br />

4.2. Construction of calibration curve of Glib in sörensen’phosphate<br />

buffer pH 7.4.<br />

0.1 gram of Glib were dissolved in 100 ml methanol to obtain a<br />

solution of concentration of 1mg/ml, 10 ml is diluted to 100 ml with<br />

sörensen’ buffer pH 7.4 to produce a solution containing 100 μg /ml of<br />

Glib . Aliquots of 0.5, 1, 1.5, 2, 2.5, and 3 ml were furtherly diluted to 10<br />

88


ml with sörensen’ buffer pH 7.4. After dilution, the solution contained 10,<br />

15, 20, 25, and 30 μg/ml of Glib respectively.<br />

The calibration equation was constructed by regressing the relative<br />

absorbances, against the corresponding Glib solutionsconcentrations at<br />

227 nm using sörensen’ buffer pH 7.4 as blank.<br />

4.3. Solubility measurements:<br />

Solubility studies were carried out according to the method of<br />

Higuchi and Connors, (1965) as mentioned before.<br />

4.4. Determination of partition coefficient of Glib:<br />

Partition coefficient of Glib in octanol/water system was determined<br />

as mentioned before.<br />

4.5. The methods of preparation of topical preparations:<br />

The following formulae were selected in which 10 mg of Glib in<br />

each 1 gm of the topical base were incorporated.<br />

4.5.1. Water soluble base:<br />

Polyethylene glycol base :( U.S.P. XXII).<br />

PEG 4000 40 gm<br />

PEG 400 60 gm<br />

Preparation<br />

Water soluble base was prepared as mentioned before.<br />

4.5.2. Absorption base: (U.S.P.XXII)<br />

White soft paraffin 95 gm.<br />

Span 80 5 gm<br />

89


4.5.3. Oleaginous base: (Ammar., et al 2007)<br />

White soft paraffin 100 gm.<br />

Preparation:<br />

Accurate amount of the drug was weighed, levigated and<br />

incorporated into the melted base with continuous stirring until congealed<br />

then packed into plastic jar until used.<br />

4.5.4. Emulsion base:<br />

O/W emulsion base (Beeler’s base) (Ezzedeen et al., 1986):<br />

White bees wax 1 gm<br />

Cetyl alcohol 15 gm<br />

Propylene glycol 10 gm<br />

Sodium lauryl sulphate 2 gm.<br />

Water 72 gm.<br />

Preparation:<br />

O/W emulsion base was prepared as mentioned previously.<br />

4.5.5. Gel bases:<br />

* Hydroxypropyl methylcellulose gel (Sobati, 1998):<br />

HPMC 12 gm<br />

water 88 gm<br />

* Sodium alginate gel (Sobati, 1998):<br />

Sodium alginate 8 gm<br />

Water 92 gm<br />

90


Preparation:<br />

The drug was dispersed in a quantity of water then the gelling agent<br />

was added with continuous stirring and was set aside for complete<br />

swelling and the weight was adjusted by the addition of the water.<br />

:<br />

4.5.6. Hydroxypropyl methylcellulose emulgel (Gehan, 1999):<br />

Liquid paraffin 20 gm<br />

Tween 80 1 gm<br />

Water 70 gm<br />

HPMC 9 gm<br />

Preparation:<br />

-A mixture of the aqueous phase containing hydrophilic emulsifier was<br />

added to the oily phase to form a primary O/W emulsion.<br />

- Drug was suspended into the primary emulsion, then the specified<br />

quantity of the gelling agent powder was sprinkled on the emulsion<br />

surface and was left a side for complete swelling and formation of<br />

emulgel.<br />

All the formulations mentioned previously were summarized in Table 24<br />

91


Table 24 : Composition of different topical formulations.<br />

Type of<br />

base<br />

PEG<br />

4000<br />

Water<br />

soluble<br />

base<br />

(PEG<br />

base)<br />

30<br />

PEG 400 70<br />

Span 80 5<br />

Absorp<br />

t-ion<br />

base<br />

Oleagino<br />

u-s base<br />

92<br />

Emulsio<br />

n base<br />

(O/W<br />

base)<br />

Gel base<br />

HPMC Sod.<br />

Algin<br />

a-te<br />

Soft<br />

paraffin<br />

95 100<br />

Propylene<br />

glycol<br />

10<br />

White<br />

bees wax<br />

1<br />

Sodium<br />

lauryl<br />

sulphate<br />

2<br />

HPMC 12 8 9<br />

Tween 80 2<br />

Liquid<br />

paraffin<br />

Sodium<br />

alginate<br />

20<br />

Water 88 92 69<br />

Emulg<br />

el base<br />

(HPM<br />

C<br />

emulge<br />

l)


4.6. In vitro release of Glib from different topical formulation:<br />

The release study was determined using the simple dialysis<br />

technique as mentioned in part one. 1 gm of the tested formulation<br />

containing (10 mg of the drug) was accurately weighted over the<br />

cellophane membrane (Donor). The diffusion cell was placed at the center<br />

of 1000 ml dissolution cell containing 100 ml of phosphate buffer pH 7.4<br />

(Receptor). The stirring rate was 100 rpm and the temperature was<br />

maintained at 37 ± 0.5 °C<br />

At suitable time intervals 2.5 ml sample was withdrawn from the<br />

sink solution assayed spectrophotometerically at 227 nm using a suitable<br />

blank.A similar volume of buffer was added to mentain the volume of<br />

receptor constant. Each experiment was done in triplicate, and the<br />

average was calculated. The cumulative amount of the drug released was<br />

calculated as mentioned in chapter one.<br />

4.7. Penetration enhancers screening procedure:<br />

In order to select penetration enhancers which lend themselves to a<br />

more detailed investigation, the screening procedure were developed<br />

based on the percentage of the drug released after six hours.<br />

4.8. Effect of incorporation of different penetration enhancers in<br />

water soluble base:<br />

On the basis of results obtained in the previous screening, different<br />

penetration enhancers with different concentrations were incorporated in<br />

the topical formulation that demonstrated the best release results (water<br />

soluble base) as shown in (Table 25 ). In vitro release of these<br />

preparations was done as mentioned above.<br />

Table 25 : Types of penetration enhancers and percentages used.<br />

93


Penetration enhancers Percentages used<br />

(A)Surfactants<br />

1.Cationic surfactant<br />

(cetrimide) 0.3 0.5 1 2<br />

2.Anionic surfactant<br />

(SLS) 0.1 0.4 0.8<br />

3. Non ionic surfactant<br />

i.Tween 80 (Tw-80) 0.3 1 4 5<br />

ii. Labrafil (Lab) 3 5 7<br />

B) Solubilizing agents<br />

Transcutol (Tc) 5 8<br />

(C) Unsaturated free<br />

fatty acids<br />

1. Oleic acid (OA)<br />

0.5<br />

94<br />

1 <br />

2. Linoleic acid (LOA) 0.8 <br />

(D) Fatty acid esters<br />

1.Isopropyl myristate<br />

(IPM)<br />

2. Isopropyl palmitate<br />

(IPP)<br />

0.5<br />

<br />

0.2


4.9. Kinetic evaluation of the in vitro release data:<br />

The data obtained from the experiments were analyzed to know the<br />

mechanism of the release of the drug using the following kinetic<br />

equations:<br />

(I) Zero order kinetics: A=A-k<br />

(II) First order kinetics: ln A = ln A- kt<br />

log A = log A- kt/ 2.303<br />

(III) Higuchi diffusion model:<br />

M = Q = 2C ) ½<br />

4.10. In vitro permeation of glibenclamide through abdominal rabbit<br />

skin:<br />

Preparation of the rabbit skin and in vitro permeation of Glib were<br />

done by the same methods mentioned in part one.<br />

4.11. Statistical analysis:<br />

Data were expressed as mean of three experiments ± the standard<br />

error (S.E.). The obtained data were compared statistically using One-<br />

way analysis of variance (ANOVA) test of significance on a computer<br />

statistical SPSS analysis program. A p-value of 0.05 or less was<br />

considered to be significant (Suwanpidokkul et al., 2004).<br />

4.12. In vivo studies:<br />

4.12.1. Animals:<br />

The animals used for the anti diabetic and hypoglycemic activity study<br />

were white adult albino rats weighing between 200-250g. The animals<br />

were housed under standard laboratory conditions.<br />

95


4.12.2. Hypoglycemic activity in normal rats:<br />

The hair on the backside of the rats was removed with an electric hair<br />

clipper on the previous day of the experiment. The oral doses were given<br />

using a round tipped stainless steel needle attached to 1 ml syringe.<br />

Following an overnight fast, rats were divided into7 groups (n=5). The<br />

rats were treated as follows:<br />

Group I (Control) – 1ml gum acacia suspension was given orally.<br />

- 5 mg/kg Glib in mucilage of gum acacia was given orally<br />

(Mutalik and Udupa, 2005).<br />

Group III - 5 mg drug incorporated in 1gm water soluble ointment base<br />

was applied topically.<br />

Group IV - 5 mg drug incorporated in 1gm water soluble ointment base<br />

containing 1% oleic acid was applied topically.<br />

Group V - 5 mg drug incorporated in 1gm water soluble ointment base<br />

containing 1% cetrimide was applied topically.<br />

Group VI - 5 mg drug incorporated in 1gm water soluble ointment base<br />

containing 1% isopropylmyristate was applied topically.<br />

Group VII - 5 mg drug incorporated in 1gm water soluble ointment<br />

base containing 5% Labrafil was applied topically.<br />

At time intervals between 2-24 h after treatment blood was collected<br />

from orbital sinuses; blood glucose levels were determined using the<br />

glucometer.<br />

96


4.12.3. Hypoglycemic activity in diabetic rats:<br />

4.12.3.1. Induction of diabetes mellitus:<br />

The overnight fasted rats were made diabetic by a single<br />

intraperitoneal injection of streptozotocin (STZ) (50 mg/kg; i.p) dissolved<br />

in citrate buffer (pH 4.5). The blood glucose was measured after 24 hrs<br />

and animals with blood glucose levels >250 mg/dL were selected<br />

(Sridevi et al., 2000).<br />

4.12.3.2. Anti-diabetic activity in diabetic rats:<br />

The anti diabetic activity of the prepared topical preparation was<br />

evaluated in overnight fasted diabetic rats.<br />

Diabetic rats were divided into 3 groups (n=5). The rats were treated as<br />

follows:<br />

Group I (Control) – 1ml gum acacia suspension was given orally.<br />

Group II -Glib 5 mg/kg was given orally (Mutalik and Udupa, 2005).<br />

Group III –5 mg drug incorporated in 1gm water soluble ointment base<br />

in presence of 1% cetrimide was applied topically.<br />

At time intervals between 2-24 h after treatment blood was<br />

collected from orbital sinuses; blood glucose levels were determined<br />

using the glucometer.<br />

4.12.4. Statistical analysis:<br />

Data were expressed as mean of three experiments ± the standard<br />

error (S.E.). The obtained data were compared statistically using One-<br />

way analysis of variance (ANOVA) test of significance on a computer<br />

statistical SPSS analysis program. A p-value of 0.05 or less was<br />

considered to be significant (Suwanpidokkul et al., 2004).<br />

97


1. UV scanning of Glib:<br />

Results and Discussion<br />

UV scanning of Glib in methanol was carried out (Figure44). Three<br />

absorption maxima were observed at 299, 278 and 227 nm at<br />

concentration of Glib of (100 μg/ml) and nearly the same wavelengths<br />

were observed at concentration of Glib of (20μg/ml) with lower intensity.<br />

The measurements were done at 227 nm (Siavoush et al., 2005).<br />

20 μg/ml<br />

227nm<br />

100 μg/ml<br />

278nm<br />

Wavelength<br />

Figure 44: UV absorption spectra for Glib in methanol.<br />

98<br />

299nm


2. Calibration curves of Glib in sörensen’s phosphate buffer pH (7.4):<br />

(Figure 45) show a linear relationship between the absorbance and<br />

the concentration of Glib in sörensen’s phosphate buffer pH 7.4 at the<br />

max in the concentration range used.<br />

3. Solubility measurements:<br />

In the present study, the solubility of the Glib in distilled water and<br />

in sörensen’s phosphate buffer pH 7.4 at 25 C was found to be 4.41<br />

μg/ml and 16.19 μg/ml respectively.<br />

4. Partition coefficient of Glib:<br />

In the present study, the partition coefficient of Glib was found<br />

to be 2.05 (log octanol/ water =0.312) and this is in agreement with that<br />

observed by Srinivas and Nayanabhirama, 2005, who found that the<br />

log octanol /buffer = 0.32.<br />

99


5. Release of Glib from different topical bases:<br />

The influence of the type of base on the in vitro release of Glib has<br />

been studied. The bases investigated consisted of ointment (water soluble<br />

base, emulsion base, absorption base, and oleaginous base), emulgel and<br />

gel (HPMC and sodium alginate gels).<br />

Results of release from different topical bases are summarized in<br />

(Table 26) and graphically illustrated in (Figures 46-47).<br />

Due to the high lipid solubility and low water solubility (4.41<br />

μg/ml) of Glib, this may explain the slow release of the drug that is<br />

observed from all bases.<br />

From the data obtained it is clear that the percentage amount of drug<br />

released from water soluble base, gel bases and emulgel base are greater<br />

than that released from other bases. The rate of drug release can be<br />

arranged in the following descending order:<br />

Water soluble base (5.94 %) > HPMC emulgel (4.6 %) > sodium alginate<br />

gel (4.38 %) > HPMC gel (3.99) >O/W emulsion base (2.5 %) ><br />

absorption base (1.94%) > oleaginous base (1.61%).It is clear that, water<br />

soluble base showed the highest release than that of emulsion, gels,<br />

emulgel, oleaginous and absorption bases.<br />

100


Table 26: In vitro release of glibenclamide from different topical bases.<br />

Glibenclamide released % ± (sd)<br />

Time (min) WSB HPMC emulgel Sodium alginate gel HPMC gel<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 0.54 ± 0 0.65 ± 0.07 0.73 ± 0.016<br />

60 1.5 ± 0.09 1.22 ± 0.06 0.95 ± 0.05 1.16 ± 0.06<br />

90 1.71 ± 0.25 2.09 ± 0.14 1.45 ± 0.08 1.71 ± 0.13<br />

120 2.25 ± 0.07 2.19 ± 0.19 1.97 ± 0.12 2.36 ± 0.03<br />

150 2.86 ± 0.14 2.46 ± 0.048 2.44 ± 0.26 2.46 ± 0.14<br />

180 3.44 ± 0.12 2.71 ± .062 2.68 ± 0.16 2.5 ± 0.25<br />

240 4.8 ± 0.25 3.39 ± 0.17 3.15 ± 0.28 2.91 ± 0.15<br />

300 5.29 ± 0.11 3.86 ± 0.18 4.14 ± 0.31 3.40 ± 0.25<br />

360 5.94 ± 0.41 4.6 ± 0.27 4.38 ± 0.3 3.99 ± 0.05<br />

101


Cont.Table 26: In vitro release of glibenclamide from different topical bases.<br />

Glibenclamide released % ± (sd)<br />

Time (min) O/W cream Absorption base Oleaginous base<br />

0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.54 ± 0 0.44 ± 0.02 0.60 ± 0.013<br />

60 0.65± 0.03 0.49 ± 0 0.89 ± 0.06<br />

90 0.97 ± 0.038 0.97 ± 0.13 0.91 ± 0.13<br />

120 1.13 ± 0.06 1.055 ± 0.21 1.22 ± 0.12<br />

150 1.97 ± 0.08 1.12 ± 0.08 1.27 ± 0.08<br />

180 1.81± 0.04 1.27 ± 0.15 1.48 ± 0.15<br />

240 1.92 ± 0.07 1.35 ± 0.56 1.34 ± 0.2<br />

300 2.2 ± 0.09 1.37 ± 0.9 1.51 ± 0.14<br />

360 2.5 ± 0.1 1.94 ± 0.38 1.61 ± 0.17<br />

102


WSB base HPMC gel HPMC emulgel Sodium alginate gel<br />

O/W cream Oleagineous base Absorption base<br />

7<br />

6<br />

5<br />

4<br />

3<br />

% drug releasesd<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 46: Release profile of glibenclamide from different topical bases.<br />

103


The highest release may be attributed to the rapid dissolution of the base<br />

in water and the possible solubilizing effect of the base components<br />

(Moes, 1982; Anshel, 1976)<br />

(Chakole et al., 2009) found that halobetasol propionate and Fusidic<br />

acid ointment formulation containing water miscible base showed better<br />

in-vitro release profile in comparison to oleaginous base.<br />

(Dhavse and Amin, 1997) stated that norfloxacin formulations<br />

containing polyethylene glycol and Carbopol gel base showed better in<br />

vitro release profile in comparison to creams and ointment base<br />

formulations.<br />

The higher release of the Glib from emulgel and gel bases than O/W<br />

emulsion base, oleaginous and absorption ointment bases is considered to<br />

be due to the high miscibility of these bases with the dissolution medium.<br />

The higher release of the Glib from emulgel than gel bases is<br />

considered to be due to the presence of Tween80 which can facilitate the<br />

release of drug.<br />

The lower release of the drug from O/W emulsion base than from<br />

water soluble base, emulgel and gels owing to its biphasic nature which<br />

leading to partitioning of the drug in 2 phases, that results in slower<br />

release of drug (Dhavse and Amin, 1997)<br />

The higher release of the Glib from O/W emulsion base than from<br />

absorbtion base and oleaginous base may be due to the formation of a<br />

continuous contact between the external phase of the O/W emulsion base<br />

and the buffer (Nakano et al., 1976).<br />

104


7<br />

WSB<br />

6<br />

WSB<br />

HPMC emulgel<br />

Sod. alginate<br />

HPMC gel<br />

O/W cream<br />

Absorption base<br />

Oleaginous base<br />

5<br />

HPMC emulgel<br />

Sod. alginate<br />

HPMC gel<br />

4<br />

3<br />

O/W cream<br />

% Drug released<br />

Absorption base<br />

Oleaginous base<br />

2<br />

1<br />

0<br />

1<br />

Figure 47: Percentage drug released from different topical bases.<br />

105


In case of oleaginous base and absorption base, the external phase is non-<br />

polar and immiscible with the polar diffusion medium hence retardation<br />

of drug release is expected. Also this low release may be attributed to the<br />

closing of the cellophane membrane pores with the fatty base and<br />

prevention of penetration of the acceptor medium through the membrane<br />

to dissolve the drug (Habib and El-Shanawany, 1989).<br />

6. Effect of incorporation of penetration enhancers:<br />

The transdermal route has been recognized as one of the highly<br />

potential routes of systemic drug delivery and provides the advantage of<br />

avoidance of the first-pass effect, ease of use and withdrawal (in case of<br />

side effects), and better patient compliance. However, the major<br />

limitation of this route is the difficulty of permeation of drug through the<br />

skin (Sinha and Maninder, 2000). Studies have been carried out to find<br />

safe and suitable permeation enhancers to promote the percutaneous<br />

absorption of Glib.<br />

106


6.1. Effect of incorporation of surfactants:<br />

The effect of surfactant on the release of Glib from the prepared<br />

water soluble base is shown in (Tables 27- 30) and (Figures 49-53).<br />

Anionic, cationic and non-ionic surfactants were used.<br />

6.1.1. Anionic surfactants:<br />

Incorporation of sodium lauryl sulphate (SLS) in concentrations of<br />

0.4% and 0.8% increased the percentage of drug released from 5.94 % to<br />

7.95% and 7.12% respectively. While incorporation of SLS in<br />

concentration of 0.1% decreased the amount of drug released by (-1.06<br />

fold) in comparison to control.<br />

Nokhodchi et al., 2003 studied the enhancing effects of SLS on the<br />

permeation of lorazepam through rat skin and he found that, sodium<br />

lauryl sulphate at a concentration of 5% w/w (the highest concentration)<br />

exhibited the greatest increase in flux of lorazepam compared with<br />

control.<br />

6.1.2. Cationic surfactants:<br />

Incorporation of cetrimide (Cetylpyridiniumbromide) in<br />

concentrations of 0.3%, 0.5% and 1% increased the percentage of drug<br />

released from 5.94 % to 7.18%, 8.75% and 9.48% respectively. While<br />

incorporation of cetrimide in concentration of 2% decreased the amount<br />

of drug released by (-1.17 fold) in comparison to control.<br />

6.1.3. Non-ionic surfactants:<br />

<br />

5% increased the percentage of drug released from 5.94 % to 9.05% and<br />

6.76% respectively. While incorporation of Tween 80 in concentration of<br />

0.3% and 1% decreased the amount of drug released by (-1.04 fold) in<br />

comparison to control. This is in accordance with (Ramadan, 2008) who<br />

studied Enhancement factors for the penetration of miconazole through<br />

cellulose barrier from different bioadhesive gels containing different<br />

107


concentrations of Tween80 and she found that, 1% concentration of<br />

enhancers used seems to be the optimum concentration at which the<br />

maximum release and concentration of enhancers beyond the maximum<br />

concentration would be responsible for permeability coefficient declined<br />

and reducing of enhancement effect. Enhancer at high level showed a<br />

lower tendency to solubilize the drug which may be attributed to complex<br />

formation.<br />

-6 glycerides) in<br />

concentrations of 3% and 5% increased the percentage of drug released<br />

from 5.94 % to 7.32% and 10.03% respectively. While incorporation of<br />

labrafil in concentration of 7% decreased the amount of drug released by<br />

(-1.11 fold) in comparison to control.<br />

Choi et al., 2003 found that incorporation of 1.5% of labrafil in<br />

50/50 buffer (pH 10)/ PG solvent mixture increased the permeability of<br />

clenbuterol through hairless mouse skin approximately 8 folds more than<br />

control without permeation enhancer.<br />

Based on the above mentioned results, it is obvious that addition of<br />

the surfactant into the ointment base could result in increased solubility of<br />

the hydrophobic drug, leading to the increase in the drug release rate<br />

(Sarisuta et al., 1999). In addition increasing the concentration of<br />

surfactant may decrease drug release and this may be attributed to<br />

miceller complexation which decreases thermodynamic activity of the<br />

drug (Fergany, 2001).<br />

108


Table 27: In vitro release of glibenclamide from water soluble base containing different concentrations of cetrimide.<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.3% 0.5% 1% 2%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 0.69 ± 0.2 0.7 ± 0.07 1.09 ± 0.035 0.6 ± 0.01<br />

60 1.5 ± 0.09 1.11 ± 0.12 2.02 ± 0.16 2.23 ± 0.11 1.1 ± 0<br />

90 1.71 ± 0.25 2.36 ± 0.14 3.26 ± 0.37 3.29 ± 0.18 1.72± 0.01<br />

120 2.25 ± 0.07 3.15 ± 0.14 4.22 ± 0.33 4.3 ± 0.07 2.18 ± 0.04<br />

150 2.68 ± 0.14 3.75 ± 0.19 5.16 ± 0.33 5.2 ± 0.51 2.57 ± 0. 2<br />

180 3.44 ± 0.12 4.41 ± 0.34 5.89 ± 0.12 6.1± 0.53 3.22 ± 0.19<br />

240 4.8 ± 0.25 5.59 ± 0.17 6.84 ± 0.12 7.34 ± 0.48 3.64 ± 0.007<br />

300 5.29 ± 0.11 6.73 ± 0.18 7.87 ± 0.24 8.6 ± 0.16 4.17 ± 0.19<br />

360 5.94 ± 0.41 7.18 ± 0.19 8.75 ± 0.34 9.48 ± 0.56 5.05 ± 0.2<br />

109


Drug alone 0.3% cetrimide 0.5% cetrimide 1% cetrimide 2% cetrimide<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 49: Release profile of glibenclamide from water soluble base containing different concentrations of cetrimide.<br />

110


Table 28: In vitro release of glibenclamide from water soluble base containing different concentrations of Sodium<br />

lauryl sulphate (SLS).<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.1% 0.4% 0.8%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 0.5 ± 0 1.96 ± 0.11 1.75 ± 0.014<br />

60 1.5 ± 0.09 1.42 ± 0.14 2.96 ± 0.07 2.59 ± 0.014<br />

90 1.71 ± 0.25 1.81 ± 0.09 3.66 ± 0.14 3.38± 0.077<br />

120 2.25 ± 0.07 2.22 ± 0.06 3.96 ± 0.16 3.83 ± 0.056<br />

150 2.68 ± 0.14 2.7 ± 0.19 4.41 ± 0.15 4.22 ± 0. 3<br />

180 3.44 ± 0.12 3.37 ± 0.11 4.96 ± 0.17 4.85 ± 0.13<br />

240 4.8 ± 0.25 4.3 ± 0.46 6.56 ± 0.16 5.9 ± 0.17<br />

300 5.29 ± 0.11 5.11 ± 0.35 7.26 ± 0. 4 6.92 ± 0.18<br />

360 5.94 ± 0.41 5.59 ± 0.41 7.95 ± 0.16 7.12 ± 0.18<br />

111


12<br />

Drug alone 0.1% SLS 0.4% SLS 0.8% SLS<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 50: Release profile of glibenclamide from water soluble base containing different concentrations of SLS.<br />

112


Table 29: In vitro release of glibenclamide from water soluble base containing different concentrations of Tween 80.<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.3% 1% 4% 5%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 0.67 ± 0.16 0.95 ± 0.1 1.72 ± 0.05 1.1 ± 0.05<br />

60 1.5 ± 0.09 1.32 ± 0.11 1.72 ± 0.2 2.47 ± 0.07 2.12 ± 0.07<br />

90 1.71 ± 0.25 1.95 ± 0.15 2.17 ± 0.07 3.45 ± 0.2 2.72± 0.04<br />

120 2.25 ± 0.07 2.44 ± 0.07 2.75 ± 0.16 4.33 ± 0.007 3.31± 0.13<br />

150 2.68 ± 0.14 3.03 ± 0.06 3.16 ± 0.03 5.3 ± 0.24 3.89 ± 0. 07<br />

180 3.44 ± 0.12 3.5 ± 0.15 3.68 ± 0.02 5.64± 0.26 4.37 ± 0.24<br />

240 4.8 ± 0.25 4.25 ± 0.1 4.99 ± 0.19 7.05 ± 0.14 5.08 ± 0.33<br />

300 5.29 ± 0.11 4.71 ± 0.09 4.97 ± 0.04 8.2 ± 0.15 5.81 ± 0.36<br />

360 5.94 ± 0.41 5.66 ± 0.09 5.69 ± 0.08 9.05 ± 0.23 6.76 ± 0.24<br />

113


Drug alone 0.3% Tw- 80 1% Tw- 80 4% Tw- 80 5% Tw- 80<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 51: Release profile of glibenclamide from water soluble base containing different concentrations of Tween 80.<br />

114


Table 30: In vitro release of glibenclamide from water soluble base containing different concentrations of labrafil.<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 3% 5% 7%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 1.68 ± 0 1.59 ± 0.27 0.84 ± 0.019<br />

60 1.5 ± 0.09 3.01 ± 0.17 3.23 ± 0.21 1.97 ± 0.2<br />

90 1.71 ± 0.25 3.56 ± 0.16 4.02 ± 0.23 2.46 ± 0.15<br />

120 2.25 ± 0.07 4.68 ± 0.25 4.96 ± 0.14 2.69 ± 0.36<br />

150 2.68 ± 0.14 5.43 ± 0.33 5.5 ± 0.37 2.98 ± 0.21<br />

180 3.44 ± 0.12 4.41 ± 0.18 5.97 ± 0.24 3.1 ± 0.17<br />

240 4.8 ± 0.25 5.86 ± 0.11 8.46 ± 0.14 4.13 ± 0.32<br />

300 5.29 ± 0.11 6.72 ± 0.22 9.3 ± 0.26 4.96 ± 0.25<br />

360 5.94 ± 0.41 7.32 ± 0.17 10.03 ± 0.18 5.33 ± 0.22<br />

115


Drug alone Lab 3% Lab 5% Lab 7%<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 52: Release profile of glibenclamide from water soluble base containing different concentrations of labrafil.<br />

116


117


Drug alone Cetrimide 0.3% Cetrimide 0.5% Cetrimide 1% Cetrimide 2%<br />

SLS 0.1% SLS 0.4% SLS 0.8% Tw 80 0.3% Tw 80 1%<br />

Tw 80 4% Tw 80 5% Lab 3% Lab 5% Lab 7%<br />

12<br />

Lab 5%<br />

Cetrimide<br />

1%<br />

10<br />

Tw 80 4%<br />

Cetrimide<br />

0.5%<br />

SLS 0.4%<br />

Lab 3%<br />

Cetrimide<br />

0.3%<br />

8<br />

Tw 80<br />

5%<br />

SLS 0.8%<br />

Tw 80<br />

1%<br />

Tw 80<br />

0.3%<br />

SLS<br />

0.1%<br />

Drug<br />

Lab 7%<br />

Cetrimide<br />

2%<br />

6<br />

% Drug released<br />

4<br />

2<br />

0<br />

1<br />

Figure 53: Percentage drug released from water soluble base containing different concentrations of different<br />

surfactants.<br />

118


6.2. Effect of incorporation of fatty acids:<br />

The effect of unsaturated fatty acids on the release of Glib from the<br />

prepared water soluble base is shown in (Tables 31-32) and (Figures 54-<br />

56).<br />

<br />

increased the percentage of drug released from 5.94 % to 6.19% and<br />

8.8%. However, concentrations of 2% and 3% increased the release of<br />

Glib when compared with the control but didn't lead to a further increase<br />

in permeation and this is with agreement with (Ammar., et al 2007) who<br />

studied the effect of oleic acid on the transdermal delivery of aspirin and<br />

he found that oleic acid enhanced aspirin permeation from CMC gel base<br />

at a concentration of 5% or 10% However, a concentration of 20%<br />

enhanced the permeation when compared with the control but didn't lead<br />

to a further increase in permeation. This may be attributed to an increase<br />

in the lipophilicity of the vehicle (Ammar., et al 2006).<br />

<br />

2% increased the percentage of drug released from 5.94 % to 7.34%,<br />

6.56% and 6.43% respectively.<br />

Gwak and Chun, 2001 studied the effect of linoleic acid on<br />

transdermal delivery of aspalatone and they found that, linoleic acid<br />

(LOA) at the concentration of 5% was found to have the largest<br />

enhancement factor. However, a further increase in aspalatone flux was<br />

not found in the fatty acid concentration greater than 5%, indicating the<br />

enhancement effect is in a bell-shaped curve<br />

119


Table 31: In vitro release of glibenclamide from water soluble base containing different concentrations of oleic acid.<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.5% 1% 2% 3%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 0.87 ± 0 1.59 ± 0.16 1.15 ± 0.023 1.31 ± 0.035<br />

60 1.5 ± 0.09 2.13 ± 0.05 2.7 ± 0.15 2.71 ± 0.36 2.08 ± 0.14<br />

90 1.71 ± 0.25 2.95 ± 0.09 3.89 ± 0.13 3.74 ± 0.35 2.81± 0.25<br />

120 2.25 ± 0.07 3.44 ± 0.07 4.5 ± 0.39 4.15 ± 0.17 3.08± 0.37<br />

150 2.68 ± 0.14 3.71 ± 0.08 4.96 ± 0.4 4.73 ± 0.24 3.5 ± 0. 32<br />

180 3.44 ± 0.12 4.24 ± 0.13 5.86 ± 0.34 5.4± 0.45 3.87 ± 0.24<br />

240 4.8 ± 0.25 5.05 ± 0.21 7.09 ± 0.32 6.82 ± 0.43 4.98 ± 0.19<br />

300 5.29 ± 0.11 5.64 ± 0.35 7.9 ± 0.45 7.4 ± 0.15 5.38 ± 0.22<br />

360 5.94 ± 0.41 6.19 ± 0.27 8.8 ± 0.4 7.67 ± 0.19 6.68 ± 0.21<br />

120


Drug alone 0.5% OA 1% OA 2% OA 3% OA<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 54: Release profile of glibenclamide from water soluble base containing different concentrations of oleic<br />

acid.<br />

121


Table 32: In vitro release of glibenclamide from water soluble base containing different concentrations of linoleic<br />

acid.<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.8% 1% 2%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 1.54 ± 0.024 1.45 ± 0.09 0.97 ± 0.1<br />

60 1.5 ± 0.09 2.42 ± 0.17 1.98 ± 0.16 1.95 ± 0.15<br />

90 1.71 ± 0.25 2.9 ± 0.17 2.73 ± 0.23 2.51 ± 0.06<br />

120 2.25 ± 0.07 3.33± 0.28 3.27 ± 0.09 3.22 ± 0.25<br />

150 2.68 ± 0.14 4.31 ± 0.38 3.84 ± 0.18 3.81 ± 0. 09<br />

180 3.44 ± 0.12 4.9 ± 0.35 4.42 ± 0.44 4.2 ± 0.26<br />

240 4.8 ± 0.25 5.89 ± 0.59 5.2 ± 0.24 5.12 ± 0.28<br />

300 5.29 ± 0.11 6.66 ± 0.52 6.45 ± 0.28 5.9 ± 0.42<br />

360 5.94 ± 0.41 7.34 ± 0.48 6.56 ± 0.47 6.43 ± 0.37<br />

122


Drug alone 0.8% LOA 1% LOA 2% LOA<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 55: Release profile of glibenclamide from water soluble base containing different concentrations of linoleic<br />

acid.<br />

123


10<br />

Drug alone OA 0.5% OA 1% OA 2% OA 3% LOA 0.8% LOA 1% LOA 2%<br />

OA 1%<br />

9<br />

OA 2%<br />

8<br />

LOA 0.8%<br />

LOA 1%<br />

OA 3%<br />

7<br />

LOA 2%<br />

OA 0.5%<br />

Drug alone<br />

6<br />

5<br />

4<br />

% Drug released<br />

3<br />

2<br />

1<br />

0<br />

1<br />

Figure 56: Percentage drug released from water soluble base containing different concentrations of fatty acids.<br />

124


6.3. Effect of incorporation of fatty acid esters:<br />

The effect of fatty acid esters namely isopropylpalmitate (IPP) and<br />

isopropylmyristate (IPM) on the release of Glib from the prepared water<br />

soluble base is shown in (Tables 33-34) and (Figures 57, 58 and 60).<br />

<br />

increased the percentage of drug released from 5.94 % to 6.11%, 10.26%<br />

and 6.82% respectively.<br />

<br />

increased the percentage of drug released from 5.94 % to 8.39%, 7.33%<br />

and 7.025% respectively.<br />

Malay et al., 2006 investigated the effect of (10% W/W) IPP and<br />

IPM on transdermal permeation of trazodone hydrochloride from matrix-<br />

based formulations through the skin and he found that, the highest<br />

enhancing effect was obtained with IPM followed by IPP. The<br />

permeation of TZN in the presence of 10% w/w of IPM and IPP was 3.79<br />

and 2.00 times greater, respectively, than that in absence of these<br />

enhancers.<br />

125


Table 33: In vitro release of glibenclamide from water soluble base containing different concentrations of<br />

Isopropylmyristate (IPM).<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.5% 1% 2%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 1.36 ± 0.019 1.67 ± 0.05 1.55 ± 0.012<br />

60 1.5 ± 0.09 2.31 ± 0.17 3.45 ± 0.05 2.61 ± 0.13<br />

90 1.71 ± 0.25 2.72 ± 0.04 4.7 ± 0.13 3.51± 0.1<br />

120 2.25 ± 0.07 3.17 ± 0.07 5.4 ± 0.24 3.71 ± 0.3<br />

150 2.68 ± 0.14 3.54 ± 0.16 6.33 ± 0.06 4.13 ± 0. 26<br />

180 3.44 ± 0.12 3.95 ± 0.28 6.94 ± 0.33 4.84 ± 0.4<br />

240 4.8 ± 0.25 4.71 ± 0.23 8.09 ± 0.41 5.67 ± 0.05<br />

300 5.29 ± 0.11 5.36± 0.51 9.33 ± 0. 14 6.07 ± 0.16<br />

360 5.94 ± 0.41 6.11 ± 0.24 10.26 ± 0.4 6.82 ± 0.27<br />

126


Drug alone 0.5%IPM 1% IPM 2% IPM<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 57: Release profile of glibenclamide from water soluble base containing different concentrations of<br />

isopropyl myristate.<br />

127


Table 34: In vitro release of glibenclamide from water soluble base containing different concentrations of<br />

Isopropylpalmitate (IPP).<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 0.2% 1% 2%<br />

0 0 ± 0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 0.86 ± 0.03 1.19 ± 0.11 0.89 ± 0.011<br />

60 1.5 ± 0.09 2.19 ± 0.09 2.15 ± 0.11 1.95 ± 0.11<br />

90 1.71 ± 0.25 3.02 ± 0.04 3.09 ± 0.28 2.75± 0.16<br />

120 2.25 ± 0.07 3.51 ± 0.02 3.58 ± 0.02 3.41± 0.12<br />

150 2.68 ± 0.14 3.94 ± 0.04 4.04 ± 0.06 3.84 ± 0. 29<br />

180 3.44 ± 0.12 4.93 ± 0.04 4.65 ± 0.24 4.65 ± 0.09<br />

240 4.8 ± 0.25 6.37 ± 0.35 5.55 ± 0.14 5.49 ± 0.24<br />

300 5.29 ± 0.11 7.34± 0.04 6.34 ± 0. 31 6.02 ± 0.042<br />

360 5.94 ± 0.41 8.39 ± 0.34 7.33 ± 0.4 7.025 ± 0.31<br />

128


Drug alone 0.2% IPP 1% IPP 2% IPP<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 58: Release profile of glibenclamide from water soluble base containing different concentrations of<br />

isopropyl palmitate (IPP).<br />

129


6.4. Effect of incorporation of solubilizing agent (Transcutol):<br />

The effect of Transcutol on the release of Glib from water soluble<br />

base is shown in (Table 35) and (Figures 59- 60).<br />

Transcutol (Tc) (Diethylene glycol monoethyl ether) is a powerful<br />

solubilizing agent used in several dosage forms and it seems to be very<br />

attractive as a penetration enhancer due to its non-toxicity,<br />

biocompatibility with the skin, miscibility with polar and non polar<br />

solvents and optimal solubilizing properties for a number of drugs<br />

(Barthelemy et al., 1995).<br />

Incorporation of Transcutol in concentrations of 5%and 8%<br />

increased the percentage of drug released from 5.94 % to 7.25% and 6.5%<br />

respectively. Mura et al., (2000) found that incorporation of 50% of<br />

Transcutol in carbapol hydrogel increased clonazepam flux three times<br />

more than control gel.<br />

The enhancing mechanism of Transcutol may be due to its powerful<br />

solubilizing ability and consequently drug leaching increased.<br />

Mutalik and Udupa, 2003 studied the effect of some penetration<br />

enhancers on in vitro permeation of Glib and glipizide through mouse<br />

skin. Ethanol in various concentrations, N-methyl-2-pyrrolidinone,<br />

Transcutol, propylene glycol and terpenes like citral, geraniol and<br />

eugenol were used as penetration enhancers. The flux values of both<br />

drugs significantly increased in the presence of all penetration enhancers,<br />

except Transcutol and propylene glycol.<br />

All data are summarized in (Figure 61).<br />

130


Table 35: In vitro release of glibenclamide from water soluble base containing different concentrations of Transcutol.<br />

Glibenclamide released % ± (sd)<br />

Time (min)<br />

0% 5% 8%<br />

0 0 ± 0 0 ± 0 0 ± 0<br />

30 0.68 ± 0 1.37 ± 0.04 1.05 ± 0.07<br />

60 1.5 ± 0.09 2.38 ± 0.017 1.82 ± 0.07<br />

90 1.71 ± 0.25 3.05 ± 0.14 2.350± 0<br />

120 2.25 ± 0.07 3.49 ± 0.19 2.82 ± 0<br />

150 2.68 ± 0.14 4.04 ± 0.28 3.29 ± 0. 03<br />

180 3.44 ± 0.12 5.4± 0.18 4.17 ± 0.07<br />

240 4.8 ± 0.25 5.65 ± 0.25 4.95 ± 0.14<br />

300 5.29 ± 0.11 6.99 ± 0.15 5.39 ± 0.13<br />

360 5.94 ± 0.41 7.25 ± 0.25 6.5 ± 0.33<br />

131


Drug alone 5% Tc 8% Tc<br />

12<br />

11<br />

10<br />

9<br />

8<br />

7<br />

6<br />

5<br />

% Drug released<br />

4<br />

3<br />

2<br />

1<br />

0<br />

0 50 100 150 200 250 300 350 400<br />

Time (min)<br />

Figure 59: Release profile of glibenclamide from water soluble base containing different concentrations of<br />

Transcutol.<br />

132


12<br />

11<br />

IPM 1%<br />

10<br />

Drug alone<br />

9<br />

IPM 0.5%<br />

8<br />

IPM 1%<br />

Tc 5%<br />

IPP 0.2% IPP1%<br />

IPP 2%<br />

IPM 2%<br />

7<br />

IPM 2%<br />

Tc 8%<br />

IPM 0.5%<br />

Drug alone<br />

6<br />

IPP 0.2%<br />

IPP1%<br />

5<br />

% Drug released<br />

IPP 2%<br />

4<br />

Tc 5%<br />

3<br />

Tc 8%<br />

2<br />

1<br />

0<br />

Different enhancers with different concentrations<br />

Figure 60: Percentage drug released from water soluble base containing different concentrations of fatty acid<br />

esters and Transcutol.<br />

133


10<br />

Drug alone<br />

Tw 80 4%<br />

Cetrimide 0.5%<br />

9<br />

Cetrimide<br />

0.5%<br />

SLS 0.4%<br />

SLS 0.4%<br />

Urea 5%<br />

LOA 0.8%<br />

OA 2%<br />

8<br />

Lab 3% Tc 5%<br />

Tw 80 4%<br />

Labrazol 5%<br />

IPM 2% IPP2%<br />

7<br />

Lab 3%<br />

Drug alone<br />

6<br />

Tc 5%<br />

5<br />

IPM 2%<br />

IPP2%<br />

4<br />

% Drug released<br />

OA 2%<br />

3<br />

LOA 0.8%<br />

2<br />

Urea 5%<br />

1<br />

Labrazol 5%<br />

0<br />

Differtent penetration enhancers<br />

Figure 48: Percentage drug released from water soluble base containing different concentrations of different<br />

enhancers.<br />

134


7. Kinetic analysis of release data:<br />

As shown in (Table 36) the release data of Glib from all different<br />

topical formulations followed diffusion controlled mechanism (Higuchi<br />

model).<br />

8. In vitro permeation of gliclazide through abdominal rat skin:<br />

Skin permeation studies indicated that Glib permeation through<br />

hairless rabbit skin was negligible. The possible reasons for this result<br />

may be i) Glib , a lipophilic drug, was retained within the stratum<br />

corneum with no portioning into the viable epidermis or ii) most of the<br />

drug was used up to saturate the binding sites in the skin and the<br />

remaining drug was probably insufficient to provide a significant<br />

concentration gradient (Srini et al., 1998).<br />

135


Table 36: Kinetic data of the release of Glib from different topical<br />

bases.<br />

Topical<br />

Cont. table 36: Kinetic data of the release of Glib from different<br />

topical bases.<br />

Correlation coefficient (R)<br />

preparation Zero First Diffusion<br />

Topical bases<br />

WSB-SLS<br />

WSB-<br />

WSB-Tw -80<br />

WSB-Tc<br />

Cetrimide<br />

136<br />

Observed<br />

order<br />

WSB 0.9833 0.9784 0.9842 D.M<br />

HPMC gel 0.9356 0.9375 0.9694 D.M<br />

HPMC emulgel 0.9622 0.9642 0.9849 D.M<br />

Sod-alginate gel 0.9804 0.9814 0.9852 D.M<br />

O/W cream 0.9196 0.9205 0.9527 D.M<br />

Oleaginous base 0.8095 0.8102 0.8960 D.M<br />

Absorption base 0.8859 0.8865 0.8904 D.M<br />

0.1% SLS 0.9813 0.9881 0.9891 D.M<br />

0.4% SLS 0.9784 0.9857 0.9991 D.M<br />

0.8% SLS 0.9652 0.9674 0.9855 D.M<br />

0.3% cetrimide 0.9742 0.9768 0.9910 D.M<br />

0.5% cetrimide 0.9521 0.9573 0.9956 D.M<br />

1% cetrimide 0.9766 0.9803 0.9981 D.M<br />

2% cetrimide 0.9789 0.9804 0.9917 D.M<br />

0.3% Tw-80 0.980 0.9818 0.9954 D.M<br />

1% Tw-80 0.9801 0.9820 0.9974 D.M<br />

4% Tw-80 0.9722 0.9761 0.9987 D.M<br />

5% Tw-80 0.9781 0.9804 0.9962 D.M<br />

5% Tc 0.9806 0.9820 0.9859 D.M<br />

8% Tc 0.9836 0.9849 0.9881 D.M


Topical<br />

preparation<br />

WSB-<br />

WSB-oleic<br />

WSB-linoleic<br />

WSB-IPP<br />

WSB-IPM<br />

labrafil<br />

acid<br />

acid<br />

Correlation coefficient (R)<br />

Zero First Diffusion<br />

137<br />

Observed<br />

order<br />

3% lab 0.9063 0.9109 0.9676 D.M<br />

5 % lab 0.9729 0.9685 .9757 D.M<br />

7 % lab 0.9668 0.9685 0.9778 D.M<br />

0.5 % OA 0.9418 0.9458 0.9906 D.M<br />

1% OA 0.9614 0.9659 0.9970 D.M<br />

2% OA 0.9521 0.9553 0.9870 D.M<br />

3% OA 0.9855 0.9808 0.9808 D.M<br />

0.8 % LOA 0.9752 0.9774 0.9895 D.M<br />

1 % LOA 0.9728 0.9755 0.9991 D.M<br />

2% LOA 0.9773 0.9791 0.9897 D.M<br />

0.2% IPP 0.9569 0.9594 0.9840 D.M<br />

1% IPP 0.9793 0.9819 0.9979 D.M<br />

2% IPP 0.9662 0.9692 0.9965 D.M<br />

0.5% IPM 0.9834 0.9851 0.9950 D.M<br />

1% IPM 0.9636 0.9688 0.9971 D.M<br />

2% IPM 0.9556 0.9587 0.9918 D.M


9- In- vivo study:<br />

The result of hypoglycemic activity of the topically applied glibenclamide<br />

and oral glibenclamide (5 mg/kg; p.o.) in both normal and diabetic rats<br />

are shown in (Table 37-41) and (Figures 61-62).<br />

The blood glucose reducing effect was significant in oral and all<br />

topically treated groups up to 24 h except groups treated with ointment<br />

contained 5% Labrafil, compared with control group (p<br />

*** Studies in normal rats<br />

Glib (oral) produced a significant decrease of 58.09 % ± 1.5 (p<br />

0.05 compared to control) in blood glucose levels at 2 hr and then the<br />

blood glucose levels decreased. The percentage reduction in the blood<br />

glucose levels at the end of 24 hr were 30.61 ± 3.4. On other hand, the<br />

blood glucose reducing response of all topical formulation was gradual<br />

and increased slowly up to 24 h.<br />

The effect of OA and cetrimide in amount of 1% within the Glib<br />

ointment on reducing the blood glucose level is shown in (Figure 62).<br />

OA and cetrimide increased significantly the blood glucose reducing<br />

activity of glib<br />

OA is a popular penetration enhancer and penetrates into the stratum<br />

corneum and decompresses this layer and hence reduces its' resistance to<br />

drug penetration (Barry and Bennett, 1987). OA can also accumulate<br />

within the lipid bilayers of stratum corneum cells and hence increase their<br />

flowability and penetration ability (Goodman and Barry, 1988).<br />

138


Table 37: Reduction in blood glucose level after oral and topical application of glibenclamide and glibenclamide with<br />

1% oleic acid in normal rats. All values are expressed as mean ± sd.<br />

.<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Absolute<br />

blood<br />

glucose level<br />

Group<br />

(mg/dl) 2hr 4hr 6hr 8hr 24hr<br />

71 ± 2.94<br />

(24.64 ± 1.5)<br />

77.5 ± 5.74<br />

(17.59 ±2.1)<br />

84.5 ± 2.08<br />

(10.53 ±1.84)<br />

84 ± 2.4<br />

(11.05± 2.49)<br />

94.5 ± 3.97 93.25 ± 3.4*<br />

(1.29±0.4)**<br />

Control<br />

(1ml gum<br />

acacia<br />

suspension)<br />

69.5 ± 4.2<br />

(30.61 ±3.4)<br />

59.25 ± 0.95<br />

(42.53 ±4.3)<br />

51.75± 4.3<br />

(49.9 ± 4.06)<br />

49.75 ± 5.3<br />

(50.48 ±6.6)<br />

103.75 ± 8.9 43 ± 4.4<br />

(58.09 ± 1.5)<br />

Oral<br />

glibenclamide<br />

(5mg/kg)<br />

51 ± 6.27<br />

(48.91 ± 4.2)<br />

57.25 ± 8.8<br />

(41.54 ± 3.6)<br />

66.00 ± 9.1<br />

(32.55± 2.14)<br />

69.25 ± 8.53<br />

(29.13± 1.75)<br />

97.75± 12.12 74.25± 9.06<br />

(23.99± 2.84)<br />

WSB<br />

(glibenclamide<br />

40.75 ± 6.84<br />

(56.86 ± 3.8)<br />

56.5 ± 7.3<br />

(40.04 ±3.7)<br />

59.5 ± 6.6<br />

(36.68 ± 2.7)<br />

60.75 ± 5.9<br />

(35.26 ± 3.2)<br />

94.5± 13.1 69 ± 6.3<br />

(26.95 ±5.7)<br />

WSB<br />

(glibenclamide<br />

+1% oleic<br />

acid)<br />

* Reduction in blood glucose level (mg/dl). **Percentage reduction in blood glucose levels.<br />

139


Table 38 : Reduction in blood glucose level after oral and topical application of glibenclamide and glibenclamide<br />

with 1% cetrimide in normal rats. All values are expressed as mean ± sd.<br />

. .<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Group<br />

2hr 4hr 6hr 8hr 24hr<br />

Absolute<br />

blood<br />

glucose<br />

level<br />

(mg/dl)<br />

71 ± 2.94<br />

(24.64± 1.5)<br />

77.5 ± 5.74<br />

(17.59 ±2.1)<br />

84.5 ± 2.08<br />

(10.53 ±1.84)<br />

84 ± 2.4<br />

(11.05 ± 2.49)<br />

94.5 ± 3.97 93.25 ± 3.4*<br />

(1.29±0.46)**<br />

Control<br />

(1ml gum<br />

acacia<br />

suspension)<br />

69.5 ± 4.2<br />

(30.61 ±3.4)<br />

59.25 ± 0.95<br />

(42.53 ±4.3)<br />

51.75± 4.3<br />

(49.9 ± 4.06)<br />

49.75 ± 5.3<br />

(50.48 ±6.6)<br />

103.75± 8.9 43 ± 4.4<br />

(58.09 ± 1.5)<br />

Oral<br />

glibenclamide<br />

(5mg/kg)<br />

51 ± 6.27<br />

(48.91± 4.2)<br />

57.25 ± 8.8<br />

(41.54 ± 3.6)<br />

66.00 ± 9.1<br />

(32.55 ± 2.14)<br />

69.25 ± 8.53<br />

(29.13 ± 1.75)<br />

74.25± 9.06<br />

(23.99± 2.84)<br />

97.75±<br />

12.12<br />

WSB<br />

(glibenclamide<br />

45.5 ± 6.3<br />

(60.9± 3.33)<br />

52.66 ± 6.8<br />

(54.54 ±3.17)<br />

60.75 ± 5.67<br />

(47.39 ± 3.3)<br />

70.33 ± 4.9<br />

(39.07 ± 2.4)<br />

115.5± 7.5 87.25 ±3.3<br />

(24.32 ±2.8)<br />

WSB<br />

(glibenclamide<br />

+1%<br />

cetrimide)<br />

*<br />

Reduction in blood glucose level (mg/dl). **Percentage reduction in blood glucose levels.<br />

140


Table 39 : Reduction in blood glucose level after oral and topical application of glibenclamide and glibenclamide<br />

with 1% isopropyl myristate (IPM) in normal rats. All values are expressed as mean ± sd.<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Absolute<br />

blood glucose<br />

level (mg/dl)<br />

Group<br />

2hr 4hr 6hr 8hr 24hr<br />

71 ± 2.94<br />

(24.64 ±<br />

1.5)<br />

77.5 ± 5.74<br />

(17.59 ±2.1)<br />

84.5 ± 2.08<br />

(10.53 ±1.84)<br />

84 ± 2.4<br />

(11.05 ± 2.49)<br />

94.5 ± 3.97 93.25 ± 3.4*<br />

(1.29±0.46)**<br />

Control<br />

(1ml gum<br />

acacia<br />

suspension)<br />

69.5 ± 4.2<br />

(30.61 ±3.4)<br />

59.25 ± 0.95<br />

(42.53 ±4.3)<br />

51.75± 4.3<br />

(49.9 ± 4.06)<br />

49.75 ± 5.3<br />

(50.48 ±6.6)<br />

103.75 ± 8.9 43 ± 4.4<br />

(58.09 ± 1.5)<br />

Oral<br />

glibenclamide<br />

(5mg/kg)<br />

51 ± 6.27<br />

(48.91 ±<br />

4.2)<br />

57.25 ± 8.8<br />

(41.54 ± 3.6)<br />

66.00 ± 9.1<br />

(32.55 ± 2.14)<br />

69.25 ± 8.53<br />

(29.13 ± 1.75)<br />

97.75 ± 12.12 74.25± 9.06<br />

(23.99± 2.84)<br />

WSB<br />

(glibenclamide<br />

50.5 ± 6.5<br />

(47.52 ±<br />

4.2)<br />

62.75 ± 12.89<br />

(35.26 ±3.2)<br />

69 ± 12.83<br />

(28.82± 2.38)<br />

78.25 ± 13.37<br />

(19.13 ± 3.23)<br />

97± 17.9 93 ± 17.75<br />

(4.16 ±0.93)<br />

WSB<br />

(glibenclamide<br />

+1% IPM)<br />

*<br />

Reduction in blood glucose level (mg/dl). **Percentage reduction in blood glucose levels.<br />

141


.Table 40 : Reduction in blood glucose level after oral and topical application of glibenclamide and glibenclamide<br />

with 5 % Labrafil in normal rats. All values are expressed as mean ± sd.<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Absolute<br />

blood glucose<br />

level (mg/dl)<br />

Group<br />

2hr 4hr 6hr 8hr 24hr<br />

71 ± 2.94<br />

(24.64 ±<br />

1.5)<br />

77.5 ± 5.74<br />

(17.59 ±2.1)<br />

84.5 ± 2.08<br />

(10.53 ±1.84)<br />

84 ± 2.4<br />

(11.05 ± 2.49)<br />

94.5 ± 3.97 93.25 ± 3.4*<br />

(1.29±0.46)**<br />

Control<br />

(1ml gum<br />

acacia<br />

suspension)<br />

69.5 ± 4.2<br />

(30.61<br />

±3.4)<br />

59.25 ± 0.95<br />

(42.53 ±4.3)<br />

51.75± 4.3<br />

(49.9 ± 4.06)<br />

49.75 ± 5.3<br />

(50.48 ±6.6)<br />

103.75 ± 8.9 43 ± 4.4<br />

(58.09 ± 1.5)<br />

Oral<br />

glibenclamide<br />

(5mg/kg)<br />

51 ± 6.27<br />

(48.9± 4.2)<br />

57.25 ± 8.8<br />

(41.54 ± 3.6)<br />

66.00 ± 9.1<br />

(32.55 ± 2.14)<br />

69.25 ± 8.53<br />

(29.13 ± 1.75)<br />

97.75 ± 12.12 74.25± 9.06<br />

(23.99± 2.84)<br />

WSB<br />

(glibenclamide<br />

75 ± 6.97<br />

(27.0±<br />

2.96)<br />

83.75 ± 9.7<br />

(18.017 ±2.5)<br />

95 ± 5.9<br />

(7.41 ± 2.14)<br />

95 ± 6.6<br />

(6.9 ± 2.5)<br />

102.75± 8.5 96.75 ± 6.3<br />

(5.7 ±1.9)<br />

WSB<br />

(glibenclamide<br />

+5% Labrafil)<br />

. *<br />

Reduction in blood glucose level (mg/dl). **Percentage reduction in blood glucose levels.<br />

142


.<br />

Control Oral glibenclamide Topical glibenclamide<br />

1% OA 1% Cetrimide 1% IPM<br />

5% Labrafil<br />

70<br />

60<br />

50<br />

40<br />

30<br />

20<br />

% Reduction in blood glucose level<br />

10<br />

0<br />

0 2 4 6 8 10 12 14 16 18 20 22 24 26<br />

Time (hr)<br />

Figure 62: Percent reduction in blood glucose levels after oral and topical administration of glibenclamide in normal<br />

rats.<br />

143


Cetrimide is a cataionic surfactant. It has a potential to solubilise<br />

lipids within the stratum corneum, swell the stratum corneum and interact<br />

with intercellular keratin so increase the permeation of the drug<br />

(Williams and Barry, 2004).<br />

Incorporation of both 1% IPM and 5% Labrafil did not show any<br />

enhancement in the blood glucose reducing activity (compared to Glib<br />

without enhancer).<br />

*** Studies in diabetic rats:<br />

Oral and topical groups showed significant hypoglycemic activity<br />

upto 24 hrs. The hypoglycemic effect produced by ointement containing<br />

Glib and 1% cetrimide in the animals is significantly less when compared<br />

to oral administration.<br />

Glib (oral) produced a significant decrease of 41.1 ± 5.25 (p<br />

compared to control) in blood glucose levels at 4 hr and then the blood<br />

glucose levels increased. On other hand, the blood glucose reducing<br />

response of topical formulation was gradual and increased slowly up to<br />

24 h.<br />

The results did not differ significantly in oral and topical groups<br />

after 24 hrs. The topically applied Glib and the oral drug produced<br />

decrease of 24.53 ± 3.74 and 25.7 ± 4.69 respectively, in the blood<br />

glucose levels after 24 hrs. This may be due to reduced insulin levels in<br />

diabetic models impairs principal metabolic pathways of sulphonylurea<br />

which resulted in its prolonged action in orally treated group (Stroev and<br />

Belkina, 1989).<br />

144


Table 41 : Reduction in blood glucose level after oral and topical application of glibenclamide and glibenclamide<br />

with 1% cetrimide in diabetic rats. All values are expressed as mean ± sd.<br />

.<br />

Reduction in blood glucose level (mg/dl)<br />

(Percentage reduction in blood glucose levels)<br />

Group<br />

2hr 4hr 6hr 8hr 24hr<br />

Absolute<br />

blood<br />

glucose<br />

level<br />

(mg/dl)<br />

223.4 ± 40.8<br />

(5.22 ± 2.74)<br />

224.4 ± 38.8<br />

(4.76±0.89)<br />

230.6 ± 40.8<br />

(2.16 ±1.68)<br />

232.2 ± 38.46<br />

(1.35 ± 1.06)<br />

228.4 ± 36.26*<br />

(2.89 ± 1.65)**<br />

235.6 ±<br />

40.3<br />

Control<br />

(1ml gum<br />

acacia<br />

suspension)<br />

364.8± 49.93<br />

(24.53 ±3.74)<br />

342 ± 51.63<br />

(29.25<br />

±3.75)<br />

318 ± 45.06<br />

(34.02± 5.67)<br />

283 ± 33.54<br />

(41.1 ±5.25)<br />

420.2 ± 62.81<br />

(15.63 ± 3.09)<br />

485 ±<br />

79.56<br />

Oral<br />

glibenclamide<br />

(5mg/kg)<br />

236.8 ± 23.53<br />

(25.7 ± 4.69)<br />

267.5± 25.99<br />

(16.16 ±2.7)<br />

277.2 ± 30.82<br />

(13.28± 3.5)<br />

286.7± 35.12<br />

(9.6 ± 4.4)<br />

319± 23.2 305± 21.37<br />

(4.28± 2.4)<br />

WSB<br />

(glibenclamide<br />

+1%<br />

cetrimide)<br />

. * Reduction in blood glucose level (mg/dl). **Percentage reduction in blood glucose levels.<br />

145


50<br />

Control<br />

Oral glibenclamide<br />

Topical glibenclamide<br />

45<br />

40<br />

35<br />

30<br />

25<br />

20<br />

15<br />

10<br />

% Reduction in blood glucose level(mg/dl)<br />

5<br />

0<br />

0 2 4 6 8 10 12 14 16 18 20 22 24 26<br />

Time (hr)<br />

Figure 63 : Percent reduction in blood glucose levels after oral and topical administration of glibenclamide in<br />

diabetic rats.<br />

146


Conclusion:<br />

From the previously demonstrated data the following results can be<br />

concluded:<br />

1- Glib has a lipophilic property.<br />

2- The percentage amount of drug released from water soluble base, gel<br />

bases and emulgel base are greater than that released from other bases.<br />

The rate of drug release can be arranged in the following descending<br />

order:<br />

Water soluble base (5.94 %) > HPMC emulgel (4.6 %) > sodium alginate<br />

gel (4.38 %) > HPMC gel (3.99) >O/W emulsion base (2.5 %) ><br />

absorption base (1.94%) > oleaginous base (1.61%).It is clear that,<br />

water soluble base showed the highest release than that of emulsion,<br />

gels, emulgel, oleaginous and absorption bases.<br />

3- The investigation showed the effect of addition of penetration<br />

enhancers on the amount of Glib released from different topical bases<br />

in vitro can be arranged in the follwing descending order:<br />

1% IPM > 5% Lab > 1% Cetrimide > 4% Tw-80 > 1% OA > 0.2%<br />

IPP > 0.8% LOA > 0.4% SLS > 5% Tc.<br />

4- Topically applied glibenclamide exhibited better control of<br />

hyperglycemia and more effectively reversed the glibenclamide side<br />

effects than oral glibenclamide administration in both normal and<br />

diabetic rats. Slow and sustained release of the drug from the<br />

transdermal system might reduce<br />

manifestations like severe hypoglycemia, sulphonylurea receptor down<br />

regulation and the risk of chronic hyperinsulinemia ( Mutalik and<br />

Udupa, 2004).<br />

147


Ointments contained 1% cetrimide and 1% OA enhanced the blood<br />

glucose reducing activity of glibenclamide . While addition of 1% IPM<br />

and 5% Lab did not show any enhancement in the blood glucose reducing<br />

activity (compared to glib without enhancer).<br />

148


General Conclusion<br />

The preceding study was an attempt to evaluate the potential of<br />

pharmaceutical formulation of certain sulfonylureas namely, gliclazide<br />

and glibenclamide in different bases for topical application.<br />

In case of gliclazide, the amount of drug released from topical bases<br />

incorporating solid dispersions can be arranged in the following<br />

descending order. Topical preparations containing (8:92) PEG 6000 ><br />

(1:10) glucose > (8:92) PEG 4000 > (1:10) urea solid dispersions > pure<br />

drug.<br />

The blood glucose reducing activity of ointment contained (10:90)<br />

gliclazide –PEG 6000 solid dispersions was significantly more when<br />

compared to ointment contained gliclazide alone.<br />

In case of glibenclamide, the presence of various penetration<br />

enhancers increase the amount of drug released from the topical base in<br />

vitro. The maximum release was obtained by using IPM (1%).<br />

Ointments contained 1% cetrimide and 1% oleic acid enhanced the<br />

blood glucose reducing activity of glibenclamide . While addition of 1%<br />

isopropyl myristate and 5% Labrafil did not show any enhancement in the<br />

blood glucose reducing activity of glibenclamide.<br />

In conclusion, it was demonstrated that sulfonylureas were absorbed<br />

through the skin and lowered the blood glucose levels. The results<br />

suggest the possibility of transdermal administration of sulfonylureas for<br />

the treatment of NIDDM.<br />

149


A<br />

Aboofazeli, R., Zia, H. and Needham, T.E., Drug Deliv., (2002), 9,<br />

239–247.<br />

Abu T.M. Serajuddin, Advanced Drug Delivery Reviews (2007).<br />

Agharkar, S., Lindenbaum, S., Higuchi, T., J. Pharm.Sci., (1976), 65,<br />

747-749.<br />

Ainsworth, M., J. Soc. Cosmetic Chemists, (1960), 11. 69.<br />

Al-Angary, A.A., Al-Mahrouk, G.M. and Al-Meshal, M.A., Pharm.<br />

Ind., (1996), 58(3), 260.<br />

Al-Suwayeh, S.A., Saudi Pharmaceutical Journal, (2003), 11(1-2), 46.<br />

Ambike, A.A., Mahadik, K.R., Paradkar, A., Int.J. Pharm., (2004),<br />

282(1), 151-162.<br />

Amin, K., Dannenfelser, R. M., J. Zielinski, Wang, B., J. Pharm. Sci.,<br />

(2004), 93, 2244-2249.<br />

Ammar H.O., Ghorab M., El-Nahas, S., Int. J. Pharm., (2006), 327, 81-<br />

88.<br />

Ammar, H. O., Ghorab, M., El-Nahhasa, S. A., Kamela, R., Asian<br />

J. Pharm. Sci., (2007), 2 (3), 96-105.<br />

Anil, J. S., Kevin, C.G., Namdeo, R. J. and Harinath, M.N., J.<br />

Pharmaceutical Reviews, (2007), 5, Issue 6.<br />

Anshel, J. and Lieberman, H.A., In The Theory and Practice of<br />

Industrial Pharmacy, Ist edn., Lachman, L., Lieberman, H.A. and Kany,<br />

J.L. (eds.), Lee and Febiger, Philadelphia, (1976), 242-249.<br />

Arias, M.J., Gines, J.M., Moyano, J.R. and Rabasco, A.M., Pharm.<br />

Acta Helv.,(1996), 71, 229.<br />

150


Arias, M.J., Gines, J.M., Moyano, J.R., Rabasco, A.M., Int.J.Pharm.,<br />

(1995), 123, 25-31.<br />

Asada, M., Takashashi, H., Okamoto, H., Tanino, H., Danjo, K.,<br />

Int.J.Pharm., (2004), 270(1), 167-174.<br />

Astley, J.P., and Levine, M., J. Pharm. Sci., (1976), 65, 210.<br />

B<br />

Babu, G.V., Prasad, C.D. and Murthy, K.V., Int J. Pharm., (2002), 234<br />

(1-2), 1.<br />

Balata, G.F., M.sc. pharm. Thesis, Faculty of pharmacy, Zagazig Univ. Egypt,<br />

(1999).<br />

Barr, M., J.Pharm. Sci., (1962), 51, 395-400.<br />

Barry, B.W., Eur. J. Pharm. Sci., (2001), 14, 101-14.<br />

Barry, B.W., In: Dermatological formulation in percutaneous absorption<br />

Marcel Dekker, New York (1983), 1-334.<br />

Barry, B.W., Molec. Aspects. Med., (1991), 12, 195-241.<br />

Barry, B.W and Bennett, L., J. Pharm. Pharmacol. (1987), 39: 535-548<br />

Barthélémy, P., Farah, N., Laforet, J.P., Transcutol-Product profile.<br />

Product Information, Gattefossé, (1995),1.<br />

Behl, C.R., Flynn, G.L., Kurihara, T., Harper, N., Smith, W.,<br />

Higuchi, W.I., Ho, N.F., Pierson, C.L. J. Invest. Dermatol., (1980), 75, 346-<br />

52.<br />

Bentley, M.V., Vianna, R.F., Wilson, S., Collett, J.H. J. Pharm.<br />

Pharmacol., (1997), 49, 397-402.<br />

Bharti D. Shewale, Ravindra A. Fursule and Nidhi P. Sapkal,<br />

International Journal of Health Research, (2008), 1(2): 95-99.<br />

Biana, G., Touitou, E. Crit. Rev. Ther. Drug Carrier Syst., (2003), 20, 63-102.<br />

Bikiaris, D., Papageorgiou, G. Z., Stergiou, A., Pavlidou, E.,<br />

Karavas, E., Kanaze F. and Georgarakis, M., Thermochim. Acta.,<br />

(2005), 439, 58–67.<br />

151


Bitzen, P.O., Melander, A. and Schersten, B., Eur. J. Clin. Pharmacol<br />

(1992), 83, 42-77.<br />

Bonina, F. P. and Montenegro, L., Int. J. Pharm., (1992), 82, 171.<br />

Bos, J.D., Meinardi, M.M. Exp. Dermatol., (2000), 9, 165-9.<br />

Bottari, F., Di Colo, G., Nannipieri, E., Saettone, M.F., and Serfaini, M.F., J.<br />

Pharm. Sci., (1977), 66, 926.<br />

Bradshaw, M., Am. Perfumer, (1961), 76, 15.<br />

Breitenbach, J., Eur. J. Biopharm., (2002), 54(2), 107.<br />

Brewster, M.E. and Loftsson, T., Pharmazie, (2002), 57(2), 94.<br />

Bronaugh, R.L.; Stewart, R.F.; Storm, J.E. Extent of cutaneous<br />

metabolism during percutaneous absorption of xenobiotics. Toxicology<br />

and Applied Pharmacology.(1989), 99: 534-543.<br />

Butler, J.A., Br. J. Dermatol., (1966), 78, 665.<br />

C<br />

Castellan, G.W., In: Physical Chemistry, Addison- Wesley, Menlo Park, (1983), 324-<br />

336.<br />

Catham, S.M., S.T.P. Pharm., (1987), 3, 575.<br />

Celebi, N., Kislal, O. and Tarimci, N., Pharmazi, 48, (1993) , 12.<br />

Ceschel, G.C., Maffei, P., and Gentile, M., (1999), Drug Dev. Ind.<br />

Pharm., 125(9), 1035-1039.<br />

Cevc, G., Crit. Rev. Ther. Drug Carrier Syst., (1996), 13, 257-388.<br />

Cevc, G., "Transferosomes: Innovative Transdermal Drug Carriers," in<br />

"Modified Release drug delivery Technology," M.J.Rathbone, J.,<br />

Hadgraft, M.S., Roberts, Eds., Marcel Dekker Inc., New York, NY,<br />

(2003), 533.<br />

Chakole C. M., Shende M. A and Khadatkar S.N., Int. J. Chem. Tech.<br />

Research, (2009), 1,103-116.<br />

Chatterjee, D. J., Li and Koda, R. T., Pharm. Res. (1997), 14, 1058.<br />

152


Chauhan, B. et al., Chauhan B, Shimpi S, Paradkar A., Eur. J. Pharm.<br />

Sci., (2005), 26, 219–230.<br />

Chaumeil, J.C., Micronisation: a method of improving the<br />

bioavailability of poorly solubledrugs, Methods and Findings in<br />

Experimental and Clinical Pharmacology, (1998), 20,211-215.<br />

Chayen, J., Bitensky, L., Butcher, R.G., Poulter, L.W., and Ubhi, G.S., Br. J.<br />

Dermatol., (1970), 82(suppl.6), 62.<br />

Chi, H.L. and Choi, H.K., AAPS Pharm. Sci. Tech., (2000), 1(2), 13.<br />

Chine, Y.W., (ed.), "Transdermal controlled release drug<br />

administration," In "Fundamentals, Development Concepts, Biomedical<br />

assessments," (1982) 1 st edn., Vol. (14), Marcel Dekker, New York, pp.<br />

179-215.<br />

Chine, Y.W., "Transdermal Therapeutic Systems," In "Controlled Drug<br />

Delivery: Fundamentals and Applications," (1987), J.R., Robonson and<br />

V.H.L., Lee, Eds., 2 nd ed., Marcel Dekker, Inc., New York, pp. 523.<br />

Chiou,W.L., Riegelman, S., J. Pharm. Sci. (1969), 56, 1505-1510.<br />

Chiou,W.L., Riegelman, S., ibid. (1971), 58, 1281-1302.<br />

Choi, H-G., Rhee, J-D., Yu, B-K. and Yong, C-S., J. Kor., Pharm. Sci.,<br />

(2003), 33(1), 29.<br />

Choi, Y-G., Cui, Y., Kim, K-N., Park, E-S. and Chi,S-C., ibid, (2003),<br />

33(2), 129.<br />

Chutimaworapan, S., Ritthidej, G.C., Yonemochi, E., Oguchi, T. and Yamamoto,<br />

K., Drug Dev. and Ind. Pharm., (2000), 26(11), 1141.<br />

Collet, D.M., "Ointment pastes and gels," In " Pharmaceutical Practice," 1 st edn.,<br />

Chapter (4), Michael, E.A. and Collet, D.M. (ed.), Longman Pte Ltd., Singapore,<br />

(1991) pp. 125-133.<br />

Cormier, M., and Daddona, P.E., "Macroflux Technology for<br />

transdermal Delivery of Therapeutic Protins and Vaccines," in "Modified<br />

Release Drug Delivery Technology," M.J.Rathbone, J., Hadgraft, M.S.,<br />

Roberts, Eds., Marcel Dekker Inc., New York, NY, 2003, 590.<br />

153


Corrigan, O.I., Murphy, C.A., Timony, R.P., Drug Dev. Ind. Pharm.,<br />

(1979), 4(1), 67-74.<br />

Craig, D.Q.M., Int. J. Pharm., (2002), 231, 131–144.<br />

Craig, D.Q.M., Drug Dev. and Ind. Pharm., (1990), 16, 2514.<br />

Cutler, L., Howes C., Deeks N. J., Buck T. L. , Jeffrey P., J. Pharm.<br />

Sci., (2006), 95, 1944–1953.<br />

D<br />

Davaran, S.; Rashidi, M.R.; Hashemi, M., J. Pharm. Pharmacol.,<br />

(2003), 55, 513-7.<br />

Dhavse V.V., Amin P.D., East Pharm. (1997), 133– 135.<br />

Di colo, G., Carelli, V., Giannaccini, B., Serafini, M.F., and Bottari, f., J. Pharm.<br />

Sci., (1980), 69, 387.<br />

Dipolo. R., Sha'afi, R.I., and Solomon, A.K., J. Gen. Physiol., (1970), 55, 63.<br />

Domain, F., Blaton, N., Naesens, L., Balzarini, J., Kinget, R., Augustijns, P. and<br />

Mooter, G.V., Eur. J. Pharm. Sci., ( 2000), 10, 313.<br />

Domb, A.J., Int.J.Pharm.Sci.,(1995), 124, 271.<br />

Doshi, D.H, Ravis, W. R. And Betageri, G.V., Drug Develop. Indust.<br />

Pharm., (1997), 23(12), 1167-1176.<br />

Duncan, J. I., Payne, S. N., Winfield, A. J., Ormerod, A.D. and<br />

Thomson, A. W., Br. J. Dermatol., (1990), 123, 631.<br />

E<br />

Egbaria, K., Ramachandran, C., Kittayanond, D., Weiner, N.<br />

Antimicrob. Agents Chemother., (1990), 34, 107-10.<br />

Ehrenstrom Reiz, G.M. and Reiz, S.L., Acta Anaesthesiol. Scand.,<br />

(1982), 26, 596-8.<br />

Elias, P.M., Cooper, E.R., Korc, A., and Brown, B.E., J. Invest. Dermatol., (1981),<br />

76, 297.<br />

154


El-Megrab, N.A., El-Nahas, H.M. and Balata,G.F. Saudi<br />

Pharmaceutical Journal, (2006), 14, Nos. 3-4, 157.<br />

El-Nahas, H.M. PhD. Thesis, Faculty of pharmacy, Zagazig Univ. Egypt, (2001).<br />

El-Nokaly M., and Cornell, D., "Microemulsion and Emulsions in foods,"<br />

Developed from a symposium sponsored by the Division of Agriculture and food<br />

chemistry at 19 th National meeting of the American Chemical Society, Boston,<br />

(1990).<br />

El-Sayed, Y.M., Suleiman, M.S., Hassan, M.M., Abdel-Hamid, M.E.,<br />

Najib, N.M., Sallam, E.S. and Shubair, M.S., Int. J. Clin Pharmacol<br />

Ther Toxicol, (1989), 27, 551-557.<br />

Emara, L.H., Badr, R.M. and Abd El-bary, A., Drug Dev. and Ind. Pharm., (2002),<br />

28(7), 795.<br />

Etman, A.M., Alex. J. Pharm. Sci., (2000), 14(1), 79.<br />

Ezzedeen, F.W., Shihab, F.A. and Stohs, S.J., Inter. J. Pharm.,<br />

(1986), 28, 113-117.<br />

F<br />

Faber, O.K., Beck-Nielsen, H. and Binder, C. Diabetes Care (1990), 31,<br />

13-26.<br />

Fang, J.Y. Wu, P.C., Huang, Y.B. and Tsai, Y.H., Int J. Pharm.,<br />

(1996), 128, 169-177.<br />

Fang, J.Y., Hawang, T.L. and Leu, Y.L., (2003), Int. J.Pharm.,250,313-325.<br />

Fergany, A.M., Drug Dev. Ind. Pharm., (2001), 27(10), 1083.<br />

Fincher, T. K., Yoo, S. D., Player, M. R., Sowell, J. w. and<br />

Minchaniak, B. B., J. Pharm. Sci., (1996), 85, 920.<br />

Fini,A., Moyano, J.R., Ginés, J.M., Martinez, J.I., Rabasco, A.M.,<br />

Eur. J. Pharm. Biopharm., (2005), 117-122.<br />

Fledmann, R.J., and Maibach, H.I., Arch. Dermatol., (1965), 91, 661.<br />

Fledmann, R.J., and Maibach, H.I., ibid, (1966), 94, 649.<br />

Fledmann, R.J., and Maibach, H.I., J. Invest. Dermatol., (1967), 48, 181.<br />

Fledmann, R.J., and Maibach, H.I., ibid, (1969), 52, 89.<br />

155


Fledmann, R.J., and Maibach, H.I., ibid, (1968), 50, 351.<br />

Fledmann, R.J., and Maibach, H.I., ibid, (1970), 54, 399.<br />

Flynn, G.L. and Roseman, T.J., ibid, (1971), 60, 1788.<br />

Ford, J.L. and Timmins, P., In Pharmaceutical Thermal Analysis, Technique and<br />

Applications, Ellis Horwood Limited, (1989).<br />

Forster, A., Rades, T. and Hempenstall, J., Pharm. Technol. Eur., (2002), 14(10),<br />

27.<br />

Francois, D. and Jones, B.E., European Capsule Technology Symposium, Constance<br />

pp. 55-61 (11-13 October 1978).<br />

Fricke, S., Dittgen, M., Gerecke, H., Osterwald, H., Pharmazie, (1995),<br />

50, 507- 508.<br />

Furia, T.E. (Ed) Hand book of food Additives, 2 nd edn., (1972), 129-137.<br />

G<br />

Gary-Bobo, C.M., Dipolo, R., and Solomon, A.K., J. Gen Physiol., (1969), 54, 369.<br />

Ghaderi, R., Artursson, P., Carlfors J., Pharm. Res., (1999), 16, 676–<br />

681.<br />

Ghazy, F.S., Ghareeb, S.A., Mahdy,M.A., El-Ghamry, H.A., Issa,<br />

M.M. and Sabaty, A.M., (2004), Alex. J. Pharm. Sci., 18(2), 151-158.<br />

Ghebremeskel, A.N. , Vemavarapu, C., Lodaya, M., Int. J. Pharm.,<br />

(2007), 328, 119–129.<br />

Ginés, J.M., Arias, M.J., Moyano, J.R. and Sanches-Soto, P.J., Int. J.<br />

Pharm., (1996), 143, 247.<br />

Gloor, M., Fluhr, J., Wasik, B. and Gehring, W., Pharmazie ,<br />

(2001), 56,. 810–814.<br />

Goldberg, A. H., Gibaldi, M. and Kanig, J.L., J.Pharm. Sci.(1966),55,<br />

482-487.<br />

Goldberg, A. H., Gibaldi, M. and Kanig, J.L., J.Pharm. Sci.(1965),54,<br />

1145-1148.<br />

156


Goodman, M. and Barry, B.W., J. Invest. Dermatol., (1988), 91, 323-<br />

327.<br />

Greenhalgh, D.J., Williams, A.c., Timmins, P. and York, P., J. Pharm.<br />

Sci., (1999), 88(11), 1182.<br />

Gul Majid Khan and Zhu Jiabi, Drug Develop. Indust. Pharm., (1998),<br />

24(5), 455-462.<br />

Gupta, P., and Garg, S., Pharmaceutical technology, 104, (2002), 144.<br />

Gwak H.S., Chun I.K., Arch Pharm Res. (2001), 24(6), 572-7.<br />

H<br />

Habib, F.S. and El- Shanawany, S.M., Bull. Pharm. Sci., Assuit<br />

University, Egypt, (1989), 12(1), 90-102.<br />

Hadgraft, J. Modulation of the barrier function of skin. Skin<br />

Pharmacology and Applied Skin Physiology. (2001), 14(suppl 1): 72-81.<br />

Heather A.E. Benson. Current Drug Delivery, (2005), 2, 23-33.<br />

Higuchi, T. and Cannors, K.A., Adv. Anal. Chem. Instrum., (1965), 4,<br />

117.<br />

Hirvonen, J., Rajala, R., Vihervaara, p., Laine, E., Paronen, P.<br />

and Urtti, A., Eur. J. Pharm. Biopharm., (1994), 40, 81.<br />

Hong, S. S., Lee, S. H., Lee, Y. J., Chung, S. J., Lee, M. H. and Shim,<br />

C. K., J. Control. Rel., (1998), 51( 2-3), 185-192.<br />

Hosny, E.A., Abdel Hady, S.S., and Niazy, E.M., (1998), Acta Pharm.<br />

Helv., 72, 247-254.<br />

Howes, D., Guy, R., Hadgraft, J., Heylings, J., Hoeck, u.,Kamper, F.,<br />

Hu, C.J. and Rhodes, D.G., Int. J. Pharm., (1999), 185, 23-35.<br />

I<br />

157


Idson, B., Drug. Metab. Rev., (1983), 14, 207-222.<br />

Ismail, S., Mohamed, A.A. and Abd El-Mohesn, M.G., Bull.<br />

Pharm.Sci. Assiut. University, (1990), 13, Part 1, 115-123.<br />

J<br />

Jachowicz, R., Nurnberg, E. and Hoppe, R., Int. J. Pharm.(1993), 99,<br />

321-325.<br />

Jachowicz, R. and Nurnberg, E., ibid, (1997), 149-158.<br />

Jia –You, F., Pao-Chu,W., Yaw-Bin,H. and Yi-Hung, T., Int. J.<br />

Pharm., (1996),128, 169.<br />

Johari, G.P., Kim, S., Shanker, R.M., J. Pharm. Sci., (2005), 94, 2207–<br />

2223.<br />

Joseph T. Rubino,Cosolvents and Cosolvency, (2002), 1, 658-670.<br />

Juppo, A.M., Boissier, C., Khoo, C., Int. J. Pharm., (2003), 250(2), 385-<br />

401.<br />

K<br />

Kang, B.K., Lee, J.S., Chon, S.K., Jeong, S.Y., Yuk, S.H., Khang, G.,<br />

Lee, H.B. and Cho , Int. J. Pharm., (2004), 274, 65–73.<br />

Karavas, E., Georgios K., Aristotelis X. and Emmanouel G. , Eur. J.<br />

Pharm. Biopharm., (2006), 63, 103–114.<br />

Kaufmann, G., Dittgen, M., GraÈser, T., Gerecke, H., Osterwald, H.,<br />

Oettel, M., Pharmazie, (1995), 50, 50-51.<br />

Kerc, J., Srcic, S.,Kofler, B., Drug Dev. Ind. Pharm., (1998), 24(4), 359-<br />

363.<br />

Khitworth, C.K. and Stephenson, R.E., J. Pharm. Sci.,(1971), 60-48.<br />

Kligman, A.M., J. Am. Med. Assoc., (1965),193, 796.<br />

158


Kreuter, J., Feste Dispersionen, in: Kreuter, J., Herzfeldt (Eds.),<br />

Grundlagen der Arzneifoemenlehre Galenik, 2, Springer, Frankfurt am<br />

Main, 1999, pp. 262-274.<br />

Kshirsagr., N.A., Indian.J. Pharm. Sci. (2000), 32, 554.<br />

L<br />

Larrucea, E., Arellano,A., Santoyo,S. and Ygartua, P.,(2001), Drug<br />

Dev. Ind. Pharm., 27(3), 251-260.<br />

Law, D., Wang, W., Schmitt, E.A., Qiu, Y., Krill, S.L. and Fort, J., J.<br />

Pharm. Sci., (2003), 92(3), 505.<br />

Law, S.L., Lo, W.Y., Lin, F.M. and Chaing, C.H., Int.J. Pharm.,<br />

(1992), 84, 161.<br />

Leuner, C. and Dressman,J., Eur. J. Pharm. Biopharm., (2000), 55, 47–<br />

60.<br />

Lloyd, G.R., Craig, D.Q.M. and Smith, A., Eur. J. Pharm. Biopharm.,<br />

(1999), 48, 59–65.<br />

Loftsson, T., Brewster, M.E. J. Pharm. Sci., (1996), 85, 1017-25.<br />

M<br />

Mackenzie, I.C., J. Invest. Derm. (1975), 65(1): 45-51.<br />

Madhusudhan, B., Rambhan, D., Gudsoorkar, V.R., Shete, J.S.,<br />

Apte, S. S., Indian J.of Pharm.Sci., (1999), 61(6), 346-349.<br />

Majerik, V., Charrbit G., Badens E., Horvath G., Szokonya L.,<br />

Bosc N., Teillaud E., J. Supercrit. Fluids, (2007), 40, 101–110.<br />

Malay, k. D., Asokangshu, B. and Sarojk, G., Acta Poloniae<br />

Pharmaceutical Drug Research, (2006), 63 (6), 535-541.<br />

Malkinson, F.D., In The Epidermis, W., Montagna, Ed., Academic press, New York,<br />

(1964), chapter 20.<br />

Malkinson, F.D., J. Invest. Dermatol., (1958), 31, 19.<br />

Malkinson, F.D., J. Soc. Cosmetic Chemists, (1956), 7, 109.<br />

159


Mallick, B., Sahoo, A. and Mirta, B.B., Boll. Chim. Farm., (2003),<br />

142(4), 180.<br />

Mario, J., Mira, B. L., Ana, K. and Biserka, C. C., Acta Pharm,<br />

(2005), 55, 223-236.<br />

Martin, A. In "Physical Pharmacy," 4 th edn., Chapter (12 and 13), B.I.<br />

Waverly Pvt. Ltd., N.D. (1994), pp. 284-289 and 324-336.<br />

Martindale, "The Extra Pharmacopeia". 31 st edn., by James, E.F<br />

Reynolds. and Kathleen Parfitt, Royal Pharmaceutical Society, London,<br />

(1996), 341-361.<br />

McKenzie, A.W., Stoughton, R.B. Arch. Dermatol., (1962), 86, 608-<br />

610.<br />

Megwa, S.A.; Cross, S.E.; Benson, H.A.E.; Roberts, M.S. J. Pharm.<br />

Pharmacol., (2000), 52, 919-28.<br />

Mei, Z., Chen, H., Weng, T., Yang, Y., Yang, X. Eur. J. Pharm.<br />

Biopharm., (2003), 56, 189-96.<br />

Mendyk, A., Jachowicz, R., Expert Systems with Applications, (2005),<br />

28, 285-294.<br />

Menton, D.N., J. Invest. Derm., (1976), 66(5): 283-291.<br />

Mezei, M. and Gulasekharam, V. Life Sci., (1980), 26, 1473-7.<br />

Michniak, B. B., Chapman, J. M. and Seyda, K. L., J. Pharm. Sci.,<br />

(1993), 82, 214.<br />

Moes, A., J.Pharm. Acta. Helv., (1982), 56,21.<br />

Moghimi, H.R., Williams, A.C., and Barry, B.W., "Stratum corneum<br />

and barrier performance; a model lamellar structure approach," In<br />

"Percutaneous Absorption," R.L., Bronaugh, and H.I., Maibach, Eds.,<br />

Marcel Dekker Inc., New York, NY, (1999),515.<br />

Moghimi, H.R., Williams, A.C., and Barry, B.W., Int. J. of Pharm.<br />

(1996), 145, 49.<br />

160


Moghimi, H.R., Williams, A.C., and Barry, B.W., Int. J. of Pharm.<br />

(1997), 146, 41.<br />

Moneghini, M., Carcano, A., Zingone, G. and Perissutti, B., Int. J.<br />

Pharm., (1998), 175. 177.<br />

Moneghini, M., Kikic, I., Voinovich, D., Perissutti, B. and Filipovic-<br />

Grcic, J., Int. J. Pharm., (2001), 222 (1). 129.<br />

Monteiro-Riviere, N.A. Comparative anatomy, physiology, and<br />

biochemistry of mammalian skin. In: Dermal and Ocular Toxicology:<br />

Fundamentals and Methods. Ed. D Hobson, CRC Press, London. (1991),<br />

pp. 3-17.<br />

Monteiro-Riviere, N.A. Dermatotoxicology. In: Introduction to<br />

Biochemical Toxicology. Eds. E. Hodgson and R.C. Smart, Wiley-<br />

Interscience. (2001a), pp. 509-537.<br />

Monteiro-Riviere, N.A. Integument. In: Biology of the Domestic Pig.<br />

Ed. W.G. Pond and H.J. Mersmann, Comstock Publishing. (2001b), pp.<br />

625-652.<br />

Monteiro-Riviere, N.A. Anatomical factors affecting barrier function. In:<br />

Dermatotoxicology. Eds. H. Zhai and H.I. Maibach, CRC Press. (2004),<br />

pp. 43-70<br />

Monteiro-Riviere, N.A. Structure and function of skin. In: Dermal<br />

Absorption Models in Toxicology and Pharmacology. Ed. J.E. Riviere,<br />

Taylor and Francis. (2006), pp. 1-19.<br />

Monteiro-Riviere, N.A.; Inman, A.O.; Mak, V.; Wertz, P.; Riviere,<br />

J.E. Effect of selective lipid extraction from different body regions on<br />

epidermal barrier function. Pharmaceutical Research. (2001), 18: 992-<br />

998.<br />

Monteiro-Riviere, N.A.; Stinson, A.W. Calhoun, HL Integument. In:<br />

Textbook of Veterinary Histology. Ed. H.D. Dellmann, Lea and Febiger.<br />

(1993), pp. 285-312.<br />

161


Morgan , T.M., Parr, R.A., Reed, B.L., and Finnin., B.C.,<br />

J.Pharm.Sci., (1998), 87, 1219.<br />

Muhrer G., Meier U., Fusaro F., Albano S., Mazzotti M., Int. J.<br />

Pharm., (2006), 308, 69–83.<br />

Mukne, A.P., Nagarsenker, M.S., AAPS Pharm. Sci. Tech., (2004),<br />

5(1), 1-9.<br />

Mummaneni, V., Vassavada, R.C., Int. J. Pharm., (1990), 66(1), 71-77.<br />

Mura, P., Faucci, M.T., Manderioli, A., Bramanti, G. and Parrini,<br />

P., Drug Develop. Indust. Pharma., (1999), 25(3), 257-264.<br />

Mura, P., Faucci, M.T., Bramanti, G. and Corti, P., Eur. J. Pharm.<br />

Sci., (2000), 9, 365-372.<br />

Mutalik S. and Udupa N., Pharmazie, (2003), 58(12), 891-894.<br />

Mutalik S. and Udupa N., J. Pharm. Sci., (2005), 8(1): 26-38.<br />

Mutalik S. and Udupa N. J. Pharm. Sci., (2004) 93(6), 1592.<br />

N<br />

Nakano, M. and Patel, N., J. Pharm. Sci., (1970), 59, 985-988.<br />

Nakano, M., Kuchiki, A., and Arita,T., Chem. Pharm. Bull., (1976) (Tokyo),23<br />

1404.<br />

Namdeo, A., and Jain., N.K., Indian.J. Pharm. Sci. (1996), 58(2), 41.<br />

Nokhodchi, A., Shokri, J., Dashbolaghi, A., Hassan-Zadeh, D.,<br />

Ghafourian, T. and Barzegar-Jalal, M., J. Int. Pharm., (2003), 250(2),<br />

359-369.<br />

Nour, S.A., Bull. Fac. Pharm. Cairo Univ., (1993), 31, Nr.2, 219.<br />

O<br />

Okonogi, S., Oguchi, T., Yonemochi, E., Puttipipatkhachorn,S. and<br />

Yamamoto, K., Int. J. of Pharm., (1997), 156, 175-180.<br />

162


P<br />

Palmieri, G.F., Welhrle, P. and Martelli, S., Drug Dev. and Ind.<br />

Pharm., (2002), 24(7), 653.<br />

Pan, R.N., Chen., J.H., Chen, R.R., Drug Dev. Ind. Pharm., (2000), 9,<br />

989-994.<br />

Pamela, G.T., Glibenclamide In: Analytical Profiles of Drug Substances,<br />

Vol (10), Klaus Florey, (1981), Academic Press, INC. (London) LTD,<br />

pp.338-355.<br />

Park, E.S., Chang, S.J., Rhee, Y.S. and Chi, S.C., Drug Dev. and Ind.<br />

Pharm., (2001), 27, 975–980.<br />

Patil, S., Singh, P., Szolar-Platzer, C., and Maibach. H., J. Pharm.<br />

Sci., (1996), 85, 249.<br />

Pinnamaneni, S., Das, N.G., Das, S.K., Pharmazie, (2002), 57, 291 – 300.<br />

Pokharkar, V.B., Mandpe, L.P., Mahesh N. Padamwar, M.N.,<br />

Anshuman A. Ambike, A.A., Kakasaheb R. Mahadik, K.R. and<br />

Paradkar, A., Powder Technol., (2006), 167, 20–25.<br />

Porzio, S., Csselli, G., Pelleggrini, L., Del Rosario, M., Coppola, A.,<br />

Boltri, L., Gentile, M., Clavenna G. and Melillo, G.,<br />

Pharmacol.Res., (1998), 37, 41.<br />

Potts, R.O.; Guy, R.H. Pharm. Res., (1992), 9, 663-9.<br />

Pouton, C.W., Eur. J. Pharm. Sci., (2006), 29, 278–287.<br />

Prausnitz, M.R, Ackley, D.E., and Gyory., J.R., "Macrophabricated<br />

Microneedles for Transdermal Drug Delivery Technology,"<br />

M.J.Rathbone, J., Hadgraft, M.S., Roberts, Eds., Marcel Dekker Inc.,<br />

New York, NY, 2003, 514.<br />

163


Price, J.C., Polyethylene glycol. In: A. Wade and Weller, P.J., Editors,<br />

Handbook of Pharmaceutical Excipients, American Pharmaceutical<br />

Association/The Pharmaceutical Press, Washington, DC/London (1994),<br />

pp. 355-361.<br />

R<br />

Ramadan, A.A., Journal of Applied Sciences Research, (2008), 4(9),<br />

1052-1065.<br />

Rasenack, N. and Muller, B.W., , Pharm. Dev. Technol., (2004), 9, 1–<br />

13.<br />

Report, Polymeric Transport System, Cosmetics and Tolietries, (1992),<br />

107, 14.<br />

Riviere, J.E. Biological factors in absorption and permeation. Cosmetics<br />

and Toiletries.(1990), 105: 85-93.<br />

Ruland, A., Kreuter, J. and Rytting, J. H., Int. J. Pharm., (1994), 101,<br />

57.<br />

S<br />

Sammour, O.A., Elkheshen, S,A., El-Shaboury, M.H., Al-Quadeib,<br />

B.T., Bull Fac Pharm Cairo Univ., (2001), 39, No. 1.<br />

Santos Maia, C., Mehnert, W., Schaller, M., Korting, H.C., Gysler,<br />

A.; Haberland, A., Schafer-Korting, M. J. Drug Target, (2002), 10,<br />

489-95.<br />

Santoyo, S., and Ygartua, P., Eur. J. Pharm. Biopharm., (2000), 50,<br />

245–250.<br />

Sarisuta, N., Saowakontha, R. and Ruangusksriwong, C., Drug Dev.<br />

Ind. Pharm., (1999), 25(3), 373.<br />

Sarpotdar, P.P. and Zatz, J.L., Drug Dev. Ind. Pharm., (1986a), 12,<br />

1625–1647.<br />

164


Sarpotdar, P.P. and Zatz, J.L., J. Pharm. Sci., (1986b), 75, 176.<br />

Sarveiya, V.; Templeton, J.F.; Benson, H.A.E. J. Pharm. Pharmacol.,<br />

(2004), 56, 717-724.<br />

Schachter, D.M., Xiong, J., Tirol, G.C., Int. J. Pharm., (2004), 28(1),<br />

89-101.<br />

Scheuplein, R.J. and Ross, L.W., J. Invest. Dermatol., (1974),62, 353-<br />

360.<br />

Schwartz, C., Mehenert., W., Lucks, J.S., and Muller, R.H.,<br />

Int.J.Pharm.Sci., (1992), 88, 53.<br />

Scott, R.C., "In-vitro absorption through damaged skin," In<br />

"Fundamentals and Applications," (1991), Bronaugh, R.L. and Maibach,<br />

H.I. (ed.), CRC Press., PP. 129-135.<br />

Sekiguchi, K. and Obi, N., Chem. Pharm. Bull (Tokyo), (1964), 12,<br />

134–144.<br />

Sekiguchi, K., Obi, N., Chem. Pharm. Bull., (1961), 9, 866-872.<br />

Sentjurc,M., Vrhovnik, K., Kristl, J. J. Control Rel., (1999), 59, 87- 97.<br />

Serajuddin, A.T., J. Pharm. Sci., (1999), 88, 1058–1066.<br />

Sethia, S. and Squillante, E., Int. J. Pharm., (2004), 272(1-2), 1181.<br />

Shahiwala, A. , Misra, A. J. Pharm. Pharmaceut. Sci., (2002), 5, 220-5.<br />

Siavoush Dastmalchi, Alireza Garjani, Nasrin Maleki, Golaleh<br />

Sheilkhee, Vida Baghchevan, Parisa Jafari-Azad, Hadi Valizaeh,<br />

Mohammad Barzegar-Jalali, J. Pharm. Pharmaceu. Sci., (2005), 8(2),<br />

175-181.<br />

Silverstein, R.M., Bassler, G.C., Morril, T.C., Wiley, New York,<br />

(1991), pp. 91-131.<br />

Singh, J., Tripathi, K. P. and Sakya, T. R., Drug Dev. Ind. Pharm.,<br />

(1993), 19,1623.<br />

Singh, U.V. and Udupa, N., Indian. J. Pharm. Sci., (1997), 59(6), 333.<br />

165


Singhal, D. and Curatolo, W., Adv. Drug. Deliv. Rev.,(2004), 56, 335-<br />

347.<br />

Sinha, V.R. and Maninder Pal Kaur, Drug Develop. Ind. Pharm.,<br />

(2000), 26(11), 1131-1140.<br />

Sloan, K.B., Prodrugs. Topical and Ocular Drug Delivery. Drugs and the<br />

Pharmaceutical Sciences. Vol. 53. New York: Marcel Dekker Inc. (1992),<br />

pp. 313.<br />

Sloan, K.B.; Wasdo, S. Med. Res. Rev., (2003), 23, 763-93.<br />

Smack, D.P.; Korge, M.D.; James, W.D. Journal of the American<br />

Academy for Dermatology. (1994), 30: 85-102.<br />

Sabati, A.M, M.sc. pharm. Thesis, Faculty of pharmacy, Zagazig Univ.<br />

Egypt, (1998).<br />

Sridevi, S; Chary, M.G.; D.R. Krishna, D.R.; Prakash V. Diwan<br />

Indian Journal of Pharmacology (2000); 32: 309-312<br />

Srini Tenjarla, Porranee Puranajoti, Ram Kasina and Tarun<br />

Mandal, J.Pharm. Sci., (1998), 87(4), 425-429.<br />

Srinivas, M. and Nayanabhirama, U., J.Pharm. Pharmaceut. Sci.,<br />

(2005), 8(1), 26-38.<br />

Stinchcomb, A.L., Swaan, P.W., Ekabo, O., Harris, K.K., Browe, J.,<br />

Hammell, D.C., Cooperman, T.A. and Pearsall, M. J. Pharm. Sci.,<br />

(2002), 91, 2571-8.<br />

Stetinova, V., Kvetina, J., Patera, J., Polakova, A. and Prazakova,<br />

M., Biopharmaceutics and Drug Deposition, (2007),28(5), 241-248.<br />

Stroev, E.A., Belkina, Z.V., Farmakol Toksikol (1989), 7,52-74.<br />

Suwanpidokkul, N., Thongnopnua, P., and Umprayn, K., AAPS<br />

Pharm Sci Tech., (2004), 5(3)48, 1.<br />

Swarbrick J., Boylan J.C., "Encyclopedia Of Pharmaceutical<br />

Technology"; 2 nd edn, (2002), 3, 2458-2479.<br />

166


T<br />

Tadeusz, W. Hermann, Roman Dobrucki, Stefan Piechocki, Matylda<br />

Resztak and Robert Reh, Med Sci Monit, (2005); 11(6),181-188.<br />

Takahashi Y, Furuya K, Iwata M, Onishi H, Machida Y, Shirotake S.<br />

Yakugaku Zasshi. (1997), 117(12):1022-7.<br />

Takahashi, H., Chen, R., Okamoto, H., Danjo, K., Chem. Pharm. Bull<br />

(Tokyo), (2005), 53, 37-41.<br />

Takeuchi, H., Nagira, S., Yamamoto, H., Kawashima, Y., Powder<br />

Technology, (2004), 141(3), 187-195.<br />

Tantishaiyakul, V., Kaewnopparat, N. and Ingkatawornwong, S., Int.<br />

J. Pharm.,(1996), 143, 59.<br />

Taylor, C.; Scogna, K.H.; Ajello, J.P. (Eds). Histology: A Text and<br />

Atlas. Baltimore: Lippencott Williams & Wilkins. (2006), pp. 98-138;<br />

442-463.<br />

Taylor, L.S. and Zografi, G., Pharm. Res., (1997), 14, 1691–1698.<br />

Tehrany, M.R. and Mehramizi, A., (2000). Drug Dev. Ind. Pharm.,<br />

26(4), 409-414.<br />

Trapani, G., Franco, M., Latrofa, A., Pantaleo, M.R. and<br />

Provenzano, M.R., Int. J. Pharm., (1999), 184, 121.<br />

Thorsteinsson, T.; Masson, M.; Loftsson, T.; Haraldsson,<br />

G.G.;mStefansson, E. Pharmazie, (1999), 54, 831-6.<br />

Torrado, S., Torrado, J., Cadorniga, R., Int. J. Pharm., (1996), 140,<br />

247-250.<br />

Touitou, E., Dayan, N., Bergelson, L.; Godin, B., Eliaz, M. J. Control<br />

Rel., (2000), 65, 403-18.<br />

Tsai, J.C., Guy, R. H., Thornfeldt, C. R., Gao, W. N., Elias, P. M.<br />

and Feingold, K. R. J. Pharm. Sci., (1996), 85, 643.<br />

167


Tupker, R.A., Pinnagoda, j. and Nater, J.P., Acta Derm. Venereol.<br />

(Stockh.), (1990), 70, 1–5<br />

U<br />

USP-25, NF-20. United State Pharmacopeia-XXV, National Formulary-<br />

XX. United State Pharmacopeial Convention INC., Twinbrook, Parkway,<br />

Rockville, MD, USA, (2002), pp.598.<br />

V<br />

Valenta, C., Siman, U., Kratzel, M. and Hadgraft, J., Int. J. Pharm.,<br />

(2000), 197, 77-85.<br />

Van den Mooter, G., Weuts, I., De Ridder, T. and Blaton, N., Int. J.<br />

Pharm., (2006), 316, 1–6.<br />

Van den Mooter, G.,Augustijns, P., Blaton, N. and Kinget, R., ibid.,<br />

1998, 164, 67-80<br />

Vasanthavada, M., Tong, W.Q., Joshi, Y. and Kislalioglu,<br />

M.S., ,Pharm. Res., (2004), 21, 1598–1606.<br />

Vasconcelos, T. and Costa, P., Development of a rapid dissolving<br />

ibuprofen solid dispersion. In PSWC – Pharmaceutical Sciences Wolrd<br />

Conference., (2007).<br />

Villiers, M.M., Wurster, D.E., Watt, J.G., Ketkar, A., Int. J. Pharm.,<br />

(1998), 163(1), 219-224.<br />

Vippagunta, S.R., Zeren, W., Stefanie, H., Steven, L. K., J. Pharm.<br />

Sci., (2006), 96, 294–304.<br />

Visser, M.R., Drooge, D.J., Hinrichs, W.L.J., Wegman, K.A.M.,<br />

Eissens, A.C., Frijlink, H.W., Eur. J. Pharm., (2004), 21(4), 511-518.<br />

W<br />

Wahlberg, J.E., Acta Derm. Venereol. (Stockh), (1965), 45, 397.<br />

168


Walking, W.D., Povidone . In: A. Wade and P.J. Weller, Editors, Handbook of<br />

Pharmaceutical Excepients. American Pharmaceutical Assosiation/ The<br />

Pharmaceutical Press, Washington, DC/London (1994), 392-399.<br />

Wang, X., Michoel, A. and Van den Mooter, G., Int. J. Pharm., (2005),<br />

303, 54–61.<br />

Watannabe, T., Hasegawa, SA., Wakiyama N., Kusai A., Senna M.,<br />

Int. J. Pharm., (2003), 250(1), 283-286.<br />

Wertz, P.W. Percutaneous absorption: role of lipids. In:<br />

Dermatotoxicology. Eds. H. Zhai and H.I. Maibach, CRC Press. pp.<br />

(2004) 71-81.<br />

Westesen, K., Siekmann, B., and Koch, M.J.H., Int.J.Pharm.Sci.,<br />

(1998), 93, 199.<br />

Wester, R. C. and Maibach, H. I. (1999). Regional variation in<br />

perctaneous absorption. In: Bronaugh, R.L. and Maibach, H.I. eds.<br />

Percutaneous Absorption; Drugs – Cosmetics – Mechanisms –<br />

Methodology, 3 rd edn. New York: Marcel Dekker, 107 -116.<br />

Wickrema Sinha, A.J. Shaw, S.R., and Weber, D.J., J. Invest. Dermatol., (1978),<br />

71, 372.<br />

William, A.C. and Barry, B.W., Advanced Drug Delivery Reviews, (2004), 56(5),<br />

603-618.<br />

Winfield, A.J., "External preparations," In" Pharmaceutical Practice," 2 nd edn., Part<br />

(2), Chapter (14)., Winfield, A.J and Richards, R.M.E. American Pharmaceutical<br />

Association, Washington, D.C., (1998), PP. 136 – 146.<br />

Williams, A.C and Barry.B.W, Advanced Drug Delivery Reviews, (2004), 56 (5),<br />

603-618.<br />

Wrenn, J.R. and Simeon, M., United State (1998), Patent No. , 6, 174,<br />

873.<br />

Wurster, D.E., and Kramer, S.F., J. Pharm. Sci. (1961), 50, 288.<br />

Wulff, M., Alden, M. and Craig, D.Q.M., Int. J. Pharm., (1996), 142, 189-198.<br />

Y<br />

169


Yakou, S., Umehara, K., Sonobe, T., Nagia, T., Sugihara, M. and<br />

Fukuyama, Y., Chem.Pharm. Bull.,(Tokyo) (1984), 32(10), 4130.<br />

Yalkowsky, S.H., Roseman, T.J. 1981. "Solubilization of drugs by<br />

cosolvents," In: Yalkowsky, S.H. (Ed.), Techniques of Soluhilization of<br />

Drugs. Dekker, New York.<br />

Yang, S., Zhu, J., Lu, Y., Liang, B., and Yang, C., Pharm. Res., (1999),<br />

16, 751.<br />

Yokomizo, Y., J. Controlled Release, (1996), 42, 217.<br />

Yoshihashi, Y., Iijima H., Yonemochi E., Terada K. , J. Therm. Anal.<br />

Calorim., (2006), 85, 689–692.<br />

Youssef, M. K., El-sayed, E. D., and Fouda, M. A., Drug Dev. and Ind.<br />

Pharm., (1988), 14(15-17), 2667-2685.<br />

170


-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

171<br />

<br />

-<br />

-


-<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

-<br />

-<br />

<br />

<br />

<br />

<br />

<br />

-<br />

-<br />

-<br />

172<br />

-<br />

-<br />

-<br />

<br />

<br />

-


-<br />

<br />

173<br />

-<br />

-<br />

<br />

<br />

<br />

-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-<br />

-


-<br />

<br />

<br />

174<br />

<br />

-<br />

-<br />

-<br />

-<br />

-<br />

<br />

-<br />

-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-


175<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

-<br />

<br />

<br />

<br />

<br />

-

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!