Sie sind auf Seite 1von 163

Enhancement of Solubility and Dissolution of Gliclazide by

Complexation with Hydroxypropyl β-Cyclodextrin


By

Mr. Amit Tondare. B. Pharm.


Reg. No. 09PI127
A dissertation submitted to the

Rajiv Gandhi University of Health Sciences, Karnataka, Bangalore


in partial fulfillment
Of the requirements for the degree of

MASTER OF PHARMACY
In
INDUSTRIAL PHARMACY
Under the Guidance of
Mr. SUNIL RAJ RAGA M. Pharm. (Ph.D)

Department of Industrial Pharmacy


K.R.E.S’s Karnataka College of Pharmacy,
BIDAR – 585403 (Karnataka State)
2010-11
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
KARNATAKA

DECLARATION BY THE CANDIDATE

I hereby declare that the matter embodied in the dissertation entitled

“Enhancement of Solubility and Dissolution of Gliclazide by Complexation with

Hydroxypropyl β-Cyclodextrin is a bonafide and genuine research work

carried out by me under the guidance of Mr. SUNIL RAJ RAGA M. Pharm. (Ph.D)

Department of Industrial Pharmacy, Karnataka College of Pharmacy, Bidar.

Date:

Place: Bidar Amit Tondare


RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
KARNATAKA

CERTIFICATE BY THE GUIDE

This is to certify that the dissertation entitled “Enhancement of Solubility and

Dissolution of Gliclazide by Complexation with Hydroxypropyl β-Cyclodextrin” is a

bonafide research work done by Mr. Amit Tondare submitted in partial

fulfillment of the requirement for the degree of “Master of Pharmacy” in

Industrial Pharmacy of the “Rajiv Gandhi University of Health Sciences,

Karnataka.

Date:

Place: Bidar
Mr. Sunil Raj Raga M. Pharm. (Ph.D)
Asst. Prof.
Department of Industrial Pharmacy,
K.R.E.S’s Karnataka College of Pharmacy,
Bidar-585 403
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
KARNATAKA

ENDORSEMENT BY THE PRINCIPAL & H.O.D.

This is to certify that the dissertation entitled Enhancement of Solubility

and Dissolution of Gliclazide by Complexation with Hydroxypropyl β-

Cyclodextrin is a bonafide research work done by Amit tondare Under the

direct supervision and guidance of Mr. SUNIL RAJ RAGA M. Pharm. Ph.d

Department of Industrial Pharmacy, Karnataka College of Pharmacy, Bidar.

Date:

Place: Bidar
Prof. Krantikumar M. Sirse M. Pharm. PhD
Principal & HOD
K.R.E.S’s Karnataka College of Pharmacy,
Bidar-585 403
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
KARNATAKA

COPYRIGHT

I hereby declare that the Rajiv Gandhi University of Health Science,

Karnataka shall have the rights to preserve, use and disseminate this

dissertation / thesis in print or electronic format for academic / research

purpose.

Date:

Place: Bidar TONDARE AMIT

@ Rajiv Gandhi University of Health Sciences, Karnataka


ACKNOWLEDGMENT

I take this privilege and pleasure to acknowledge the contributions of many


individuals who have been inspirational and supportive throughout my work
undertaken and endowed with the most precious knowledge to see success in my
endeavor.

It is with pleasure of immense gratitude that I express my most cordial and


humble thanks to my esteemed teacher and guide Shri. Sunil raj Raga, M. Pharm.
PhD, assistant Professor, Department of Industrial Pharmacy, Karnataka College
of Pharmacy, for his valuable guidance, keen interest, perennial inspiration and
everlasting encouragement. I shall for ever remain indebted to him for having
inculcated in me a quest for excellence, a spirit of diligence and perseverance, a
sense of humility, honesty and respect for the moral and ethics which govern our
sciences and without whom this work would not have seen the light of the day.

I am gratefully indebted to Prof. Sirse KrantiKumar, M. Pharm. PhD, the


honorable principal of Karnataka College of Pharmacy, Bidar, for providing
necessary facilities to carryout my work and for his constant support and
encouragement.

I consider it as a great privilege to express my heartfelt gratitude and sincere


thanks to Mr. S.B.Dana, M. Pharm. (Ph.D) and Mr. AjayKumar Patil, M. Pharm. (Ph.D),
Assistant Professor, Industrial Pharmacy, Karnataka College of Pharmacy, for his
valuable suggestions, deep inspiration and cooperative nature, with due respect in
my heart.

I extend my sincere thanks to Dr. Baswaraj Raga, Dr. Kashinath Naubade,


Dr. Malikarjun patil, Mr.Vijayakumar Trilapure, Mr.Shivanand Noola
Department of Pharmaceutical chemistry, Mr. Raghavendra Patil, Mr.V V Shastri
& Mr. Sunil G Department of Pharmacology, and I am thankful Mr. Mallikarjun
Mugli, Mr. S. Chikpety, and other staff members of our college for their help and
interest in the present work

I take this opportunity to express my sincere thanks to teaching and non-


teaching staff members, Karnataka College of Pharmacy, Bidar.
My special thanks and appreciation goes to all my seniors, especially, Mr.
irfani ,bilal,vijaya,vasisht,sudhir,zaheer,imran, Nadeem, Waseem for their good
wishes timely help and unstinted support during the course of study.

I will not forget the help and company given by my junior, Sandeep, Shrutin,
Satish, Azhar, Vinod, Zia, Younus, Sharfaraz, Ilyas,
Om,Ramya,Priyanka,Shivleeia,Nauman,Anil and dearest friends Mr
Ravi,abhijit,shridhar,yogesh,sachin,Anil,Neha,Akash,Ganesh,Nilesh,Gargi and all
my friends for their moral support good company and encouragement.

I would like to express my sincere thanks to Aditya,atul,varsha,Deepak and


all the seniors of M. Pharm for their moral support, good company and
encouragement.

I would like to express my sincere thanks to Deepak,basavraj,Kvamshi,


Madhuri,Swati,shravanti,Mrudula and all the juniors of B. Pharm for their
moral support good company and encouragement.

Words are not sufficient to express my deepest love and appreciation to my


affectionate, beloved Parents and my sister ashwini for their support and boosting
encouragement throughout my career.

Finally I consider this as an opportunity to express my gratitude to all the


dignitaries who have been involved directly or indirectly with the successful
completion of this dissertation.

Date:

Place: TONDARE AMIT.


Dedicated to

My

Beloved

Psarents

IX
&

sirshreeji

X
ABSTRACT

Gliclazide is a second generation sulphonylurea oral hypoglycemic agent. It is

practically insoluble in water. In the present study attempt has been made to prepare and

characterize inclusion complexes of Gliclzide with HP-β-CD. The phase solubility

analysis indicated the formation of 1:1 molar inclusion complex of Gliclazide and HP-β-

CD. Apparent stability constant (KC) was 256.3 M-1 for HP-β-CD complexes

respectively. The inclusion complexes were prepared by three different methods viz.

physical, kneading and method. The prepared complexes were characterized using FT-IR

DSC and differential scanning calorimetry. The inclusion complex prepared with HP-β-

CD by kneading method exhibited greatest enhancement in solubility and fastest

dissolution) of Gliclazide.

Further, this inclusion complex GLZ: HP-β-CD with different ratios was

formulated into tablets using microcrystalline cellulose, potato starch, Talc, Mg sterare

and lactose. The tablets prepared with HP-β-CD by kneading method exhibited fastest

dissolution of Gliclazide. The prepared tablets were evaluated for various post

compression parameters like hardness, friability, weight variation, thickness, drug

content, in-vitro dissolution and FT-IR and DSC studies.

Key words: Gliclazide, β-CD, HP-β-CD, Physical method, Kneading method,

XI
LIST OF ABBREVIATIONS

Abbreviations Complete Name

β-CD Betacyclodextrin

GLZ Gliclazide

HP- β-CD Hydroxypropyl betacyclodextrin

o
C Degree centigrade

hrs Hours

Mcg/μg Microgram

mg Milligram

min Minute

ml Milliliter

nm Nanometer

% Percent/ Percentage

rpm Rotations per minute

MC Microcrystalline cellulose

SD Standard deviation

λmax Wavelength maximum

X
TABLE OF CONTENTS

SR.NO CHAPTER PAGE NO.

1 INTRODUCTION 1

2 OBJECTIVES 47

3 REVIEW OF LITERATURE 48

4 METHODOLOGY 62

5 RESULTS 77

8 SUMMARY, CONCLUSION 111

9 BIBLIOGRAPHY 119

XII
TABLE OF CONTENTS

SR.NO CHAPTER PAGE NO.

1 INTRODUCTION 1

2 OBJECTIVES 47

3 REVIEW OF LITERATURE 48

4 METHODOLOGY 62

5 RESULTS 77

8 SUMMARY, CONCLUSION 111

9 BIBLIOGRAPHY 119

XII
LIST OF TABLES

Table
No. Title Page No.

1 Characteristics of Cyclodextrins 2

2 Physicochemical properties of cyclodextrin 3

3 CD-enhanced Solubility and Dissolution 12

4 CD Effect on Drug Stability 17

5 Modification of the Drug Release 30

6
Applications of Various CD Derivatives 31

7 59
Materials and their Sources
Equipments and their source
8 60

9 68
Formulae of Tablets.

10 Calibration curve data of Gliclazide 75

11 Solubility (mg/ml) of gliclazide in various fluids 76

12 Effect of pH on solubility of glicazide. 76

Phase Solubility Study Profile of gliclazide with HP β-CD


13 77

Drug Content Profile of Gliclazide HP-β-CD inclusion

Complexes
14 80

XIII
Drug Content Profile of Gliclazide HP-β-CD Physical
86
Mixtures
15

Percentage of drug release Profile of Gliclazide inclusion


16 complex with HP-β-CD 87
(Kneading Method)

Percentage of drug release Profile of Gliclazide complex with


HP-β-CD
17 (Physical Mixture) 88

Correlation Coefficients (R2) Values in the analysis of


Glicazide HP-β-CD Dissolution data profile as per Zero order
18 and First order 86

Dissolution Parameter Profile of Gliclazide- HP-β-CD


19 95

Percentage of drug release Profile of Gliclazide- HP-β-CD


20 Tablets 96

. Correlation Coefficients (R2) Values in the analysis of


Glicazide HP-β-CD Tablets Dissolution data profile as per Zero
21 order and First order 98

Dissolution Parameter Profile of Gliclazide- HP-β-CD

Tablets
22 100

Hardness Profile of Gliclazide-HP-β-CD Tablets.


23 100

24 Disintegration Time Profile of Gliclazide HP-β-CD Tablets. 101

25 Friability Percentage Profile of Gliclazide-HP-β-CD Tablets 101

26 Weight Variation Profile of Gliclazide HP-β-CD Tablets. 102

XIV
Drug Content Profile of Gliclazide HP-β-CD Tablets
27 after Stability Test 102

Percentage of drug release Profile of Gliclazide- HP-β-CD


28 Tablets After Stability Study 103

Hardness Profile of Gliclazide-HP-β-CD Tablets after stability


29 test. 105

Disintegration Time Profile of Gliclazide HP-β-CD Tablets


30 after stability test 105

Friability Percentage Profile of Gliclazide-HP-β-CD Tablets

after stability test.


31 106

Weight Variation Profile of Gliclazide HP-β-CD Tablets after


32 Stability Study. 106

XV
LIST OF FIGURES

Page
Fig. No. Title
No.

1 Structure of Cyclodextrins 11

2 the effect of complex stability constant on drug degradation. 19

3 Mode of penetration enhancement by CDs 23

4 Phase solubility diagram 42

5 Standard Calibration Curve for Gliclazide 75

6 Phase Solubility Study Profile of Gliclazide with HP β-CD 77

7 IR Profile of Gliclazide and its HP β-CD inclusion complexes 78

8 IR spectra of Gliclazide and its HP β-CD Physical Mixture 79

DSC Profile of Gliclazide and its HP β-CD Inclusion


9 complexes 80

10 DSC Profile of Gliclazide and its HP β-CD Physical Mixture 81

XRD Profile of Gliclazide and its HP β-CD Inclusion


11 complexes 82

12 XRD Profile of Gliclazide and its HP β-CD Physical Mixture 84

Dissolution Rate Data Profile graph of Gliclazide and its


13 Inclusion Complex (Kneading Method) 89

XVI
Dissolution Rate Data Profile graph of Gliclazide and it’s
14 Complex(Physical Mixture) 90

% Drug Remained Profile of Glaclazide HPβ-CD Inclusion


15 complex. 87

% Drug Remained Profile of Glaclazide HPβ-CD Physical


16 Mixture 92

Log % Drug Remained Profile of Glaclazide HPβ-CD Inclusion


17 complex 93

Log % Drug Remained Profile of Glaclazide HPβ-CD Physical


18 Mixture 94

Dissolution Rate Data Profile graph of Gliclazide- HP-β-CD


19 Tablets 97

20 % Drug Remained Profile of Glaclazide HPβ-CD Tablets. 99

21 Log % Drug Remained Profile of Glaclazide HPβ-CD Tablets 99

Dissolution Rate Data Profile graph of Gliclazide- HP-β-CD


22 Tablets After Stability Study. 104

XVII
INTRODUTION

Cyclodextrins have been used as complexing agents to increase the aqueous

solubility of poorly water soluble drugs and to increase their membrane permeability,

bioavailability and stability.1, 2 In addition; CDs have been used to reduce gastrointestinal

or ocular irritation, to mask unpleasant taste and to prevent drug-drug and drug-additive

interactions1, 2
The permeability through biological membrane is enhanced by the

presence of cyclodextrin. Cyclodextrin and their derivatives play an important role in the

formulation development due to their effect on solubility, dissolution rate, chemical

stability and absorption of a drug 3, 4.

Cyclodextrins are cyclic (α-1, 4)-linked oligosaccharides of α-D-glucopyranose

containing a hydrophilic outer surface and hydrophobic core. Owing to lack of free

rotation about the bonds connecting the glucopyranose units, the cyclodextrins are not

perfectly cylindrical molecules but are toroidal or cone shaped.1 Based on this

architecture, the primary hydroxyl groups are located on the narrow side of the cone

while the secondary hydroxyl groups are located on the wider edge as shown in Figure 1.

The commonly used CDs are α-cyclodextrin (α-CD), β-cyclodextrins (β-CD), and γ-

cyclodextrins (γ-CD), which consist of six, seven and eight glucopyranose units,

respectively.

Chemical and physical properties of the most common CDs are given in Table N0

1. Partially prehydrolysed starch, a mixture of acyclic dextrins, is converted by

cyclodextrin-glucosyl transferase enzyme, which is produced by microorganism Bacillus

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 1 
macerans to a mixture of cyclic and acyclic dextrins. From the mixture, cyclic dextrins,

the pure homogenous crystalline CD are isolated.

The aqueous solubility of β-CD is low; hence chemically modified β-CD is used.

Β-CD is used widely because of low cost, optimum cavity size and better complexing

efficiency.2 Modified β-CD has more aqueous solubility and improves the association

with guest molecules. Modified β-CD such as methylated cyclodextrin (Mβ-CD),

hydroxypropylated cyclodextrins (HPβ-CD), hydroxyethyl cyclodextrin (HPβ-CD) and

their physicochemical properties are given in Table N0 2.

The main reason for the solubility enhancement in these derivatives is that

chemical manipulation frequently transforms the crystalline CDs into amorphous

mixtures isomeric derivatives. For e.g.,(2-hydroxy propyl)- β-CD is obtained by treating

a base –solubilized solution of β-CD with propylene oxide, resulting in an isomeric

system that has an aqueous solubility well in excess of 60%(w/v)197. Several studies

reported the Cyclodextrin complexation of a variety of drugs for various purposes.

Table 1: Some characteristics of α-, β-, and γ-cyclodextrins

No of glucopyranose units α β γ

Molecular weight 972 1135 1297

Central cavity diameter (Ao) 4.7-5.3 6.0-6.5 7.5-8.3

Water solubility (g/100 ml at RT) 14.5 1.85 23.2


(α) D 250
(optical rotation) 150 + 0.5 162.5+ 0.5 177.4 + 0.5

Cavity diameter (Ao) 4.7-5.3 0.0-6.5 7.5-8.3

Height of torus (Ao) 7.9 ± 0.9 7.9 ± 0.9 7.9 ± 0.9

Diameter of outer periphery (Ao) 14.6 + 0.4 15.4 + 0.4 17.5 + 0.4

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 2 
Approximate Volume of Cavity (Ao)3 174 262 427

Approximate Volume of Cavity in


1 mol -CD (ml) 10 157 250

Table 2: Some physicochemical properties of β-cyclodextrin derivatives and

γ- cyclodextrin.

Name of the cyclodextrin Molecular Weight Solubility in water Specific Rotation


(G/ml, 25o C)

β-Cyclodextrin 1135 1.88 +162.5

Methyl β-Cyclodextrin 1303 >25 +160 – 165

Hydroxyethyl β-Cyclodextrin 1355 >50 +137

Hydroxypropyl β-Cyclodextrin 1250-1400 >50 +125-150

γ-cyclodextrin 1297 25.6 +175.98

Absorption and Toxicity

Cyclodextrins are not absorbed orally and not hydrolyzed during their transit

through the small intestine. They are totally resistant to β-amylase, but can be attacked by

α-amylases. The ability of these enzymes and cyclodextrintransglycoslases to hydrolyze

HPβ-CD is limited. Substitution provides steric hindrance to the binding of the HPβ-CD

to the active site of the enzymes and as a result, the amount of hydrolysis is reduces.

Hydrolysis occurs only in colon (partial hydrolysis occurs with α-CD). The oral

administration of CDs does not result in acute toxicity. Long term administration leads to

no significant change in organs or biological values. Natural CDs are highly toxic when

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 3 
given parentrally. α- and β-cyclodextrins induce haemolysis and nephrotoxicity upon i.v.

injection γ-CD is relatively less toxic parenterally5.

Formation of Complexes

One of the most important characteristics of CDs is their ability to form inclusion

complexes. Inclusion complexation involves entrapment of a guest molecule totally or

partially in the cavity of host molecule without formation of any covalent bonds. CDs are

typical host molecules and can entrap a wide variety of drug molecules resulting in the

formation of mono molecular inclusion complexes6.

Inclusion complexation occurs when aqueous solutions of CD are shaken with

drug molecules or its solution. In aqueous solution the hydrophobic cavities of CD are

occupied by water molecules, which can be replaced by appropriate drug molecules that

are CD and hence the complex may be precipitated from its saturated solution, as micro

crystalline powder and this powder is subsequently separated by filteration7 .

The central cavity of the CD molecule is linked with skeletal carbon and ethereal

oxygen’s of the glucose residue. It is there fore lipophilic; the polarity of the cavity has

been estimated to be similar to that of the aqueous ethanolic solution8. It provides a

lipophilic microenvironment into which suitably sized drug molecules may enter and be

included. No covalent bonds are formed or broken during CD complex formation, and in

aqueous solutions, the complexes are readily dissociated. Free drug molecules are in

equilibrium with the molecules bound within the CD cavity. Measurements of stability or

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 4 
equilibrium constants (Kc) or the dissociation constants (Kd) of the drug CD complexes

are important properties of a compound upon inclusion.

Stoichiometry of Complexes

Stoichiometry is usually 1:1 molar ratio of CD to drug, but guest molecule is large

which will not fit into cavity of one host molecule, instead of that 2:1 (CD: Drug) 9 some

times 3:110 or 5:111 complexes are formed. It may also be possible to form 1:212 and 1:3

(CD: Drug) complexes if guest molecule is smaller in size. The stoichiometry of the

complexes can be determined by phase solubility studies, mass spectroscopy, and 1H-

NMR studies. The self-association and association of complexes are also possible.13

In the case of a 1:1 complex, using the following equation one can determine the

equilibrium binding or association constant, K, from the slope of the linear portion of the

curve.

Ka: b = slope/S0 (1 – slope)

Where so is the intrinsic solubility of the drug studied under the conditions.

Equilibrium binding of drug and CD to form a 1:1 complex can be represented as

Drug + CD Drug: CD Complex

Since equilibrium binding usually establishes with half-lives of much less than 1

second, the kinetics of dissociation of drug/CD complexes are generally expected to be

much faster than many physiological processes.14, 15


CDs and their derivatives have

received considerable attention in the pharmaceutical field for the past few years and an

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 5 
increased number of reviews have been dedicated to their industrial and pharmaceutical

applications.16, 17-25

Degree of Substitution

The physicochemical properties of CDs, including their complexation ability, may

be greatly affected by the type, number, and position of the substituent on the parent CD

molecule. The “degree of substitution” per se does not uniquely characterize a β-CD

derivative such as HP-β-CD. When produced under different conditions, the

physicochemical properties of HP-β-CD samples with same degree of substitution may

not be identical owing to the possible occupancy of hydroxypropyl groups at different

positions on the parent CD molecule. Since the purity of CD can have a significant effect

on the final quality of the drug product and its marketability, it is necessary to have a

proper understanding of the following terms that are used in identification of CD purity.26

Degree of substitution (DS):

The average number of substituted hydroxyls per glucopyranose unit of CD ring.

Since the number of reactive hydroxyls per mole of glucopyranose unit is 3, the

maximum numbers of substituent’s possible for α-, β-, and γ-CDs are 18, 21, and 24,

respectively.

Average molar degree of substitution (MS):

The average number of moles of the substituting agent, e.g., hydroxypropyl, per

mole of glucopyranose. It may not necessarily describe the extent to which the reactive

sites are substituted when the substituting agent itself has reactive sites, or when new

reactive sites are generated during the substitution reaction.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 6 
Thus the value of MS can be more than 3 for each glycopyranose unit of

substituted CDs, or more than 18, 21,and 24 for α-, β-, and γ-CDs, respectively.

Degree of polymerization (DP):

The ratio of MS to DS (MS / DS). If no additional reactive sites are produced

during the substitution, MS and DS are equal and the DP becomes 1.

Total Degree of Substitution (TDS):

It avoids the confusion between DS and MS and represents the average number of

substituted groups (e.g., hydroxypropyl) per CD molecule. If the MS and DS are known,

one can calculate the molecular weight (Mw) of HP-β-CD from the following equation

Mw = 58:08* (TSD) + 1135

Where 1135 and 58.08 are the molecular weights of β-CD and propylene oxide

respectively. In the case of β-CD with 7 glycopyranose units, the TDS is 7*MS and hence

the equation becomes

Mw = 406:56* (MS) + 1135

HP-β-CDs

Degree of substitution (DS) plays an important role in balancing the CD water

solubility and its complexing ability. It was reported that increasing the degree of

substitution up to an optimum level improves the CD aqueous solubility, but beyond that,

the steric hindrances of the host molecule impair CD complexing (efficiency) capacity.

HP-β-CD derivatives with a low degree of substitution showed the best complexing

properties with low surface activities. Binding of guests to these CD derivatives was very

similar to β-CD at low degrees of substitution, but, as the substitution increased, the steric

hindrances weakened the binding and the effect was dependent upon the particular guest.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 7 
Though increasing the degree of substitution can increase the binding of guests to CDs by

increasing the surface area of binding, in many cases the differences in the binding of

guests with degree of substitution were small, if detectable.27

Drug Solubilization and Enhanced Dissolution:

Solubility of slightly and sparingly soluble drugs can be enhanced by molecular

inclusion within the CDs. The solubility and dissolution rate of sparingly soluble drugs is

enhanced using CD complexes; hence the drug reaches the blood faster and in higher

concentration suggesting the need for reducing the dose size.28 CD applications as

solubilizing agents are summarized in Table No 3.

Charged CD can be powerful solubilizer29 but their solubilizing effects appear to

depend on the relative proximity of the charge to the CD cavity. The farther away the

charge is located the better complexing abilities. For example, (2-hydroxy-3-

[trimethylammonio] propyl-β-cyclodextrin possesses excellent solubilizing effects while

β-cyclodextrin sulfate has a comparatively low complexation potential.

Sulfobutyl β-cyclodextrin, where a butyl ether spacer group has moved the anion

away from the cavity, is an excellent solubilizer. Compared to neutral CD, enhanced

complexation is frequently observed when the drug and CD molecules have opposite

charge but decreased complexation is observed if they carry same type of charge. For

example, (2-hydroxy-3-[trimethylammonio] propyl-β-cyclodextrin is an excellent

solubilizer for many acidic drugs capable of forming anions.

Many ionizable drugs are capable of forming CD complexes; the stability

constant of the complex is much larger for the un-ionized than for the ionized form. For

example, both the un-ionized and the cationic form are 4 times larger than for the cationic

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 8 
form. In the case of ionisable drugs, the presence of charge may play a significant role in

drug/CD complexation and hence a change in the solution pH can vary the complex

constant. In general, ionic forms of drugs are weaker complex forming agents than their

nonionic forms.30, 31

β-CD complexes.32 When added in small amounts, water-soluble polymers or ion

pairing agents enhance CD solubilizing effect by increasing the apparent complex

stability constant. The polymers or ion pairing agents due to their direct participation in

drug complexation, improve both pharmaceutical and biological properties of drug/CD

complexes, independent of drug’s physiochemical properties.25,33,34-38 Certain additives

may compete with drug molecules for CD cavities and thus decrease the apparent

complex stability constant, eg, additives with positive and negative hydrotropic

effect.39,40 Though water structure forming agents added to CD solutions generally

increase the total drug solubility, they showed opposite effects with clotrimazole.56

Simultaneous complexation and salt formation with hydroxy carboxylic acid (HA)

significantly increased the CD solubilizing power for a sparingly water-soluble amine

type drug by forming drug/ CD/HA multicomponent systems.41 Co-solvents can improve

the solubilizing and stabilizing effects of CDs, eg, use of 10% propylene glycol in

development of an oral itraconazole preparation containing 40% of HP-β-CD.42

Sometimes co-solvents may hinder drug complexation by competitive inclusion, eg,

presence of 10% propylene glycol decreased the solubilizing effect of HP-β-CD for

itraconazole. On dilution, the presence of propylene glycol favored absorption and

precipitation of itraconazole in GI fluids and formulation by providing increased

percentage of the free drug. The increased percentage of the free drug in presence of co-

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 9 
solvent was reported to be a result of lesser intrinsic solubility of the drug compared with

the dilution concentration line at a given HP-β-CD concentration.43

Temperature changes can affect drug/CD complexation. In most cases, increasing

the temperature decreased the magnitude of the apparent stability constant of the drug/CD

complex and the effect was reported to be a result of possible reduction of drug/CD

interaction forces, such as van derWaals and hydrophobic forces with rise of

temperature.31, 44, and 45

However, temperature changes may have negligible effect when the drug/CD

interaction is predominantly entropy driven (ie, resulting from the liberation of water

molecules hydrated around the charges of guest and host molecules through inclusion

complexation).46

Method of preparation, viz co-grinding, kneading, solid dispersion, solvent

evaporation, co-precipitation, spray drying, or freeze drying can affect drug/CD

complexation. The effectiveness of a method depends on the nature of the drug and

CD.47, 48, 49 in many cases, spray drying, 49-51 and freeze drying52, 53, 54 were found to be

most effective for drug complexation. However, method of preparation showed no

influence on the dissolution performance of tolbutamide.

Reduction of drug crystallinity on complexation or solid dispersion with CDs also

contributes to the CD increased apparent drug solubility and dissolution rate.57, 58 CDs, as

a result of their ability to form in situ inclusion complexes in dissolution medium, can

enhance drug dissolution even when there is no complexation in the solid state.59

SBE-β-CD was shown to be an excellent solubilizer for several drugs and was

more effective than β-CD but not as effective as DM-β-CD.60 CDs can also act as release

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 10 
enhancers, for example β-CD enhanced the release rate of poorly soluble naproxen and

ketoprofen from inert acrylic resins and hydrophilic swellable (high-viscosity hydroxy

propyl methyl cellulose [HPMC]) tableted matrices. Β-CD also enhanced the release of

theophylline from HPMC matrix by increasing the apparent solubility and dissolution

rate of the drug.193, 194

Figure No.1. The chemical structure (A) and the toroidal shape (B) of the â-cyclodextrins

molecule.

(C) HPβ-CD

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 11 
C R= CH2CH (OH) CH3 or H

Table No 3. Examples of CD-enhanced Solubility and Dissolution

CD Drug(s) Ref.

β-CD Nimesulide, Sulfomethiazole, Lorazepam,


Ketoprofen, Griseofulvin, 48, 54, 59, 61-
69
Praziquantel,Chlorthalidone, Etodolac,
Piroxicam,, Itraconazole
Ibuprofen

α-CD Praziquantel 59

γ-CD Praziquantel, Omeprazole, Digoxin 59,70,71

HP-β-CD Albendazole, DY–9760e, ETH–615,


Levemopamil HCl, 52, 46,
Sulfomethiazole, Ketoprofen, 33, 72, 54
Griseofulvin, Itraconazole, 62, 64, 68, 57,
Carbamazepine Zolpidem, Phenytoin, Rutin 73-75

DM-β-CD Naproxen, Camptothesin 58, 76

SBE-β-CD DY– 9760e, Danazol, Fluasterone, Spiranolactone 45,46,77,78

RM-β-CD ETH–615, Tacrolimus 33, 79


Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 12 
80

Permeability Enhancement

Masson81 reported that cyclodextrins could enhance the permeation of poorly

soluble drugs through biological membranes. However, the permeability will decrease at

higher concentration of cyclodextrin in excess of the concentration needed to solvate the

drug. The effect of cyclodextrins cannot be explained as solely due to increased solubility

of the drug in the aqueous donor phase nor can it be explained by assuming that

cyclodextrins act classical permeation enhancer. It was concluded that cyclodextrins act

as permeation enhancers by carrying the drug through the aqueous barrier which exists

before the lipophilic surface of biological membranes.

Martin82 explained nasal permeation is enhanced by interaction with nasal

epithelium by modifying tight junction and lipid and protein content of the membrane,

which enhances the permeation of membrane. The enhanced effect is proportionally some

extent with increase in concentration up to a critical value and further increase in

concentration failed to increase the permeation. At high concentrations of CD, the

permeability coefficient decreases due to decreased drug activity in the donor phase.83

The transport enhancing properties of the CD were found to follow the

lipophilicity of the core in their cyclic structure was observed by Hovgaard.84 Dimethyl-

β-CD (DM-β-CD) caused in an increase in the permeability of the cytoplasm membrane

in a concentration dependent manner. Hovgaard et al further observed that the

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 13 
basolateral membrane was significantly more sensitive to cyclodextrin than the apical

membrane.

HPβ-CD with a molar substitution of 0.6 and randomly methylated β-CD with

molar substitution of 1.8 and carboxymethyl-β-CD sodium salt with molar substitution of

0.6 were evaluated by Masson et al. Excess of the concentration needed to solvate the

drug the effect of permeation enhancer reduces.85

Moreover it was found from in-vitro electrophysiological experiment that the

change in enhanced permeability caused by β-CD occurred primarily in the transcellular

pathways, rather than in the paracellular pathways of the small intestine. They suggested

that the enhancement of intestinal absorption by β-CD, after removal of the mucin layer

from the intestinal surface, is due to the interaction between the membrane components

and CD. This interaction would induce disorder in cell membrane lipid, resulting in the

increased permeability of the transcellular route.2

Safety of CD in Oral Drug Delivery

Safety of CD in oral drug delivery is proved. It was reported that the acute LD50

of β-CD is more than 12.5 g/kg in mice, 18.8 g/kg in dogs. The acute LD50 of γ-CD is

more than 16 g/kg in mice and more than 8 g/kg in rats.87, 88 In another 90-day and 6-

month oral toxicity studies in rat (Long Ewans, 5 weeks old), daily of 200, 400, and 600

mg/kg β-CD did not show any growth depressant effect. At end of the experiment,

examination of the urine for colour, pH, protein, blood glucose, urobilinogen, bilirubin,

ketenes’ and sediment did not show any changes due to treatment with CD. Even

hematological examination, which was conducted at the end of experiment and

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 14 
comprised of hemoglobin, cell volume, counts of erythrocytes, and of total and

differential leucocytes, showed only values that were within the normal range.

Biochemical variables, including glucose, serum alkaline phosphatase; total

bilirubin, total protein and creatinine did not show any abnormalities.89, 90
Further in

autopsy, the weights of the heart, lungs, liver, kidneys and spleen were found to be in

normal range. Microscopic examination of the liver, spleen, kidneys, gonads, uterus,

stomach, small intestine, pancreas, adrenals, lungs, myocardium and brain showed that

there are no pathological changes that could be attributed to treatment with β-CD.

Based on other studies, CD administered orally to rats and dogs is considered to

be non-toxic if the daily dose is less than 600 mg/ kg or not more than 3% of the diet. No

signs of teratogenicity at 200, 400, 600 mg/kg β-cyclodextrin as an oral daily dose in

female rats. There was no evidence for an effect of treatment on the number of

implantations, the percentage of resorptions, and the percentage of dead and live fetus.

CDs have been used to ameliorate the irritation caused by drugs.91 the increased

drug efficacy and potency (i.e., reduction of the dose required for optimum therapeutic

activity), caused by CD-increased drug solubility, may reduce drug toxicity by making

the drug effective at lower doses.

β-CD enhanced the antiviral activity of ganciclovir on human cytomegalovirus

clinical strains and the resultant increase in the drug potency reduced the drug toxicity.92

The toxicities associated with crystallization of poorly Water-soluble drugs in parenteral

formulations can often be reduced by formation of soluble drug: CD complexes.

Formulation of phenytoin with HP2-β-CD showed considerably reduced tissue

irritation compared with a commercial injection of the drug in a BALB/c mouse model.93

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 15 
Further CD entrapment of drugs at the molecular level prevents their direct contact with

biological membranes and thus reduces their side effects (by decreasing drug entry into

the cells of nontargeted tissues) and local irritation with no drastic loss of therapeutic

benefits.23

Inclusion complexation with HP-β-CD reduced the side effects of 2–ethyl hexyl–

p–dimethyl aminobenzoate (a UV filter) by limiting the interaction of the UV filter with

skin.94 In a study with patients, piroxicam/β-CD inclusion complex showed better

tolerance with lower incidence and severity of gastrointestinal side effects compared with

the free drug.95

HP-β-CD alleviated the intrinsic irritancy effect observed on IV administration of

CKD-732 hemioxalate against blood vessels.96 SBE7-β-CD inhibited DY-9760e-induced

cytotoxicity toward human umbilical vein endothelial cells and significantly suppressed

the drug-induced vascular damage in rabbits.97 Inclusion complexation with CDs also

reduces ocular drug irritation by limiting the free drug concentration on the precorneal

area to a nonirritating level.98

Effect on Drug Stability

CDs can improve the stability of several labile drugs against dehydration,

hydrolysis, oxidation, and photodecomposition and thus increase the shelf life of drugs.16

Table No 4 summarizes CD effects on drug stability.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 16 
Table No 4. CD Effect on Drug Stability

Effect Drug CD Ref.

↑Photostability Promethazine HP-β-CD, DM-β-CD


33
DY–9760e SBE-β-CD
46
2–ethyl hexyl p–dimethyl HP-β-CD
94
aminobenzoate
↑ Shelf life with unaffected
dissolution rates for 4 years Glibenclamide β-CD
99

↑ Thermal stability in solid state Diclofenac sodium β-CD


100
↑Stability against intramolecular
cyclization in solid state Quinaril β-CD, HP-β-CD
101
↑Stability to acid hydrolysis and
photodecomposition Doxorubicin HP-β-CD, HP-γ-CD
102
↑Stability against hydrolysis acyl ester prodrugs of HP-β-CD
62
Ganciclovir
Digoxin γ-CD
71
Rutin HP-β-CD
75
Camptothesin RDM-β-CD
76
Melphalan and Carmustine SBE -β-CD, HP-β -CD
103
Paclitaxel γ-CD, HP-γ-CD, HP-
β-
CD
104
↑ Deacetylation or degradation Spiranolactone SBE-α-CD, SBE-β-
CD,
HP-β-CD, γ-CD,β-CD
105

↑Photoreactivity Flutamide β-CD 106

↑, increased effect

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 17 
It was reported that CD-induced enhancement of drug stability may be a result of

inhibition of drug interaction with vehicles and/or inhibition of drug bioconversion at the

absorption site.107 by providing a molecular shield, CD complexation encapsulates labile

drug molecules at the molecular level and thus insulates them against various degradation

processes. SBE-β-CD showed greater stability enhancement of many chemically unstable

drugs than other CDs.60

The stabilizing effect of CDs depends on the nature and effect of the included

functional group on the drug stability and the nature of the vehicle. Both the catalyzing

effects of the nitro group as well as the stabilizing effect of the halogen and cyanogen

groups on photodegradation of 1, 4 dihydropyrimidine derivatives were reduced by

complexation with CDs.108

HP-β-CD significantly reduced the photodegradation of 2–ethyl hexyl p–

dimethyl aminobenzoate in solution than in emulsion vehicle.94 CDs improved the

photostability of trimeprazine (when the solution pH is reduced) 109 and promethazine.110

CDs also enhanced the solid state stability and shelf life of drugs.99-101

CDs were reported to enhance the physical stability of viral vectors for gene

therapy, and the formulations containing sucrose and CDs were stable for 2 years when

stored at 20°C.111 Since the hydrolysis of drugs encapsulated in CDs is slower than that

of free drugs, 102 the stability of the drug/ CD complex, ie, the magnitude of the complex

stability constant, plays a significant role in determining the extent of protection.46,


76,103,112

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 18 
Very low concentrations of HP-β- CD (1% or lower), due to formation of a more

physically unstable complex, did not protect taxol as effectively as higher CD

concentrations. The effect of complexation on drug stability can be represented by the

following equation (Figure 2)

1 1 1

---------- = ---------------------- + ----------

k0 − kobs Kc (k0− kc) (CD) (k0− kc)

Figure No.2. A simple model representing the effect of complex stability constant on

drug degradation.104

Enzymatic Degradation, Absorption

One of the outstanding properties of CDs is their resistance to enzymes that

hydrolyse starch. CDs are resistant to β-amylases, since they do not contain terminal

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 19 
groups susceptible to attack by the enzyme. By contrast, α-amylases, which do not

require free terminal groups and attack the inside of the molecule is capable of

hydrolyzing cyclodextrins, though, usually at a low rate. Gerloczy reported that 2% of the

input radioactivity could be detected in blood after 6 to 12 hours, when the labeled β-

cyclodextrin was administered orally. Hence oral absorption of cyclodextrin is very less.
113,114

Increased Bioavailability

Enhanced oral bioavailability of a drug from a cyclodextrin-containing

formulation mainly results from increases in the apparent solubility of the drug. The

mean absorption time and mean residence time was shortened with an increase in the

molar ratio of cyclodextrin to drug. The maximal AUC was observed with a 1:30

kneaded mixture. The bioavailability of each mixture was almost the same as or slightly

higher than a commercial cyclosporine preparation.

CDs enhance the bioavailability of insoluble drugs by increasing the drug

solubility, dissolution, and/or drug permeability. CDs increase the permeability of

insoluble, hydrophobic drugs by making the drug available at the surface of the biological

barrier, e.g., skin, mucosa, or the eye cornea, from where it partitions into the membrane

without disrupting the lipid layers of the barrier. In such cases it is important to use just

enough CD to solubilizing the drug in the aqueous vehicle since excess may decrease the

drug availability (Figure 3).115, 116, 117

At low RM-β-CD concentrations, when hydrocortisone was in suspension,

increasing the CD concentration increased the drug flux. At higher CD concentrations,

when the drug was in solution, increasing the CD concentration decreased the flux.118 It

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 20 
was found that addition of polymers can further enhance the drug permeability from

aqueous CD solutions. Carboxy methyl cellulose (CMC) enhanced triclosan

bioavailability from toothpastes containing β-CD by forming a drug/CD/CMC complex

with improved substantivity.119

CDs increased the bioavailability of lipophilic itraconazole from both an oral

solution and an intravenous formulation by improving the drug solubility and

absorption.120

In the case of water-soluble drugs, CDs increase drug permeability by direct

action on mucosal membranes and enhance drug absorption and/or bioavailability.107, 115

Solubilization of specific membrane lipids of human erythrocytes through inclusion

complexation with CDs and their ability to cause perturbation of membrane integrity,

were suggested to contribute to CD-induced promotion of drug absorption and toxicity.

It was reported that CDs, because of their ability to remove cholesterol, may

increase membrane fluidity and induce membrane invagination through a loss of bending

resistance and cause cell lysis. On the other hand, removal of phospholipids, especially

phosphatidyl choline and sphingomyelin from the outer half of the membrane bilayer by

CDs causes bilayer imbalance; the removal may also contribute in part to the formation

of stomatocytes through an inward bending of membranes. CD induced lysis of artificial

membranes composed of lecithin and cholesterol by a similar solubilization process.

Detergents first incorporate themselves into membranes, and then extract the

membrane components into micelles and cause membrane solubilization/ lysis. However,

unlike detergents, CDs were reported to solubilizing membrane components without

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 21 
entering into the membrane, and hence the perturbing effects of CDs can be mild and

reversible.

In the presence of CDs, the new lipid containing compartment in the aqueous

phase with extracted components from the erythrocyte surface equilibrated freely with

the cell surface by a reversible process.

Compared with other absorption-promoting agents and preservatives commonly

used in nasal formulations, CDs exerted a rather mild and reversible effect on the ciliary

beat frequency of both chicken embryo trachea and human nasal adenoid tissue in vitro in

a concentration-dependent manner.21 DM- β-CD caused enhancement of enoxaparin nasal

absorption by solubilizing membrane components and opening tight junctions but the

effect was reversible after 6 hours.121

Watanabe et al122 reported that rectal membrane recovers its barrier function

probably 24 hours after the administration of DM-β-CD (at least 30 mg).107 Even at high

doses, the effects of HP-β-CD on kidneys were reversible and similar to those of osmotic

agents currently used in parenteral formulations.115 Labile drug stabilization by CDs71,75

and their ability to ameliorate drug irritation, and thus improve drug contact time at the

absorption site in nasal, ocular, rectal, and transdermal delivery,115 are some other

important factors that contribute to the CD-improved bioavailability. α-CD improved the

rectal bioavailability of morphine by inhibiting the drug’s upward movement from areas

impacted by first pass metabolism.107

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 22 
Figure No.3. Mode of penetration enhancement by CDs.91

CD APPLICATIONS IN DRUG DELIVERY:

Oral Drug Delivery

When a drug becomes part of a CD complex, its physicochemical properties such

as crystallinity, Solubility, dissolution and chemical stability are modified.123 hence, some

of pharmaceutical formulation problems may be solved. CD technology has also been


124
applied for naproxen, indomethacin125 in order to decrease drug irritation in oral

administration.

Applications of CDs in oral drug delivery include improvement of drug

bioavailability due to increased drug solubility, improvement of rate and extent of

dissolution, and/ or stability of the drug at the absorption site, eg, the gastrointestinal tract

(GIT) or in formulation, reduction of drug induced irritation, and taste masking (Table

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 23 
10). CD complexation was found to decrease local drug irritation and also modify the

time of drug release during GI transit.115, 22

CDs enhance the mucosal drug permeability mainly by increasing the free drug

availability at the absorptive surface.126, 127


CD complexation can provide better and

uniform absorption of low-soluble drugs with poor and erratic absorption79 and also

enhance the drug activity on oral administration128,129,130 CD complexation increased the

absorption of poorly water-soluble drugs, delivered via buccal or sublingual mucosa.131-


134

Complexation can also mask the undesirable taste of drugs. With the assumption

that only the free drug molecule exhibits bitter taste, the extent of the suppression was

reported to be dependent on the availability of free drug, regardless of the kind and

concentration of CD.135

The relative safety, efficacy in terms of complexation, cost, and acceptance in

pharmacopeias are some important factors to be considered in selecting a CD for drug

complexation. HP-β-CDs were shown to have a better oral safety profile than β-CD and

other parent CDs, but only limited data are available on the oral safety of the methylated

CDs. However, for oral administration all CDs can be considered practically nontoxic

due to lack of CD absorption through GIT and, hence, the relative safety profile of CDs is

a concern of drug doses used in drug/CD complexes and the LD50 of CD.115 β-CD is the

most cost-effective compound of all CDs, whereas HP-β- and SBE-β-CDs are more

expensive. Monograph of β-CD is already incorporated in various pharmacopeias and

national formularies (NF).16 Hence, β-CD can be considered optimum for oral use when

it is effective for drug complexation and modified CDs like HP-, SBE-β-, and DM-β-CDs

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 24 
may be used when they are more effective and when their peculiar property is required in

formulation, e.g., SBE-β-CD, owing to its osmotic property, was used in the preparation

of osmotic pump tablets.136,137

In Parenteral Drug Delivery

The sparingly water soluble drugs for intravenous and intramuscular dosing can

be given in the form of cyclodextrin complex by increasing solubility, decreasing

irritation at the site of administration of these parentrally administered drugs and

stabilization of drugs which are unstable in an aqueous environment. γ-CD, 2-HP-β-CD,

sulfobutylether β-CD, sulphated-β-CD and maltosyl-β-CD seems to be safe when

administered parentrally other cyclodextrins cannot be used in parenteral dosage forms.

One factor is whether the target drug solubility can be achieved with the use of an

acceptable cyclodextrin concentration. In addition, the cyclodextrin safety data must

support the concentration and total dose of cyclodextrin required to solubilizing the

required amount of drug. Many other factors such as the linearity of the relationship

between drug solubility and cyclodextrin concentration might also affect the acceptability

of a given concentration. Important critical factor could be whether the drug will be

completely released from the cyclodextrin dosage form.13

Intravenous Administration

So far SBE4-β-CD or HP-β-CD was utilized for I.V. administration. Significant

changes in the Pharmacokinetics of a drug were also demonstrated in the work of Frijlink
139.
They reported that higher values in some tissues may be due to transitory alteration in

protein binding.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 25 
The absorption, distribution and excretion of intravenously and orally

administered β-CD and glucosyl-β-CD in rats were studied by Kubota140. They reported

that 90% unchanged β-CD and glucosyl-β-CD was recovered in urine after 10 hours and

pharmacokinetics of the both complexing agents is similar.

Ophthalmic Applications of Cyclodextrin

Recently the strengths and weaknesses of the use of CD in this application have

been recognized. First to be considered is there should have affect on integrity of cornea.

It was assumed that only free drug, not the complex of drug with cyclodextrin penetrates

across biological membranes.

Cyclodextrin and Corneal Permeability

CD may increase the ophthalmic delivery of drugs through multiple routes. One

route would be to increase the solubility of poorly water-soluble drugs. A second route

would be to alter corneal permeability by damaging the corneal membrane. The most

beneficial area of CDs might be the solubilization of drugs intended for ophthalmic use.

Usayapant141 studied and showed that the solubility, chemical stability, and

ophthalmic delivery of dexamethasone were improved in the presence of HPβ-CD.

Cyclodextrins as anti-irritants

As high concentration of drug is used in ophthalmic, irritation is a common

drawback in ophthalmic drug development, which may decrease patient compliance.

Mainly CDs may decrease the irritation of drugs by formation of inclusion complexes,

thereby masking the irritating nature of the drug. Since ophthalmic drug irritation is

mostly related to drug concentration on the corneal surface tissues, CDs may be able to

eliminate the ocular irritation by changing the rate of drug absorption.142

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 26 
Cyclodextrins are useful excipients in eye drop formulations for a variety of

lipophilic drugs. They will facilitate eye drop formulations for drugs that otherwise might

not be available for topical use, while improving absorption and stability and decreasing

local irritation.

Prevention of additive-drug interaction

Due to carbopol-propranolol interaction, insoluble complex is formed which alters

the properties of polymer such as swelling, releasing and bioadhesion. This interaction

was reduced when drug is complexed with β-CD.143

Cyclodextrins are not only well-known solubilizers, but constitute very powerful

tool as permeation enhancers. CD enhances the permeability of drug either by decreasing

the resistance of aqueous barrier or by modification of the structural lipid layer of

biological membrane. In addition, CDs are used to reduce or prevent GI or ocular

irritation, reduce or eliminate unpleasant tastes, prevent drug-drug interactions or drug-

additive interaction.

Nasal Drug Delivery

CDs are effective excipients in nasal drug delivery. CDs improve nasal drug

absorption either by increasing aqueous drug solubility and/or by enhancing nasal drug

permeability. However, large interspecies differences were found in CD-enhanced nasal

drug absorption. The safety and nontoxicity of CDs in nasal drug formulations have been

demonstrated by the clinical data with CDs showing no adverse effects.

Merkus et al195 demonstrated that CDs can be safely used to improve nasal

bioavailability of drugs, especially peptides.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 27 
CDs can also be used to reduce the nasal toxicity of other enhancers without

affecting their absorption-enhancing property.

Rectal Drug Delivery

Applications of CDs in rectal delivery include enhancing drug absorption from a

suppository base either by enhancing drug release from the base or by increasing drug

mucosal permeability, increasing drug stability in the base or at the absorption site,

providing sustained drug release, and alleviating drug induced irritation.107, 15

Drug release from the suppository base is important in rectal absorption because

of the high viscosity of rectal fluids. The effect of CDs on rectal drug absorption can be

influenced by partition coefficient of the drug and its CD complex, magnitude of the

complex stability constant, and nature of the suppository base (oleaginous or

hydrophilic). Hydrophilic CDs (especially methylated and hydroxylpropyl CDs) enhance

the absorption of lipophilic drugs by improving the drug release from oleaginous vehicles

and/or by increasing the drug dissolution rate in rectal fluids.

Formation of hydrophilic CD complexes was found to inhibit the reverse

diffusion of drugs into oleaginous vehicles by reducing the drug/vehicle interaction. CDs

may not affect drug release if the drug/CD complex dissociates in the vehicle itself.

The CD complex, once released from the base, mostly releases the free drug for

absorption. The competing sites for the free drug released at the absorption site are CD

cavity, suppository base, and the rectal mucosa. The extent of drug diffusion into these

sites depends on drug’s partition coefficient, magnitude of the stability constant of the

drug/CD complex, and the relative lipophilicity of the competing sites. In the case of

lipophilic drugs with a high partition coefficient, there might be some back diffusion of

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 28 
the released free drug into the lipophilic base. Since a part of drug may get absorbed as

the CD complex, the partition coefficient of the complex also becomes important.

CDs enhance the rectal absorption of inabsorbable, hydrophilic drugs such as

antibiotics, peptides, and proteins by their direct action on the rectal epithelial cells.107

CDs enhance rectal drug stability either by inhibiting the drug/vehicle interaction

(by making the drug insoluble in oleaginous base) or by inhibiting the drug bioconversion

in the rectum.

Controlled Drug Delivery

CDs, due to their ability either to complex drugs or to act as functional carrier

materials in pharmaceutical formulations, can serve as potential candidates for efficient

and precise delivery of required amounts of drugs to targeted site for a necessary period

of time.

β-CD derivatives are classified as hydrophilic, hydrophobic, and ionizable

derivatives. The hydrophilic derivatives improve the aqueous solubility and dissolution

rate of poorly soluble drugs, while the hydrophobic derivatives retard the dissolution rate

of water-soluble drugs from vehicles. Hence hydrophilic and hydrophobic CD derivatives

are used in immediate and prolonged release type formulations, respectively.

The ionizable CD derivatives, on the other hand, improve inclusion capacity;

modify drug dissolution rate, and all eviate drug irritation, e.g., use of CME-β-CD to

obtain delayed release type formulations. Highly hydrophilic derivatives, such as 2HP-β-,

G2-β-, and SBE-β-CDs were used in immediate release formulations that dissolve readily

in the GIT and enhance the oral bioavailability of poorly soluble drugs.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 29 
CDs, both natural and chemically modified, are used in the design of immediate,

delayed release, and targeted drug delivery systems (Table 5). The pH-dependent

solubility of CME-β-CD (ie, limited solubility under the acidic conditions of stomach

with the complex solubility increasing with pH), which provides selective dissolution of

drug/CD complex, makes it useful in the design of enteric formulations. When

molsidomine tablets containing CME-β-CD were studied in gastric acidity–controlled

dogs, the absorption of the drug was significantly retarded under high gastric acidity

compared with low gastric acidity conditions.145, 146

Table No 5. Modification of the Drug Release Site and/or Time Profile by CDs145

Release Pattern Aim Use of CD

Immediate release Enhanced dissolution and absorption HP-β-, DM-β-,


SB-β-,
of poorly water soluble drugs and branched-
β-CDs

Prolonged release Sustained release of water-soluble Ethylated β-


CDs, acylated β-CDs drugs

Modified release More balanced oral bioavailability Simultaneous


use of
With prolonged therapeutic effect different
CDs and/or
other
excipients

Delayed, pH-dependent
Release (Enteric) Acid protection of drugs CME-β-CD

Site-specific release Colon-targeting Drug/CD


conjugate

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 30 
Hydrophobic CDs, such as alkylated and acylated derivatives, are useful as slow-

release carriers in prolonged release formulations of water-soluble drugs. Applications of

various CD derivatives in formulation of modified-release preparations are summarized

in Table 6.

Table 6. Applications of Various CD Derivatives in the Formulation of Modified

Release Preparations145, 146

Derivative Drug Summary

Diethyl-β-CD Diltiazem Sustained release for oral use


Buserelin acetate Sustained release for
subcutaneous use
Nitroglycerine Sustained release for
percutaneous use
Isosorbide dinitrate Sustained release
Tiaprofenic acid Delayed release

Triacetyl-β-CD Flufenamic acid Prolonged release for oral use

Peracylated-β-CD (TB-β-CD) Molsidomine Sustained release for oral use


Salbutamol Prolonged release for oral use
Captopril Sustained release

Al-β-CD-sulfate Recombinant
human basic
fibroblast Sustained release for oral use
growth factor enhanced stability

O-carboxymethyl- Molsidomine
O-β-CD Diltiazem HCl Delayed release

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 31 
Combination of short-chain and long-chain peracylated β-CDs in an appropriate

molar ratio was suggested to be useful to provide an effectively controlled release rate of

water-soluble drugs, eg, markedly retarded release rate of molsidomine on complexation

with peracylated β-CDs.147

CDs can also be used along with other carrier materials to optimize drug release

rate. Quaglia et al148 reported that CDs can be used to modulate drug delivery from

swellable systems.

HP-β-CD, because of its dissolution enhancing effect, was found to be more

effective than β-CD in the development of controlled release nicardipine formulations.149

Combination of drug complexes with hydrophilic and hydrophobic CDs in appropriate

ratios can be a promising drug delivery system for prolonged therapeutic effect and

balanced bioavailability.

In rabbits, a sustained release nicardipine formulation, developed by mixing the

drug complexes with HP-β-CD (fast release fraction) and with hydrophobic TA-β-CD

(sustained releasing portion) in appropriate ratios, showed markedly retarded drug release

with prolonged maintenance of plasma levels.150

A sustained release 2-layered nifedipine tablet formulation was developed by

using the drug complexes with β- and HP-β-CDs.151

Use of CDs with a hydroxyapatite matrix was indicated to control the release of

chemotherapeutic agents containing toxic metals, such as Rhodium II citrate and

butyrate, and to provide localized antitumor chemotherapy with minimal side effects.15

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 32 
Colon-Specific Drug Delivery

CDs are barely hydrolyzed and only slightly absorbed in the stomach and small

intestine but are absorbed in the large intestine after fermentation into small saccharides

by colonic microbial flora. The peculiar hydrolyzing property of CDs makes them useful

for colon drug targeting.

CD-based prodrug approach was used for colon-specific and delayed drug

delivery, eg, when tested in rats with carageenan induced inflammation, the absorption of

BPAA from γ-CD prodrugs was found to be from cecum and colon in contrast to that

from the highly soluble β-CD complex, which was mainly from the small intestine.153

When studied in rats, it was found that both sugar-degrading and ester-

hydrolyzing enzymes are necessary for colon-specific release of butyric acid from its β-

CD ester conjugates.154

Drug conjugation with α-CD resulted in a delayed release–type prodrug

formulation for colon-specific delivery that all eviates the side effects of drugs while

maintaining their therapeutic effect. Complexation of triamcinolone acetonide (TA) with

β-CD improved the sphericity of microcrystalline cellulose (MCC)–β-CD-TA spherical

pellets (5:90:5) prepared by extrusion and spheronization for colon targeting.

TA complexation with the CD also facilitated the application of coating resistant

to gastric and small intestinal media and maintained the pellet integrity in dissolution

medium with no premature bursting of coatings on granule swelling.155

Peptide and Protein Delivery

Various problems associated in practical use of therapeutic peptides and proteins

are their chemical and enzymatic instability, poor absorption through biological

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 33 
membranes, rapid plasma clearance, peculiar dose response curves, and immunogenicity.

CDs, because of their bioadaptability in pharmaceutical use and ability to interact with

cellular membranes, can act as potential carriers for the delivery of proteins, peptides, and

oligonucleotide drugs.156

The existence of efflux pumps may serve as an additional barrier for nonspecific

uptake of peptides and thus can cause low peptide bioavailability. P-glycoprotein (P-gp)

is an efflux transporter present in the apical region of epithelial cells in the brain, kidney,

liver, and GI tract. Pgp opposes the transcellular drug movement in the epithelial cells

and many peptide drugs, especially hydrophobic peptides like cyclosporin A, 157

Therapeutic use of peptides across the blood brain barrier (BBB) is greatly

hindered by their very low penetration and it was reported that P-gp substrates, such as

synthetic hydrophobic peptides, can stimulate the transport of drugs across the BBB.

It was also reported that Pgp–mediated transport of peptides might play an

important role in greatly reducing their delivery to the central nervous system in vivo.158

Hence, whenever unexplainable poor peptide absorption is seen, the role of efflux pumps

should be examined.159 It was found that CDs can inhibit or impair the efflux function of

P-gp and multidrug resistance associated proteins (MRP2).

CDs can enhance physical and chemical stability of protein and peptide drugs,

and the maximum enhancing effects were reported at low CD concentrations. The

proteolytic degradation of basic fibroblast growth factor was decreased by water-soluble

β-CD sulfate.16

Β-CD improved insulin loading of alginate microspheres prepared by an

emulsion-based process. The process was suggested to be useful in the development of an

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 34 
oral insulin drug delivery system as the absorption of insulin from optimized

microspheres was found to take place from the GI region.160

Gene and Oligonucleotide Delivery

The toxicity and immunogenicity associated with viral vectors led to the

development of nonviral vectors for gene delivery. Besides the plasmid or virus-based

vector systems, “naked” nucleotide derivatives have also been investigated for possible

use as therapeutic agents through several routes of administration. Gene delivery

technologists are now testing CD molecules in the hope of finding an optimal carrier for

the delivery of therapeutic nucleic acids, however, the limitations of CDs, such as CD-

associated toxicity (e.g., DM-β-CD) have to be considered before their clinical use.161

CDs can solve many of the problems associated with in-vivo delivery of

oligonucleotides (Ons), such as their limited ability to extravasate from blood stream and

traverse cellular membranes, high degree of susceptibility to endonucleases with potential

toxicity of their breakdown products, polyanionic nature leading to nonspecific

interactions with extracellular and intracellular cationic molecules, and potential

immunogenicity. CDs can improve cellular uptake of ONs and also delay their

degradation by increasing their stability against endonucleases. Ons-adamatane

conjugates associated with HP-β-CD provided significantly increased cellular uptake of

Ons. Substitution of at least a single nucleotide of Ons with CDs improved the cellular

uptake and/or stability of Ons.

On conjugation with CDs, Ons may be delivered to the colon, an advantageous

absorption site to achieve acceptable therapeutic levels of Ons. CDs can also modulate

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 35 
undesirable side effects of Ons treatment such as immune stimulation and reduction of

platelet counts.162, 163

Neutral and amphipilic as well as cationic CDs have been used for synthesis of

novel gene delivery vectors. Neutral CDs like β-, DM-β-, and HP-β-CDs were reported to

increase DNA cellular uptake by increasing its permeability.

Polyplexes (polycation polymer/DNA composite structures) of linear, cationic, β-

CD–containing polymers (βCDPs) were found to be suitable for DNA delivery due to

their increased transfection efficiency and stability against enzymatic degradation with

low in vitro and in vivo toxicity.164

The ability of CDs to complex hydrophobic adamantine was exploited for steric

stabilization of βCDPs with hydrophilic polymers like poly (ethylene glycol). Steric

stabilization of βCDPs prevents their self-aggregation but facilitates their targeted

delivery by preventing their undesired interactions with non–self-entities.165

CDs were also found to enhance plasmid or viral-vector–based delivery of genes.

Positively charged quarternary amino and tertiary amino β-CDs significantly enhanced

the transfection efficiency of negatively charged adenoviral vector-based gene

formulations.

It was reported that the transfection enhancement by the cationic β-CDs could be

a result of increased viral internalization caused by increased viral binding to cell and

improved cell membrane permeability.166 CDs also enhanced the physical stability of

viral vector formulations for gene therapy.111

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 36 
Dermal and Transdermal Delivery

CDs have been used to optimize local and systemic dermal drug delivery.

Applications of CDs in transdermal drug delivery include enhancement of drug release

and/or permeation, drug stabilization in formulation or at absorptive site, alleviation of

drug-induced local irritation, sustaining of drug release from vehicle, and alteration of

drug bioconversion in the viable skin. Parent CDs (α-, β-,and γ-CDs) and various

chemically modified CD derivatives with extended physicochemical properties and

inclusion capacity have been used in transdermal drug delivery.107

Drug thermodynamic activity in vehicles as well as its skin/vehicle partition

coefficients can significantly affect CD induced changes in the drug permeability through

skin. CDs, by enhancing apparent drug solubility, enhance the drug thermodynamic

activity in vehicles and thus cause enhancement of drug release from vehicles. The

enhancement of drug release from vehicles by CDs in turn enhances the dermal drug

absorption by improving the drug availability at the lipophilic absorptive barrier surface

(ie, skin).107, 23

Although the drug partition coefficient (eg, a lipophilic drug) may be decreased

on complexation with CDs (eg, with hydrophilic CDs), the increased drug solubility and

thermodynamic activity in vehicles can lead to increased drug permeability through skin.

The ability of CDs to increase stability (against light and oxygen) and solubility

of sparingly water-soluble molecules made them useful in the formulation of cosmetic

products.167

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 37 
Brain Drug Delivery or Brain Targetting

The concept of Bodor’s chemical delivery system (CDS) (ie, covalent coupling of

drugs to 1-methyl-1, 4-dihydronicotinic acid through an enzymatically labile linkage,

which increases drug lipophilicity) was applied for targeting drugs such as steroids,

antitumor agents, and calcium channel antagonists to brain.

Formulation is an important and integral concern in the development of CDS,

especially those for brain targeting. Formulation development of CDS is based on the

need for appropriate dosage form, stability, solubility, and dissolution characteristics.

Brewster and Loftsson168 discussed the use of chemically modified, especially

water-soluble, CD derivatives such as HP-β-CD in the formulation development of CDS.

HP-β-CD contributed to the development and preclinical testing of several CDS by

providing a stable and water-soluble dosage form suitable for parenteral administration.

Use of CDs in the formulation of CDS can be demonstrated by the significantly improved

solubility, stability, and pharmacologic activity of CDS of thyrotropin-releasing hormone

analogs on complexation with HP-β-CD.169

The very low penetration across the BBB greatly hinders the therapeutic use of

peptides, and whenever unexplainable poor peptide absorption is seen the role of the

efflux pumps should be examined.

It was reported that P-gp–mediated peptide transport may play an important role

in greatly reducing the peptide delivery to the central nervous system in-vivo158, 159 it was

also indicated that CDs such as DM-β-CD, due to their inhibitory effect on P-gp efflux

function, may enhance drug delivery to brain.170

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 38 
CD APPLICATIONS IN THE DESIGN OF SOME NOVEL DELIVERY

SYSTEMS:

Liposomes

In drug delivery, the concept of entrapping CD-drug complexes into liposomes

combines the advantages of both CDs (such as increasing the solubility of drugs) and

liposomes (such as targeting of drugs) into a single system and thus circumvents the

problems associated with each system.

Liposomes entrap hydrophilic drugs in the aqueous phase and hydrophobic drugs

in the lipid bilayers and retain drugs en route to their destination. The fact that some

lipophilic drugs may interfere with bilayer formation and stability limits the range and

amount of valuable drugs that can be associated with liposomes. By forming water

soluble complexes, CDs would allow insoluble drugs to accommodate in the aqueous

phase of vesicles and thus potentially increase drug-to-lipid mass ratio levels, enlarge the

range of insoluble drugs amenable for encapsulation (i.e., membrane-destabilizing

agents), allow drug targeting, and reduce drug toxicity.

Problems associated with intravenous administration of CD complexes such as

their rapid removal into urine and toxicity to kidneys, especially after chronic use, can be

circumvented by their entrapment in liposomes.171-174

Liposomal entrapment can also alter the pharmacokinetics of inclusion

complexes. Liposomal entrapment drastically reduced the urinary loss of HP-β-CD/drug

complexes but augmented the uptake of the complexes by liver and spleen, where after

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 39 
liposomal disintegration in tissues, drugs were metabolized at rates dependent on the

stability of the complexes.172, 175 CD complexation can increase liposomal entrapment of

lipophilic drugs and also reduce their release from the carrier, ie, liposomes. To

encapsulate large amounts of lipophilic drugs in liposomes, a CD molecule forming an

inclusion complex with a high drug:CD ratio should be selected.

HP-β-CD, with a more lipophilic interior and considerably higher aqueous

solubility incorporated higher drug amounts in vesicles than β-CD. However, HP-β-CD,

as a result of its ability to get entrapped in higher amounts in the vesicles, also showed a

higher velocity of destabilizing effect on vesicles than β-CD.196 Complexation with CDs

can improve the stability of liposomes. Inclusion complexation can greatly increase the

chemical stability of labile drugs in multilamellar liposomes.

Microspheres

In the presence of a high percentage of highly soluble hydrophilic excipients,

complexation may not improve the drug dissolution rate from microspheres. Study of in

vivo release behavior (over 24 hours) of β-CDfrom β-CD/poly (acrylic acid) (PAA)

microspheres, prepared by a water/oil solvent evaporation technique, and indicated a high

encapsulating efficiency (990%) with potential covalent binding of the CD.176

Microcapsules

It was suggested that cross linked β-CD microcapsules, because of their ability to

retard the release of water-soluble drugs through semi permeable membranes, can act as

release modulators to provide efficiently controlled release of drugs.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 40 
Nanoparticles

Nanoparticles are stable systems suitable to provide targeted drug delivery and to

enhance the efficacy and bioavailability of poorly soluble drugs. However, the safety and

efficacy of nanoparticles are limited by their very low drug loading and limited

entrapment efficiency (with classical water emulsion polymerization procedures) that

may lead to excessive administration of polymeric material.177, 178

Two applications of CDs have been found very promising in the design of

nanoparticles: one is increasing the loading capacity of nanoparticles and the other is

spontaneous formation of either nanocapsules or nanospheres by nanoprecipitation of

amphiphilic CDs diesters.

Both the new techniques were reported to be useful due to great interest of

nanoparticles in oral and parenteral drug administration. CDs increased the loading

capacity of poly (isobutylcyanoacrylate) nanoparticles. The increased loading capacity

was reported to be a result of increased drug concentration in the polymerization medium

on addition of the drug: CD complex and increased number of hydrophobic sites in the

nanosphere structure on association of large amounts of CDs to the nanoparticles.178, 179

It was indicated that during nanoparticle formation the free drug gets

progressively incorporated into polymer network, driven by the drug partition coefficient

between the polymer and polymerization medium though there may be a simultaneous

direct entrapment of some drug/CD complex.179, 180

Compared with natural CDs, β-CDs, particularly those derivatives with 6C

aliphatic chains on the primary face, form biodegradable, nonsurfactant, highly loaded

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 41 
nanospheres and nanocapsules with low hemolytic activity.177,181,182 The their ability to

nanoassociate or form stable nanospheres.

The amount of partially acylated species in β-CDsa was also found to play an

important role in regulating the mean diameter size and suspension stability of

nanospheres.183 The β-CDa derivatives formed inclusion complexes with the drugs and

with the nanoprecipitation technique the derivatives gave nanospheres of less than 300

nm with no use of surfactants. Loading techniques and also the type of β-CDa can

influence the loading and release properties of the nanospheres.

CD USE AS EXCIPIENTS IN DRUG FORMULATION:

As excipients, CDs have been finding different applications in the formulation

and processing of drugs. Β-CD, due to its excellent compactability (varied with source)

and minimal lubrication requirements, showed considerable promise as a filler binder in

tablet manufacturing but its fluidity was insufficient for routine direct compression.

Β-CD was also found to be useful as a solubility enhancer in tablets. The ability

of β-CD to complex progesterone by wet granulation was found to be dependent on both

binder solution and mixture type.184 Complexation can cause subtle changes in the

tabletting properties of drugs or CDs that can substantially affect the stability and

tabletting performance of tablet formulations containing drug/CD complexes.

CDs also affect the tabletting properties of other excipients, eg, microcrystalline

cellulose codried with β-CD showed improved flowability, compactability, and

disintegration properties suitable for direct compression.185

In the case of high-swelling wheat starches, β-CD (1%) increased the peak

viscosity (PV) but decreased the cool paste viscosity (CPV) and in the case of low

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 42 
swelling starches, the same CD slightly decreased the PV but increased the CPV.

However, β-CD reduced the heat paste viscosity of both the starches.186

CDs were used as pellatization agents in extrusion and spheronization processes

and in the presence of β-CD up to 90% by weight, the process provided satisfactory

products.187 CDs were also indicated to stabilize protein and peptide pharmaceuticals

during spray drying.

SBE- and HPderivatives of β- and α-CDs, with different total degree of

substitution (TDS), exhibited different colligative properties, especially their osmotic

pressure (OP) increased with their TDS. With substituted CDs, the OP was above their

theoretical values but with unsubstituted γ-CD, the OP was below the expected value.

Self-association of the unsubstituted γ-CD molecules was reported to be the possible

reason for the observed low OP value of the CD.

These findings relating the OP properties of CDs can be useful in the formulation

of parenteral and ophthalmic solutions, where maintenance of OP is an important

consideration.188

Interaction of CDs with the preservatives in the formulation is an important factor

and should be investigated. It was reported that such interaction can result in reduction of

both the solubilizing effect of CD and the antimicrobial activity of preservatives, e.g.,

interaction of HP-β-CD with preservatives like benzalkonium chloride, chlorhexidine

gluconate, chlorambutanol, methylparaben, and propylparaben.189

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 43 
Methods for detection of inclusion complex formation and Determination of

complex stability constant

One of the most interesting properties of CDs is their ability to form inclusion

complexes with a wide variety of guest molecules. Molecular encapsulation may occur

both in solution and solid state. In solution there is equilibrium between complexed and

non complexed guest molecules, in solid state guest molecules can be enclosed within

the cavity or may be aggregated to the outside of CD molecule190. Upon inclusion within

the CD cavity a guest molecule experiences changes in its physico-chemical properties.

These changes provide method to detect whether guest molecules are really included in

the in the CD cavity

Detection inclusion complexation in the solution state

Detection of inclusion complexation in solution states can be done by

spectroscopic methods like UV/VISIBLE, Fluorescence, circular dichroism, ESR, NMR,

methods. The 1H-NMR and 13


C-NMR spectroscopic studies can also be used to

determine the direction of penetration of the guest molecules into the CD cavity. Other

methods include Polarography, conductivity measurement, Microcalorimetry and

Solubility methods191. Phase solubility technique 192 is one of the widely used methods to

detect the inclusion complexation in solution state.

The general experimental operation in studying molecular interaction by means of

phase solubility methods entails the addition of an equal weight (inconsiderable excess of

its normal solubility) of a slightly soluble compound, S (substrate or guest) into each of

several vials containing increasing concentrations of a relatively soluble compound, L

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 44 
(ligand or host or complex agent), which are closed and brought to solubility equilibrium

at constant temperature. The solution phases are then analyzed, by any suitable means,

for their total concentration of compound S (guest), no matter what its molecular state

may be.

A phase diagram is constructed by plotting, on the vertical axis, total molar

concentration of S found in the solution phase against the molar concentration of L.

Figure No. 4. Theoretical phase solubility diagram

The phase diagrams (Fig 4) are observed to fall into two main classes, type A and

type B with some variation within the classes.

The type A can be further classified in sub types AL , AP and AN, where the guest

solubility of first type increases linearly with CD concentration while those of second and

third type deviate positively and negatively, respectively from the straight line. The

complex formation with a 1:1 stoichiometry gives the AL type diagram where as the

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 45 
higher order complex formation in which more than one CD molecules are involved in

the complexation gives the AN-type is complicated, because of a significant contribution

of solute-solvent interaction of the complexation. In the case of the Bs type, the initial

ascending portion of the solubility change is followed by a plateau region and then a

decrease in the solubility at higher CD concentration accompanying a microcrystalline

precipitation of the complex. The HP-type diagram is indicative of the formation of

insoluble complexes in water.

The stability constant (Ks) and stoichiometry of complexes are determined by

analyzing quantitatively the phase solubility diagram.

Detection of inclusion complexation in the solid state

Detection of the inclusion complexation in solid state can be done by powder X-ray diffractometry, single

crystal X-ray structure analysis, thermo analytical, thin layer chromatography, Paper chromatography, IR spectroscopy,

Scanning electron microscopy and Dissolution study methods40.

CDs, as a result of their complexation ability and other versatile characteristics, are continuing to have

different applications in different areas of drug delivery and pharmaceutical industry.

However, it is necessary to find out any possible interaction between these agents and other formulation

additives because the interaction can adversely affect the performance of both.

It is also important to have knowledge of different factors that can influence

complex formation in order to prepare economically drug/CD complexes with desirable

properties.

Since CDs continue to find several novel applications in drug delivery, we may

expect these polymers to solve many problems associated with the delivery of different

novel drugs through different delivery routes.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 46 
Objectives of the study:

• To perform preformulation studies like, solubility of a drug in water, Blend of water

glycerin and water SLS etc.

• To carry out the Phase solubility study of Gliclazide with HP-β-CD.

• To formulate the inclusion complexes of Gliclazide with HP-β-CD in a different ratios.

• To formulate the inclusion complexes by different methods,(Physical and Kneading

methods).

• To perform compatibility studies using FTIR, DSC and XRD.

• To perform the characterization studies like Drug content, in - Vitro Dissolution studies

and Dissolution parameters of inclusion Complexes.

• To perform the characterization studies of Tablets like Weight variation, Friability test,

Hardness test and Disintegration time.

• To ascertain the release mechanism and kinetics of drug release from tablets.

• To perform the stability studies of Tablets as per ICH guidelines.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 47 
REVIEW OF LITERATURE

PROFILE OF DRUG AND EXCIPIENT

Drug Profile:

GLICLAZIDE:

Gliclazide is a second generation sulfonylurea oral hypoglycemic agent, widely used for

the treatment of non insulin dependent diabetes mellitus.

Definition: Gliclazide contains not less than 99.0 per cent and not more than the

Equivalent of 101.0 per cent of 1-(hexahydrocyclopenta[c]pyrrol-2(1H)-yl)-3-[(4-

methylphenyl)sulphonyl]urea, calculated with reference to the dried substance.

Chemical Name : 1-(3-azabicyclo [3.3.0] oct-3-yl)-3-p-tolylsulphonylurea.

Molecular Formula : C15H21N3O3S

Molecular Weight : 323.42

Melting point : 180 - 182oC

Structural Formula:

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 48 
Gliclazide

Action and use: Inhibition of ATP-dependent potassium channels (sulfonylurea);

treatment of diabetes mellitus.

Characters

Appearance : White or almost white powder.

Solubility : Practically insoluble in water, freely soluble in methylene

Chloride, sparingly soluble in acetone, slightly soluble in ethanol

(96 per cent).

Assay : Dissolve 0.250 g in 50 ml of anhydrous acetic acid R.

Titrate with 0.1 M perchloric acid, determining the end-

Point potentiometrically.1ml of 0.1 M perchloric acid is

equivalent

To 32.34 mg of C15H21N3O3S.

Storage : In an airtight container, protected from light.

Pharmacokinetics

Absorption, Fate, and Excretion.

The sulfonylurea has similar spectra of activities; thus, their pharmacokinetic

properties are their most distinctive characteristics. Although there are differences in the

rates of absorption of the different sulfonylureas, all are effectively absorbed from the

gastrointestinal tract. However, food and hyperglycemia can reduce the absorption of

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 49 
sulfonylureas. (Hyperglycemia per se inhibits gastric and intestinal motility and thus can

retard the absorption of many drugs.) In view of the time required to reach an optimal

concentration in plasma, sulfonylureas with short half lives may be more effective when

given 30 minutes before eating. Sulfonylureas in plasma are largely (90 to 99%) bound to

protein, especially albumin. The volumes of distribution of most of the sulfonylureas are

about 0.2 liter/kg.

The first generation sulfonylureas vary considerably in their half lives and extents

of metabolism. The second generation agents are approximately 100 times more potent

than are those in the first group. Although their half lives are short (3 to 5 hours), their

hypoglycemic effects are evident for 12 to 24 hours, and it is often possible to administer

them once daily. The reason for the discrepancy between the half life and duration of

action of these drugs is not clear.

All of the solfonylureas are metabolized by the liver, and the metabolites are

excreted in the urine. The solfonyluraes should be administered with caution to patient

with either renal or hepatic insufficiency.

Dose: For treating type 2 diabetes:

Adults- 80 milligrams (mg) a day with a meal as a single dose or 160 to 320 mg

divided into two doses taken with the morning and evening meals.

Children- The type of diabetes treated with this medicine is rare in children. However, if

child needs this medicine, the dose would have to be determined by the doctor.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 50 
Mechanism of action

Sulfonylureas lower blood glucose in patients with type 2 diabetes by directly

stimulating the acute release of insulin from functioning beta cells of pancreatic islet

tissue by an unknown process that involves a sulfonylurea receptor on the beta cell.

Sulfonylureas inhibit the ATP-potassium channels on the beta cell membrane and

potassium efflux, which results in depolarization and calcium influx, calcium-calmodulin

binding, kinase activation, and release of insulin-containing granules by exocytosis, an

effect similar to that of glucose. Insulin is a hormone that lowers blood glucose and

controls the storage and metabolism of carbohydrates, proteins, and fats. Sulfonylureas

are effective only in patients whose pancreas is capable of producing insulin.

With chronic sulfonylurea treatment, insulin production is not increased and may

return to pretreatment values, but insulin efficacy continues and is thought to involve

extrapancreatic mechanisms to increase insulin sensitivity in target tissues, such as liver,

muscle, and fat as well as in other cells, such as monocytes and erythrocytes. This can

result in a decrease in hepatic glycogenolysis and gluconeogenesis. It is unclear if the

sulfonylurea's extrapancreatic actions that increase insulin's efficacy are direct or indirect

effects, but it is clear that the mechanism of action is not due to a direct sulfonylurea

action on the insulin receptor. Because this peripheral effect is not apparent in patients

with type 1 diabetes, the evidence suggests that this may not be the clinically significant

mechanism of sulfonylurea action in patients with type 2 diabetes either. However, it is

clear that tissues of sulfonylurea-treated patients with type 2 diabetes become more

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 51 
responsive to lower concentrations of endogenous insulin. Primary failure of sulfonylurea

therapy may occur if beta-cell function is severely impaired. In addition to stimulating

insulin secretion through the beta cell–sulfonylurea receptor, gliclazide may have a direct

effect on intracellular calcium transport that specifically improves the biphasic response

of the beta cell to a meal, that is, the immediate first phase of insulin release.

Adverse Reactions

Sulfonylureas may cause hypoglycemic reactions, including coma. This is particular

problem in elderly patients with impaired hepatic or renal function who is taking longer

acting sulfonylurea.

Other side effects of sulfonylureas include nausea and vomiting, cholestatic jaundice,

agranulocytosis, aplastic and hemolytic anemia, generalized hypersensitivity reactions,

and dermatological reactions.

Therapeutic uses.

Sulfonylureas are used to control hyperglycemia in type 2 diabetes mellitus patients who

cannot achieve appropriate control with changes in diet alone. Patients with type 2 DM

whose disease is controlled with relatively low doses of insulin (less than 40 U per day)

are more likely to respond to sulfonylureas.

Contraindications to the use of these drugs include type 1 DM, pregnancy, lactation, and

significant hepatic or renal insufficiency.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 52 
Excipients Profile:

Hydroxypropyl β-Cyclodextrin

Synonyms: 2-hydroxypropyl β-Cyclodextrin.

Functional category: Solubilizing and stabilizing agents.

Molecular weight: 1548

Structural formula:

Where, R= CH2CH (OH) CH3 or H

Description: White crystalline powder.

Solubility: Greater than 1 in 2 parts of water at 25°C.

Stability and storage conditions: Are stable in the solid state if protected from high humidity. It should be stored in a tightly

sealed container, in a cool and dry place.

Incompatibilities: The activity of some antimicrobial preservatives in aqueous solution

can be reduced in the presence of hydroxypropyl β-Cyclodextrin.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 53 
¾ Patel VP, Patel NM, Chaudhary BG investigated the effect of complexation of
glipizide, a practically insoluble antidiabetic drug, with cyclodextrin in presence

of different concentration of water soluble polymers (HPMC,PVP,PEG 4000 and

PEG 6000) on the dissolution rate of the drug. The dissolution rate of the drug

from ternary systems containing PEG 4000 or PEG 6000 seems to be generally

higher than from systems containing HPMC or PVP. The dissolution rate of the

drug from the ternary systems glipizide-HP-β-cyclodextrin-5% PEG 4000 was

high compare to the other systems193.

¾ Doijad RC, Kanakal MM and Manvi FV investigated influence of processing


variables on the solid-state of a model drug, piroxicam in cyclodextrin-based

system and its effect on dissolution behaviour of drug. The complex prepared by

co-evaporation method with hydroxy propyl β-cyclodextrin and was found to

yield better dissolution rate and stability194.

¾ Seoung Wook Jun, Min-Soo Kim prepared the inclusion complexes of insoluble
drug simvaststin (SV),with HP-β-cyclodextrin using a supercritical antisolvent

(SAS) process he complexes were investigated to improve the aqueous solubility

and the dissolution rate of drugs .Inclusion complexation in aqueous solution and

solid state was evaluated by the phase solubility diagram, DSC, PXRD, FTIR and

SEM. FTIR study demonstrated that the presence of intermolecular hydrogen

bonds between SV and HP-β-cyclodextrin in inclusion complex, resulting in the

formation of amorphous form. Aqueous solubility and dissolution studies

indicated that the dissolution rates were remarkably increased inclusion complex,

compared with the physical mixture and pure drug195.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 54 
¾ Deshmukh SS, Potnis VV, Shelar DB made an attempt to prepare and
characterized inclusion complexes of ziprasidone hcl with β-cyclodextrin and HP-

β-cyclodextrin. The phase solubility analysis indicated that the formation of 1:1

molar inclusion complex of a drug with β-cyclodextrin and HP-β-cyclodextrin.

The inclusion complexes were prepared co-precipitation, kneading and

microwave method. The prepared complexes were characterized using solubility

study, DSC and XRD. The inclusion complexes prepared with HP-β-

cyclodextrins by microwave method exhibited greatest enhancement in solubility

and fastest dissolution of ziprasidone HCl196.

¾ Tayade PT, Vavia PR prepared ketoprofen cyclodextrins solid systems by


kneading, co-evaporation and freeze drying. The formation of inclusion

complexes with β-cyclodextrins and HP-β-cyclodextrins in the solid state, were

confirmed by DSC, FT-IR, powder X-ray diffractometry, SEM studies and

comparative studies on the in-vitro dissolution and in-vivo absorption of

ketoprofen in human volunteers were carried out .the dissolution rate of

ketoprofen in the inclusion complexes was 15 fold higher, than that of plain drug

powder197.

¾ Chowdary KPR and Vijay Srinivas studied the effect of polyvinyl pyrrolidone on

complexation and dissolution rate of β-and hydroxypropyl β-cyclodextrin

complexes of celecoxib and were markedly enhanced the solubility and

dissolution rate by complexation with β-cyclodextrin and hydroxypropyl β-

cyclodextrin. Solid inclusion complexes of cyclodextrin with polyvinyl

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 55 
pyrrolidone gave several times higher rates of dissolution than those of celecoxib

and its complexes with cysclodextrin alone.198

¾ Deelip Derle, Sai Hanuman Sagar Boddu and Manoj Magar prepared complexes

of satranidazole inclusion complexes with β -CD. The solid complex prepared by

kneading method was characterized using differential scanning calorimetry,

powder X-ray diffractometry. In vitro study showed that the solubility and

dissolution rate of satranidazole was significantly improved with β-CD.199

¾ Monique WJ den Brok et al. studied the potency of the commercially available
and used cyclodextrins, 2-HP-β-cyclodextrin to extract the preservative 2-Phenyl

Phenol (2PP) from platinum cured silicon tubing the presence of 2PP was

structurally confirmed with HPLC-UV and LC/MS/MS in HP-β-cyclodextrins

solutions after incubation with platinum cured silicone tubing. HP-β-cyclodextrins

concentration and prior tubing sterilization were found not to influence the levels

of 2PP 200.

¾ Nagesh Bandi and Uday B Kompella had determined whether budesonide and

indomethacin HPβ-CD complexes could be formed using a supercritical fluid

process. The process involved the exposure of drug-HPβ-CD mixtures to

supercritical carbon dioxide. The ability of supercritical fluid process to form

complexes was assessed by determining drug dissolution, drug crystallinity and

drug excipients interactions. Thus budesonide and indomethacin HPβ-CD

complexes with enhanced dissolution can be formed using a single step organic

solvent free SCF process.201

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 56 
¾ Sunil Jambhekar, Casella R and Maher T made an effort to improve the
bioavailability of the insoluble drug indomethacin. Three complexes were

prepared with indomethacin and the soluble complexing agents (β-, hydroxyethyl-

β- and hydroxyl propyl-β-cyclodextrin). Phase solubility analysis confirmed the

formation of complexes and suggested the three complexes were found similarly.

Solubility studies show complexation increase indomethacin solubility. The

bioavailability studies were performed by administering the indomethacin

complex or Indocin capsule to male-ablino, New Zealand rabbits. Hence, it was

observed that the bioavailability of indomethacin was improved by complexation

with only β-cyclodextrins


¾
Indap MA et al. have prepared inclusion complexes of quercetin with

hydroxypropyl β-cyclodextrin in 1:5 molar ratios. The maximum tolerated dose

corresponding to the Ld10 was >400 mg/Kg of hydroxypropylβcyclodextrins

complex of quercetin obtained after single intraperitoneal application which

proved to be less toxic than quercetin. There was no apparent toxicity to bone

marrow of irradiated Swiss mice. Previously administered HPBC complex of

quercetrin. The ability to selectively target quercetin via its cyclodextrins

inclusion complex against cancer growth could improve the therapeutic

effectiveness of cyclodextrins preparation as well as reduce adverse effect

associated with quercetin.203


¾
Mourya VK, Saini TR et al. have prepared molecular inclusion complex of

hydroxypropyl β-cyclodextrin with Sparfloxacin. The 1:1 stiochemistry of the

components is suggested by phase solubility studies and Benesi-Hildebrand Plot.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 57 
Stability constant evaluated by phase solubility analysis and Benesi-Hildebrand

plot showed deviations. The complex displayed enhanced aqueous solubility and

dissolution.204

¾ Tayade NG and Nagarsenker MS have prepared inclusion complexes of

susquiterpene lactone with hydroxypropyl β-cyclodextrin in 1:1 ratio, the drug in

cyclodextrin when administered orally to the mice infected with plasmodium

yoelli, improved antimalarial activity in terms of increase mean survival time

(MST) and percent inhibition in parasitaemia was observed suggesting in-vivo

advantage.205

¾ Raida S. Al-Kassas et.al. This work investigates preparation of

biodegradable beads with alginate polymer by ionotropic gelation method to

take the advantages of the swelling and mucoadhesive properties of alginate

beads for improving the oral delivery of the antidiabetic agent gliclazide. It

demonstrates that the ionic gelation of alginate molecules offers a flexible

and easily controllable process for manipulating the characteristics of the

beads which are important in controlling the release rate and consequently

the absorption of gliclazide from the gastrointestinal tract. The in vitro

release experiments revealed that the swelling is the main parameter

controlling the release rate of gliclazide from the beads. In vivo studies on

diabetic rabbits showed that the hypoglycemic effect induced by the

gliclazide loaded alginate beads was significantly greater and more

prolonged than that induced by the marketed conventional gliclazide tablet.

The results clearly demonstrated the ability of the system to maintain tight

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 58 
blood glucose level and improved the patient compliance by enhancing,

controlling and prolonging the systemic absorption of gliclazide.206

¾ S Biswal et.al. The present study was to characterize gliclazide solid

dispersions (SDs) prepared with polyethylene glycol (PEG) 8000 and

compare them with SDs in PEG 6000. Gliclazide SDS containing varying

concentrations of PEG 8000 was prepared using the fusion – solvent

technique, and their phase solubility behavior and dissolution in 0.1N HCl

were assessed at 37 oC. The solubility of gliclazide increased with

increasing amount of PEG 8000 in aqueous medium. Gibbs free energy

values were all negative, indicating the spontaneous nature of Gliclazide

solubilisation. Dissolution studies indicated a significant increase in the

dissolution of gliclazide when dispersed in PEG 8000. FTIR analysis

demonstrated the absence of well-defined gliclazide - PEG 8000 chemical

interaction while DSC and XRD studies indicated the amorphous

microcrystalline state of gliclazide in the SDs207.

¾ Hussien Allaboun et. al. studied to improve dissolution rate of gliclazide

using anionic and cationic surfactants. The intrinsic dissolution rates of

gliclazide in solutions of sodium dodecyl sulfate (SDS) and in solutions of

tetradecyltrimethyl ammonium bromide (TDTMAB) were measured using

the rotating disk method to study the convective diffusion transport of drug-

loaded micelles. Two different approaches were applied to the experimental

data; the convective diffusion model and the film equilibrium model. The

two approaches are based on the same fundamental assumptions differing

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 59 
only in their interpretation of the diffusional boundary layer. The results

obtained from the film equilibrium model were less satisfactory, and in case

of TDTMAB the model was inapplicable (negative diffusion coefficient).

While excellent results were obtained from the convective diffusion 252

model.208
¾
Roya Talari et.al. The present study is to use in situ micronization process

through pH change method to produce micron-size gliclazide particles for

fast dissolution hence better bioavailability. Gliclazide was recrystallized in

presence of 12 different stabilizers and the effects of each stabilizer on

micromeritic behaviors, morphology of microcrystals, dissolution rate and

solid state of recrystallized drug particles were investigated. The results

showed that recrystallized samples showed faster dissolution rate than

untreated gliclazide particles and the fastest dissolution rate was observed

for the samples recrystallized in presence of PEG 1500. Some of the

recrystallized drug samples in presence of stabilizers dissolved 100% within

the first 5 min showing at least 10 times greater dissolution rate than the

dissolution rate of untreated gliclazide powders.209

¾ Gopal Venktesh Shavi et.al. Study was aimed to formulate solid dispersions

using different water soluble polymers such as polyethylene glycol 4000

(PEG 4000), polyethylene glycol 6000 (PEG 6000) using fusion method and

polyvinyl pyrrolidone K- 30 (PVP K 30) by solvent evaporation method.

The interaction of gliclazide with the hydrophilic polymers was studied by

Differential Scanning Calorimetry (DSC). Pharmacokinetic studies of

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 60 
optimized formulation were compared with pure drug and marketed

formulation in wistar rats. It was observed that there is a twofold increase in

peak plasma concentration compared to pure drug.210

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 61 
METHDOLOGY

The solubility, stability and therapeutic efficacy of drugs are closely related to

their physicochemical properties and those of vehicles or carriers used along them. The

dissolution rate of drug from its dosage form is considered as an important parameter in

bioavailability, dissolution is the rate limiting step in the absorption of poorly water

soluble drugs from its dosage forms.

Various intermolecular forces such as Vander Waals forces, ion-dipole and non-

induced dipole forces, hydrogen bonding, crystal lattice interactions etc. between the drug

and vehicle, affect the drug solubility and dissolution rate. By selecting suitable

excipients in proper proportion, drugs stability and desired dissolution rate can be

achieved. The dissolution rate of drug from its dosage form is considered as an important

parameter in bioavailability.

Cyclodextrins (CDS) are cyclic oligosaccharides contains six (α-cyclodextrin)

seven (β-cyclodextrin) or eight (γ-cyclodextrin) α-1, 4 linked glucopyranose units with

hydrophilic outer surface and a hydrophobic cavity . CDS are capable of forming

inclusion complexes with many drugs by taking up a whole drug molecule or part of it,

into the CDs cavity. Such molecular encapsulation will affect many of the

physicochemical properties of drugs such an aqueous solubility and rate of dissolution.

Among the various approaches, preparation of inclusion complexes with cyclodextrin has

proven to be successful in enhancing the solubility of poorly water soluble drugs.

Gliclazide is second generation hypoglycemic sulfonylurea which is useful in the

treatment of type 2 diabetes mellitus and has its poor aqueous solubility5. This limits

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 62 
several advantages of the drug with respect to its absorption, distribution and therapeutic

efficacy. Hence in the present work cyclodextrin inclusion complexes of Gliclazide will

be prepared to enhance its solubility and dissolution rate

• To perform preformulation studies like, solubility of a drug in water, Blend of water

glycerin and water SLS etc.

• To carry out the Phase solubility study of Gliclazide with HP-β-CD.

• To formulate the inclusion complexes of Gliclazide with HP-β-CD in a different ratios.

• To formulate the inclusion complexes by different methods,(Physical and Kneading

methods).

• To perform compatibility studies using FTIR, DSC and XRD.

• To perform the characterization studies like Drug content, in - Vitro Dissolution studies

and Dissolution parameters of inclusion Complexes.

• To perform the characterization studies of Tablets like Weight variation, Friability test,

Hardness test and Disintegration time.

• To ascertain the release mechanism and kinetics of drug release from tablets.

• To perform the stability studies of Tablets as per ICH guidelines.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 63 
A) Materials
Table No.7: Materials and their Sources

Materials Sources

Gliclazide Wallace Pharmaceuticals Pvt.Ltd, Goa.

Hydroxypropyl β-Cyclodextrin Gangwal Chemicals Pvt. Ltd., Mumbai

Potato starch Gangwal Chemicals Pvt. Ltd., Mumbai

Microcrystalline cellulose S.D. Fine chem.

Lactose S.D. Fine chem.

Talc S.D. Fine chem.

Magnesium stearate S.D. Fine chem.

* All the materials were used as procured; without further purification.

* All the other solvents and reagents used for the study were of Analytical Reagent grade.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 64 
Table No. 8: Equipments and their sources

Equipments Sources

Dissolution test apparatus Electrolab, USP XXIV

UV Visible spectrophotometer U.V. – 1800, Shimadzu

FTIR Spectrophotometer 8400s, Shimadzu

Differential Scanning Calorimeter Mettler Toledo

Hot air Oven M.C. Dalal and Co., Chennai

Digital Balance Citizen

Gyratory Flask Shaker Remi Electrical

Sieve 100 mesh Indicot India

Glassware Borosil (A) – grade

Tablet Punching Machine (Pilot Press


Chamuda Pharma, Ahemadabad
10 Station)

Stability Chamber Osworld, IRIC-11

Friability Tester Electrolab, USP EF-2

Hardness Tester Monsanto

Digital pH meter Italy(H96107)

Vernier caliper Pico India Ltd.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 65 
Analytical method and its Validation.

Methods for the estimation of Gliclazide:

A spectrophotometric method based on the measurement of absorbance at

227nm in phosphate buffer of pH 7.4 was used in the present study for the estimation of

gliclazide.

Reagents:

Phosphate buffer of pH 7.4:

Phosphate buffer of pH 7.4 was made by dissolving 6.8gm of potassium

dihydrogen ortho phosphate and 1.5gm of sodium hydroxide in 1000ml with distilled

water. The pH was adjusted to 7.4.

Standard solution:

10mg of gliclazide was dissolved in methanol in 25ml volumetric flask and the

solution was made up to the volume with methanol.

Procedure:

The standard solution of gliclazide was subsequently diluted with phosphate buffer

of pH 7.4 to obtain series of dilutions containing, 10,20,30,40, and 50µg of gliclazide per

ml of solution. The absorbance of the above dilutions was measured in Elico-UV-Visible

spectrophotometer at 227nm using phosphate buffer of pH 7.4 as blank. The

concentration of Glicazide and corresponding absorbance were given in Table 10. The

absorbance values were plotted against concentration of Glicazide as show in Figure 5.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 66 
Validation:

The method was validated for linearity, precision and accuracy. The method

obeyed Beer’s law in the concentration range of 0-50µg/ml, when standard drug solution

was assayed repeatedly (n=6), the mean error (accuracy) and relative standard deviation

(precision) were found to be 0.7 and 0.9% respectively. No interference from excipients

used was observed. Thus method was found suitable for the estimation of gliclazide

content in dissolution fluids. Calibration curve shown in fig: was used for the purpose.

Melting Point:

The melting point of gliclazide was determined by conventional melting point apparatus.

Solubility of Gliclazide.

Solubility of gliclazide in water, the effect of pH (4, 6, 7.4,8 and9) and blend of

water with cosolvents (alcohol, glycerin) and surfactant (SLS) in different concentrations

(0.5%, 1%, 2%, 3%, 4% and 5%) the aqueous solubility of gliclazide was determined.

10mg amount of gliclazide was added to 10ml of distilled water, different pH and blend

of water with cosolvents and solvent respectively in a stoppered conical flasks. The

mixtures were frequently shaken for 1 hour and filtered using wattman filter paper. The

aliquots were diluted suitably and assayed for gliclazide by UV spectrophotometer

(SHIMATZU 1700) at 227 nm. against blanks prepared in the same concentration of

water, pH solutions and different concentrations of co-solvents and solvents respectively

so as to cancel any absorbance that may be exhibited by water, pH and a blend of co-

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 67 
solvents, solvents. The solubility experiments were conducted in triplicate and calculated

mg/ml.

Phase solubility studies.

The phase-solubility technique permits the evaluation of the affinity between HP-β

CD and gliclazide in water. Phase solubility studies were performed according to the

method reported by Higuchi and Connors192. Gliclazide, in amounts that exceeded its

solubility, was taken in to 25ml stoppered conical flasks to which were added 15 ml of

distilled water containing various concentrations of HP-β-cyclodextrin (1-15 mM). These

stoppered conical flasks were shaken for 48 hours at room temperature on a rotary

shaker. This amount of time is considered sufficient to reach equilibrium. Subsequently,

the aliquots were withdrawn (2 ml), using a syringe at 12 hours intervals, and samples

were filtered immediately by using wattman filter paper and diluted suitably. A portion of

a sample was analyzed by UV spectrophotometer (SHIMATZU 1700) at 227 nm against

blanks prepared in the same concentration of HP-β-cyclodextrin in water so as to cancel

any absorbance that may be exhibited by the HP-β-CD. Shaking was continued until 3

consecutive estimations were equivalent. The solubility experiment was conducted in

triplicate. The apparent stability constant (Kc) according the hypothesis of 1:1

stoichiometric ratio of complexes was calculated from the phase-solubility diagrams

using the following equation1.

K1:1 = Slope / So (1- Slope)

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 68 
The slope is obtained from the initial straight-line portion of the plot of gliclazide

concentration against HP-β-CD concentration, and So is equilibrium solubility of

gliclazide in water.

Thin layer chromatography (T L C).

The stability study of the drug in the formulation was confirmed by thin layer

chromatography analysis. The TLC studies were carried out on Aluminium preparative

sheets using Methanol: Trimethylamine: water: Acetronitrile (20:65.9:15) as developing

solvent (mobile phase) and dil, sulphuric acid as spraying solution. The standard TLC

plates are used. The plates were spotted with pure gliclazide in methanol at about 2 cm

above from the bottom as standard. The sample (Gliclazide +HP-β-cyclodextrin) in

methanol were spotted adjacent to the pure gliclazide spot at a distance of 2 cm, the plate

was kept in an enclosed chamber saturated previously with mobile phase solvent system.

The solvent was allowed to rise on the plate to a sufficient level (⅔). The distance

travelled by solvent front was noted. The distance travelled by the solute was determined.

The Rf value was calculated for both the standard and sample, using the following

formula.

Rf Value = Distance travelled by solute / Distance travelled by mobile

solvent.

Rf value of Gliclazide= 0.781

Rf value of Gliclazide-HPβ-CD=0.842

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 69 
IR Spectral Analysis:

The compatibility between drug and the formulation components were confirmed by

IR Spectral studies. The IR Spectra were recorded using mulling agent KBr pellets. The

IR spectrum of gliclazide and gliclazide-HP-β-CD is shown in Fig.7&8. The IR

spectroscopy was conducted using a Shimadzu FTIR 8400 spectrophotometer and the

spectrum was recorded in the wavelength region of 4000 to 400 cm-1. The procedure

consists of dispersing a sample (pure drug alone and drug-CD in different ratio of mM

complexes) in KBR and compressing in to disc / film by applying a pressure of 8 Tunes

for 5 min hydraulic press. The pellet was placed in the light and the spectrum was

obtained.

Differential Scanning Calorimetry (DSC):

DSC thermograms of the gliclazide, HP-β-cyclodextrin and solid complex

prepared by kneading method and physical mixtures were recorded on NETZSCH DSC

204 METTLER STARe Model. Samples (2-5 mg) were sealed in to aluminum pans and

scanned at a heating rate of 100c min-1 over a temperature range 30-3000c under a

nitrogen gas stream. The DSC thermograms are shown in fig.9&10.

X-ray Diffractometry:

X-ray powder diffraction patterns were recorded using a Phillips model powder

diffractometer with monochromatized Cu-Kα radiation. The solid inclusion complexes of

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 70 
gliclazide HP-β-CD prepared by kneading method and physical mixtures were scanned at

room temperature in the continuous scan mode over the 50-500 2θ range with 0.1 2θ step

size and with counting time of 0.6 sec. The X-ray diffractograms are shown in

figs.11&12.

Preparation of Gliclazide-HP-β-CD Complex.

Kneading Method:

HP-β-cyclodextrin (1 mM) and distilled water (1.5 ml) were mixed together in a

mortar so as to obtain a homogeneous paste. Gliclazide (2 mM) was then added slowly.

The mixture was then kneaded for 45 min. During this process, an appropriate quantity of

water was added to the mixture in order to maintain a suitable consistency. The paste was

then dried in oven at 700C for 1-2 hours. The dried complex was milled and passed

through sieve no. 100. The same procedure was used to prepare a formulation of

gliclazide-HP-β-CD complexes in different mM ratios.

Physical mixture:

Gliclazide with HP-β-CD in different molar ratios (1:2M) were mixed in a mortar

for about one hour with constant trituration, passed through sieve No. 80 and stored in

desiccators over fused calcium chloride. The same procedure was used to prepare a

formulation of gliclazide-HP-β-CD complexes in different mM ratios.

Estimation of Drug Content.

The gliclazide content in the inclusion complex was estimated by UV

spectrophotometric (SHIMADZU 1700) method. The method was validated for linearity,

accuracy and precision. A sample of gliclazide-HP-β-CD complex equivalent to 100 mg

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 71 
was dissolved with 25 ml methanol in a 100 ml volumetric flask, and the volume was

adjusted up to 100ml by using phosphate buffer of pH 7.4. The solution was filtered

through Wattman filter paper. The filtrate was assayed for drug content by measuring the

absorbance at 227 nm after suitable dilution, against phosphate buffer of pH 7.4 as blank.

Then the amount of gliclazide in the complex was calculated as % yield (% yield =

practical yield / theoretical yield x 100), mean drug content and weight equivalent to 100

mg of drug is estimated. The same procedure was used to estimate drug content of

gliclazide-HP-β-CD complexes in different mM ratios.

IN-VITRO Drug release from inclusion complex.

Drug release was studied by using USP-Thermo Lab eight station dissolution test

apparatus in phosphate buffer solution of pH 7.4. The test sample of drug-HP-β-CD

complex (100 mg equivalent to pure drug in complex) was tightened in a muslin bag with

a paddle stirrer, which rotates at a speed of 50 rpm, the temperature of water bath, was

maintained 370C ± 0.50C throughout the experiment. 5 ml aliquot of dissolution medium

was withdrawn at a specific time intervals and replaced with 5 ml fresh medium of

phosphate buffer solution of pH 7.4 to maintain sink condition. The withdrawn sample

was filtered through wattman filter paper, after suitable dilutions with phosphate buffer

solution of pH 7.4 the absorbance was determined by using UV spectrophotometer

(SHIMADZU 1700) at 227 nm against blank (phosphate buffer solution of pH 7.4). The

absorbance was recorded. The release of pure drug was also studied to compare with

release of drug from complexes. All studies were conducted in triplicate; the average data

obtained were computed for further calculations.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 72 
Formulation of Gliclazide-HP-β-CD tablets.

Gliclazide-HP-β-CD Tablets were prepared by two methods, wet granulation and

direct compression. In wet granulation method, the weighed quantities of gliclazide, HP-

β-CD ,MCC and ½ portion of potato starch (dry) were mixed in geometric ratios in

mortar, to which alcohol : water (1:1 ratio)containing 0.5% PVP were added and mixed

to get a lumps, the lumps were passed through sieve no 12, to get granules. These

granules were dried at 600c for 2-4 hours in a oven until they dry. To the dried granules,

remaining ½ portion of potato starch (dry), talc and magnesium stearate were added and

passed through sieve no 14. These granules were punched in to tablets (8 mm punch) by

using Rimek mini press 10 station rotary tablet punching machine. In direct compression

method, drug and all other ingredients were mixed in geometric ratio in mortar, and

directly compressed to get desired tablets.

Table No. 9. Formulae of Tablets.


Gliclazide : 60mg
HP β-CD : 120mg
MCC : 60mg
Potato Starch (Dry) : 30mg
Talc : 15mg
Mag, Stearate : 15mg

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 73 
Estimation of drug content from tablets.

The gliclazide content in the tablets were estimated by UV spectrophotometric

(SHIMADZU 1700) method. The method was validated for linearity, accuracy and

precision. Five gliclazide-HP-β-CD tablets (wet granulation) weighed and were crushed

in to powder. The Tablet powder equivalent to 100mg of gliclazide was dissolved with 25

ml methanol in 100 ml volumetric flask, and the volume was adjusted up to 100ml by

using phosphate buffer of pH 7.4. The solution was frequently shaken for 1hr, and

filtered through Wattman filter paper. Then, the filtrate was assayed for drug content by

measuring the absorbance at 227 nm after suitable dilutions, against phosphate buffer of

pH 7.4 as blank. The amount of gliclazide in the tablet was calculated as % yield (%

yield = practical yield / theoretical yield x 100), mean drug content and weight equivalent

to 100 mg of drug is estimated. The same procedure was used to estimate drug content of

gliclazide-HP-β-CD tablets prepared by direct compression method.

IN-VITRO Drug release from Gliclazide-HP-β-CD tablets.

Drug release was studied by using USP-24 Electrolab eight station dissolution test

apparatus in phosphate buffer solution of pH 7.4. A tablet of gliclazide-HP-β-CD

(formulated by wet granulation method) was placed in a basket, which rotates at a speed

of 50 rpm; the temperature of water bath was maintained 370C ± 0.50C throughout the

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 74 
experiment. A 5 ml aliquot of dissolution medium was withdrawn at a specific time

intervals and replaced with 5 ml fresh medium of phosphate buffer solution of pH 7.4 to

maintain sink condition. The withdrawn sample was filtered through wattman filter

paper, after suitable dilutions with phosphate buffer solution of pH 7.4 the absorbance

was determined by using UV spectrophotometer (SHIMADZU 1700) at 227 nm against

blank (phosphate buffer solution of pH 7.4). The absorbance was recorded. Same

procedure was used to estimate the drug content in gliclazide-HP-β-CD tablet formulated

by direct compression method. All studies were conducted in triplicate; the average data

obtained were computed for further calculation ns.

Evaluation of Gliclazide-HP-β-CD tablets.

Weight variation of tablets was determined by weighing 20 tablets individually in

a electronic weighing machine, calculating the average weight, and comparing the

individual tablets weights to the average.

Hardness of tablets was tested by using Monsanto hardness tester.

Friability of tablets was determined in Roche Friabilator (Electrolab). Preweighed

tablets were placed in a friabilator, which is then operated for 100 revolutions. The tablets

were dusted and reweighed.

Disintegration of the tablets was determined by using phosphate buffer solution of

pH 7.4 in Disintegration test machine.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 75 
Stability studies of Gliclazide-HP-β-CD tablets.

Stability studies of the tablets are carried out according to ICH guidelines. In the

present study, as the tablets developed are solid dosage forms, a storage condition of 400c

± 20c; 75% RH ± 5% RH for 3 months was used for accelerated testing.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 76 
RESULTS

Concentration
Sl No (mcg/ml) Absorbance
1
0 0
2
10 0.13
3
20 0.231
4
30 0.324
5
40 0.436
6
50 0.558

Table No.10. Calibration curve data of Gliclazide in phosphate buffer pH 7.4

Standard Calibration Curve

0.6

0.5
Absorbance

0.4

0.3
0.2

0.1

0
0 10 20 30 40 50 60
Concentration (mcg/ml)

Figure No. 5. Standard Calibration Curve for Gliclazide in phosphate buffer pH 7.4

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 77 
Fluid Solubility(mg/ml)

Conc.(%) of added solvent/surfactant

0 0.5 %(SD) 1.0 %(SD) 2.0 %(SD) 3.0 %(SD) 4.0 %(SD) 5.0 %(SD)

Water 0.086±0.005 - - - - - -

Water+Alcohol 0.123±0.003 0.197±0.005 0.318±0.003 0.533±0.004 0.780±0.006 0.998±0.003

Water+Glycerine - 0.114±0.006 0.158±0.005 0.232±0.004 0.302±0.005 0.321±0.061 0.412±0.005

Water+SLS 0.153±0.003 0.409±0.005 0.785±0.013 1.106±0.010 1.357±0.005 1.537±0.011

Table No.11. Solubility (mg/ml) of gliclazide in various fluids

pH 4 pH 6 pH 7.4 pH 8 pH 9

Solubility
3.168 6.174 6.058 0.097 0.096
(mg/ml)
Solubility
3.181 6.185 6.052 0.096 0.085
(mg/ml)
Solubility
3.168 6.181 6.054 0.011 0.087
(mg/ml)

3.172 6.8 6.055 0.101 0.089


Average

0.165 0.278 0.256 0.102 0.113


(±SD)

Table No. 12. Effect of pH on solubility of glicazide.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 78 
HP-B-CD Drug Dissolved
Sl No (Mm) (Mm)

1 0 0

2 1 0.03

3 3 0.08

4 6 0.14

5 9 0.23

6 12 0.32

7 15 0.41

Table No.13. Phase Solubility Study Profile of gliclazide with HP β-CD

Phase Solubility Study of Gliclazide-HP-B-CD

0.45
Conc of Gliclazide (Mm)

0.4
0.35
0.3
0.25
0.2
0.15
0.1
0.05
0
0 2 4 6 8 10 12 14 16
Conc of HP-B-CD (Mm)

Figure.No.6. Phase Solubility Study Profile of Gliclazide with HP β-CD

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 79 
Figure No.7. IR Profile of Gliclazide and its HP β-CD inclusion complexes

Infrared Spectra of inclusion complex


1st I.R. spectra is Glicazide drug, GZ
2nd I.R. spectra is HP-β-CD drug, (HP-β-CD)
3rd I.R. Spectra is Glicazide: HP-β-CD (1:0.5) G1
4th I.R. Spectra is Glicazide: HP-β-CD (1:1) G2
5th I.R spectra is Glicazide: HP-β-CD (1:1.5) G3
6th I.R. spectra is Glicazide: HP-β-CD (1:1.5) G4
7th I.R. spectra is Glicazide: HP-β-CD (1:2) G5

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 80 
Figure No.8. IR spectra of Gliclazide and its HP β-CD Physical Mixture.

1st I.R. spectra is Glicazide drug


2nd I.R. spectra is HP-β-CD drug
3rd I.R. Spectra is Glicazide: HP-β-CD (1:0.5) GP1
4th I.R. Spectra is Glicazide: HP-β-CD (1:1) GP2
5th I.R spectra is Glicazide: HP-β-CD (1:1.5) GP3
6th I.R. spectra is Glicazide: HP-β-CD (1:1.5) GP4
7th I.R. spectra is Glicazide: HP-β-CD (1:2) GP5

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 81 
Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 82 
Figure No.9. DSC Profile of Gliclazide and its HP β-CD Inclusion complexes

1st DSC spectra is Glicazide drug


2nd DSC spectra is HP-β-CD drug
3rd DSC Spectra is Glicazide: HP-β-CD (1:0.5) G1
4th DSC Spectra is Glicazide: HP-β-CD (1:1) G2
5th DSC spectra is Glicazide: HP-β-CD (1:1.5) G3
6th DSC spectra is Glicazide: HP-β-CD (1:1.5) G4
7th DSC spectra is Glicazide: HP-β-CD (1:2) G5

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 83 
Figure No.10. DSC Profile of Gliclazide and its HP β-CD Physical Mixture.

1st DSC spectra is Glicazide drug


2nd DSC spectra is HP-β-CD drug
3rd DSC Spectra is Glicazide: HP-β-CD (1:0.5) GP1
4th DSC Spectra is Glicazide: HP-β-CD (1:1) GP2
5th DSC spectra is Glicazide: HP-β-CD (1:1.5) GP3
6th DSC spectra is Glicazide: HP-β-CD (1:1.5) GP4
7th DSC spectra is Glicazide: HP-β-CD (1:2) GP5

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 84 
Figure No.11. XRD Profile of Gliclazide and its HP β-CD Inclusion complexes.

1st XRD spectra is Glicazide drug


2nd XRD spectra is HP-β-CD drug
3rd XRD Spectra is Glicazide: HP-β-CD (1:0.5) G1
4th XRD Spectra is Glicazide: HP-β-CD (1:1) G2
5th XRD spectra is Glicazide: HP-β-CD (1:1.5) G3
6th XRD spectra is Glicazide: HP-β-CD (1:1.5) G4
7th XRD spectra is Glicazide: HP-β-CD (1:2) G5

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 85 
Figure No.12. XRD Profile of Gliclazide and its HP β-CD Physical Mixture.

1st XRD spectra is Glicazide drug


2nd XRD spectra is HP-β-CD drug
3rd XRD Spectra is Glicazide: HP-β-CD (1:0.5) GP1
4th XRD Spectra is Glicazide: HP-β-CD (1:1) GP2
5th XRD spectra is Glicazide: HP-β-CD (1:1.5) GP3
6th XRD spectra is Glicazide: HP-β-CD (1:1.5) GP4
7th XRD spectra is Glicazide: HP-β-CD (1:2) GP5

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 86 
Sample. G1 G2 G3 G4 G5

1 95.13 95.18 96.41 97.38 97.07

2 94.86 93.78 95.79 96.81 98.15

3 96.04 96.26 96.19 97.03 98.43

AVG mg 95.34 95.07 96.13 97.07 97.88

std dev+- 0.62 1.24 0.31 0.29 0.72

c.v. (%) 0.006 0.013 0.0032 0.0029 0.00733

Table No.14. Drug Content Profile of Gliclazide HP-β-CD inclusion Complexes

G1 = Gliclazide: HP-β-CD (1:0.5)

G2 = Gliclazide: HP-β-CD (1:1)

G3 = Gliclazide: HP-β-CD (1:1.5)

G4 = Gliclazide: HP-β-CD (1:2)

G5 = Gliclazide: HP-β-CD (1:2.5)

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 87 
Sample. GP1 GP2 GP3 GP4 GP5
82.97
1 81.3 81.25 82.1 82.46

2 81.56 81.98 82.23 82.67 83.45

3 81.78 82.1 82.46 82.89 83.78

AVG mg 81.54 81.77 82.26 82.67 83.4

std dev+- 0.24 0.46 0.18 0.22 0.41

c.v. (%) 0.003 0.006 0.002 0.003 0.005

Table No.15. Drug Content Profile of Gliclazide HP-β-CD Physical Mixtures

GP1 = Gliclazide: HP-β-CD (1:0.5)

GP2 = Gliclazide: HP-β-CD (1:1)

GP3 = Gliclazide: HP-β-CD (1:1.5)

GP4 = Gliclazide: HP-β-CD (1:2)

GP5 = Gliclazide: HP-β-CD (1:2.5)

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 88 
Time (min)

Gliclzide Pure G 1(± Stdv) G 2 (± Stdv) G 3 (± Stdv) G 4 (± Stdv) G5 (± Stdv)

(± Stdv)

0 0 0 0 0 0 0

5 10.56 (±0.147) 20.34(±0.132) 27.74(±0.143) 33.29(±0.126) 37.60(±0.167) 40.64(±0.145)

10 12.71( ± .134) 60.55(±0.145) 66.01(±0.147) 70.49(±0.167) 77.58(±0.143) 79.18(±0.153)

20 17.37(±0.123) 71.71(±0.156) 76.41(±0.135) 82.89(±0.187) 85.06(±0.189) 86.21(±0.156)

30 22.64(±0.167) 79.03(±0.154) 84.58(±.0176) 86.21(±0.198) 89.84(±0.145) 89.05(±0.176)

40 24.96±(0.135) 84.11(±0.165) 87.51(±0.132) 89.29(±0.187) 91.92(±0.154) 91.97(±0.176)

50 27.19(±0.123) 86.19(±0.145) 88.36(±0.126) 90.37(±0.176) 92.15(±0.165) 93.21(±0.134)

60 37.19(±0.123) 96.19(±0.145) 98.36(±0.126) 98.37(±0.176) 98.15(±0.165) 98.21(±0.134)

Table No.16. Percentage of drug release Profile of Gliclazide inclusion complex with
HP-β-CD
(Kneading Method)

G 1 = Gliclazide-HP-β-CD Inclusion Complex

G 2 = Gliclazide-HP-β-CD Inclusion Complex

G 3 = Gliclazide-HP-β-CD Inclusion Complex

G 4 = Gliclazide-HP-β-CD Inclusion Complex

G 5 = Gliclazide-HP-β-CD Inclusion Complex

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 89 
Time Gliclazide GP1(± Stdv) GP2(± Stdv) GP3(± Stdv) GP4(± Stdv) GP5(± Stdv)
(min) pure(± Stdv)

0 0 0 0 0 0 0

5 10.56(±0.107) 15.19(±0.127) 19.87(±0.184) 22.48(±0.105) 29.34(±0.132) 31.29(±0.122)

10 12.71(±0.167) 39.47(±0.148) 40.45(±0.159) 42.72(±0.195) 45.61(±0.162) 47.83(±0.153)

20 17.37(±0.187) 45.54(±0.187) 47.91(±0.187) 50.86(±0.187) 53.74(±0.187) 54.67(±0.187)

30 22.64(±0.184) 50.71(±0.163) 51.04(±0.129) 54.84(±0.141) 55.27(±0.127) 58.54(±0.137)

40 24.96(±0.129) 53.02(±0.169) 54.37(±0.149) 56.52(±0.150) 59.81(±0.138) 60.69(±0.169)

50 27.19(±0.149) 56.11(±0.139) 59.52(±0.166) 60.94(±0.181) 61.54(±0.167) 62.15(±0.148)

60 29.93(±0.178) 58.19(±0.162) 61.21(±0.158) 62.61(±0.138) 63.04(±0.139) 64.21(±0.128)

Table No.17. Percentage of drug release Profile of Gliclazide complex with HP-β-CD

(Physical Mixture)

GP 1 = Gliclazide-HP-β-CD Physical Mixture

GP 2 = Gliclazide-HP-β-CD Physical Mixture

GP 3 = Gliclazide-HP-β-CD Physical Mixture

GP 4 = Gliclazide-HP-β-CD Physical Mixture

GP 5 = Gliclazide-HP-β-CD Physical Mixture

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 90 
Dissolution Profile

100

GP
P e r c e n ta g e o f D r u g

80
G1
R e le a s e d

60 G2

40 G3
G4
20 G5
0
0 10 20 30 40 50 60
Time (min)

Figure No.13. Dissolution Rate Data Profile graph of Gliclazide and its Inclusion
Complex (Kneading Method)

GP = Gliclazide Pure

G 1 = Gliclazide-HP-β-CD Inclusion Complex

G 2 = Gliclazide-HP-β-CD Inclusion Complex

G 3 = Gliclazide-HP-β-CD Inclusion Complex

G 4 = Gliclazide-HP-β-CD Inclusion Complex

G 5 = Gliclazide-HP-β-CD Inclusion Complex

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 91 
Dissolution Profile

100
P e r c e n ta g e o f D r u g

80 GP
GP2
R e le a s e d

60
GP3
40 GP4

20 GP5

0
0 10 20 30 40 50 60
Time (min)

Figure No.14. Dissolution Rate Data Profile graph of Gliclazide and its Complex

(Physical Mixture)

GP = Gliclazide Pure

GP 1 = Gliclazide-HP-β-CD Physical Mixture

GP 2 = Gliclazide-HP-β-CD Physical Mixture

GP 3 = Gliclazide-HP-β-CD Physical Mixture

GP 4 = Gliclazide-HP-β-CD Physical Mixture

GP 5 = Gliclazide-HP-β-CD Physical Mixture

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 92 
CORRELATION COEFFECIENT

Formulations Zero order Equation First order Equation


Regression coefficient (R2) Regression coefficient (R2)

GP 0.8965 0.9126

G1 0.6815 0.8347

G2 0.6661 0.376

G3 0.6305 0.8466

G4 0.5846 0.6178

G5 0.59 0.8756

GP1 0.7336 0.7234

GP2 0.7477 0.844

GP3 0.7302 0.8169

GP4 0.6595 0.7486

GP5 0.6488 0.7603

Table No.18. Correlation Coefficients (R2) Values in the analysis of Glicazide HP-β-CD
Dissolution data profile as per Zero order and First order of physical and inclusion
complexes

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 93 
100
Percentage of Drug Remaind

90
80
GP
70
G1
60
G2
50
G3
40
G4
30
20 G5
10
0
0 10 20 30 40 50 60
Time (min)

Figure No. 15. % Drug Remained Profile of Glaclazide HPβ-CD Inclusion complex.

100
Percentage of Drug Remaind

90
80
GP
70
GP1
60
GP2
50
GP3
40
GP4
30
20 GP5
10
0
0 10 20 30 40 50 60
Time (min)

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 94 
Figure No. 16. % Drug Remained Profile of Glaclazide HPβ-CD Physical
Mixture

2
Dissolution profile
1.8
1.6
log cum % drug rem ain

1.4 G1
1.2 G2
1 G3
0.8 G4
0.6 G5

0.4 Linear (G1)


Linear (G2)
0.2
Linear (G3)
0
Linear (G4)
0 10 20 30 40 50 60 70
Linear (G5)
Time(min)

Figure No. 17. Log % Drug Remained Profile of Glaclazide HPβ-CD Inclusion complex.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 95 
Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 96 
2.5

GP1
2
log cum % drug remain
GP2
GP3
1.5 GP4
GP5
1 Linear (GP1)
Linear (GP2)
Linear (GP3)
0.5
Linear (GP4)
Linear (GP5)
0
0 10 20 30 40 50 60 70
Time (min)

Figure No. 18. Log % Drug Remained Profile of Glaclazide HPβ-CD Physical Mixture.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 97 
DE Increase in
Formulations T 50% T 80% T 90% (30%) K1 (min-1) K1 (Folds)

GP - - - 13.62 0.005

G1 8.12 32.12 - 55.60 0.030 5.98

G2 7.24 27.12 - 60.10 0.023 4.6

G3 6.48 15.48 45.12 65.16 0.039 7.8

G4 6.24 11 30.48 68.98 0.042 8.6

G5 6 10.24 29.48 70.30 0.048 9.6

GP1 28.24 - - 36.03 0.012 2.4

GP2 26 - - 37.90 0.014 2.8

GP3 18.36 - - 40.52 0.014 2.8

GP4 14 - - 43.58 0.012 2.4

GP5 11.48 - - 45.09 0.014 2.8

Table No.19. Dissolution Parameter Profile of Gliclazide- HP-β-CD Inclusion

complexes and Physical Mixtures.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 98 
GZ TAB W GZ TAB D GZ TAB 1W GZ TAB D
Time (min) (± Stdv) (± Stdv) (± Stdv) (± Stdv)

0 0 0 0 0

5 12.18(±0.134) 14.49(±0.168) 47.25(±0.134) 52.05(±0.123)

10 15.53(±0.187) 18.62(±0.145) 68.68(±0.186) 74.27(±0.167)

20 20.87(±0.145) 23.21(±0.137) 79.37(±0.119) 83.58(±0.129)

30 24.06(±0.167) 26.35(±0.147) 86.41(±0.187) 91.46(±0.148)

40 27.12(±0.148) 29.12(±0.146) 89.16(±0.178) 95.17(±0.128)

50 29.03(±0.167) 32.98(±0.138) 93.59(±0.187) 97.09(±0.128)

60 30.91(±0.187) 34.48(±0.149) 95.87(±0.170) 98.21(±0.193)

Table No.20. Percentage of drug release Profile of Gliclazide- HP-β-CD Tablets.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 99 
GZ TAB W = Gliclazide Tablet (without HP-β-CD) by Wet Granulation method

GZ TAB D = Gliclazide Tablet (without HP-β-CD) by Direct compression method

GZ TAB 1 W = Gliclazide-HP-β-CD Tablet by Wet Granulation method

GZ TAB 2 D = Gliclazide-HP-β-CD Tablet by Direct compression method.

100
90
80
70
GZ TAB W
60
GZ TAB D
50
GZ TAB 1 W
40
GZ TAB 2 D
30
20
10
0
0 10 20 30 40 50 60

Figure No.19. Dissolution Rate Data Profile graph of Gliclazide- HP-β-CD Tablets

GZ TAB W = Gliclazide Tablet (without HP-β-CD) by Wet Granulation method

GZ TAB D = Gliclazide Tablet (without HP-β-CD) by Direct compression method

GZ TAB 1 W = Gliclazide-HP-β-CD Tablet by Wet Granulation method

GZ TAB 2 D = Gliclazide-HP-β-CD Tablet by Direct compression method.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 100 
CORRELATION COEFFECIENT

Formulations Zero order Equation First order Equation


Regression coefficient (R2) Regression coefficient (R2)

GZ TAB W 0.8431 0.8797

GZ TAB D 0.8292 0.8860

GZ TAB1 W 0.6624 0.9584

GZ TAB 2 D 0.3038 0.9743

Table No.21. Correlation Coefficients (R2) Values in the analysis of Glicazide HP-β-CD
Tablets Dissolution data profile as per Zero order and First order

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 101 
100

90

80
GZ TAB W
70
GZ TAB D
% Drug remain

60 GZ TAB 1 W
GZ TAB 2 D
50
Linear (GZ TAB W )
40 Linear (GZ TAB D )
Linear (GZ TAB 1 W )
30
Linear (GZ TAB 2 D )
20

10

0
0 10 20 30 40 50 60
Time(min)

Figure No. 20. % Drug Remained Profile of Glaclazide HPβ-CD Tablets.

2
1.8 GZ TAB W
Log cum % drug remain

1.6 GZ TAB D
1.4 GZ TAB W 1
1.2 GZ TAB 2D
1
Linear (GZ TAB W)
0.8
Linear (GZ TAB D)
0.6
Linear (GZ TAB W 1)
0.4
Linear (GZ TAB 2D)
0.2
0
0 10 20 30 40 50 60
Time(min)

Figure No. 21. Log % Drug Remained Profile of Glaclazide HPβ-CD Tablets.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 102 
Increase in
Formulation T 50% T 80% T 90% DE (30%) K1 (min-1) K1 (Folds)
23.3 37.2 46.2 10.07
GZ TAB W 1.001 -
17.8 31.2 42.6 12.71
GZ TABD 1.086 2.987
15.9 19.6 30.8 65.90
GZ TAB 1W 2.781 2.778
13.2 17.5 27.6 70.34
GZ TAB 2D 0.951 0.875

Table No.22. Dissolution Parameter Profile of Gliclazide- HP-β-CD Tablets

Formulation Hardness Hardness Hardness Avg. Hardness Std. Dev.

GZ Tabw 5.12(±0.132) 5.8(±0.132) 5.3(±0.132) 5.41(±0.132) 0.356

GZ Tab D 4.98(±0.132) 5.02(±0.132) 5.03(±0.132) 5.01(±0.132) 0.026

GZ Tab1w 5.03(±0.132) 5.12(±0.132) 5.09(±0.132) 5.08(±0.132) 0.045

GZ Tab 2D 4.97(±0.132) 5.06(±0.132) 5.12(±0.132) 5.05(±0.132) 0.075

Table No.23. Hardness Profile of Gliclazide-HP-β-CD Tablets.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 103 
Formulation Time(min) Time(min) Time(min) Avg. Time Std. Dev.

GZ Tabw 4.98 4.93 4.89 4.93 0.04


GZ Tab D
4.56 5.59 4.52 4.89 0.60
GZ Tab1w
4.79 4.76 4.71 4.75 0.04
GZ Tab 2D
4.26 4.21 4.29 4.25 0.04

Table No.24. Disintegration Time Profile of Gliclazide HP-β-CD Tablets.

Formulation Initial wt Weight lost % friability

GZ Tabw 1.537(±0.162) 0.009(±0.156) 0.455(±0.187)

GZ Tab D 1.502(±0.149) 0.004(±0.158) 0.226(±0.136)

GZ Tab1w 1.457(±0.176) 0.005(±0.137) 0.343(±0.187)

GZ Tab 2D 1.431(±0.165) 0.002(±0.149) 0.139(±0.129)

Table No.25. Friability Percentage Profile of Gliclazide-HP-β-CD Tablets

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 104 
Average %Deviation
Average Deviation Wt from Avg,
Formulation Weight(gm) Wt(±) Wt(±)

GZ Tabw 0.301 ±0.004 ±0.842

GZ Tab D 0.303 ±0.005 ±0.928

GZ Tab1w 0.301 ±0.004 ±0.938

GZ Tab 2D 0.301 ±0.003 ±0.906

Table No.26. Weight Variation Profile of Gliclazide HP-β-CD Tablets.

Sample GZ TAB W GZ TAB D GZ TAB 1 W GZ TAB 2 D

1 98.08 99.22 99.76 99.79

2 99.34 99.58 99.44 99.01

3 99.47 99.29 98.89 99.58


AVG
mg 98.96 99.36 99.36 99.46
std
dev± 0.77 0.19 0.44 0.40
c.v.
(%) 0.007 0.001 0.004 0.004

Table No.27. Drug Content Profile of Gliclazide HP-β-CD Tablets after Stability Test.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 105 
GZ TAB W = Gliclazide Tablet (without HP-β-CD) by Wet Granulation method

GZ TAB D = Gliclazide Tablet (without HP-β-CD) by Direct compression method

GZ TAB 1 W = Gliclazide-HP-β-CD Tablet by Wet Granulation method

GZ TAB 2 D = Gliclazide-HP-β-CD Tablet by Direct compression method.

GZ TAB W GZ TAB D GZ TAB 1 GZ TAB 2D


Time min (± Stdv) (± Stdv) W(± Stdv) (± Stdv)

0 0 0 0 0

5 11.83(±0.168) 14.1(±0.187) 46.86(±0.130) 51.64(±0.141)

10 15.16(±0.176) 18.19(±0.189) 68.22(±0.167) 73.82(±0.139)

20 20.48(±0.158) 22.92(±0.167) 79.02(±0.120) 83.19(±0.187)

30 23.74(±0.156) 26.04(±0.167) 86.02(±0.183) 91.05(±0.189)

40 26.74(±0.138) 28.71(±0.187) 88.71(±0.165) 94.78(±0.174)

50 28.58(±0.145) 32.59(±0.187) 93.18(±0.175) 96.68(±0.148)

60 30.5(±0.138) 34.09(±0.176) 95.45(±0.187) 97.8(±0.136)

Table No.28. Percentage of drug release Profile of Gliclazide- HP-β-CD Tablets

After Stability Study

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 106 
GZ TAB W = Gliclazide Tablet (without HP-β-CD) by Wet Granulation method

GZ TAB D = Gliclazide Tablet (without HP-β-CD) by Direct compression method

GZ TAB 1 W = Gliclazide-HP-β-CD Tablet by Wet Granulation method

GZ TAB 2 D = Gliclazide-HP-β-CD Tablet by Direct compression method.

Dissolution Profile

100
P e r c e n ta g e o f D r u g

80
GZ TAB W
R ele ase d

60 GZ TAB D
40 GZ TAB 1 W
GZ TAB 2 D
20

0
0 10 20 30 40 50 60
Time (min)

Figure No.22. Dissolution Rate Data Profile graph of Gliclazide- HP-β-CD Tablets

After Stability Study.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 107 
GZ TAB W = Gliclazide Tablet (without HP-β-CD) by Wet Granulation method

GZ TAB D = Gliclazide Tablet (without HP-β-CD) by Direct compression method

GZ TAB 1 W = Gliclazide-HP-β-CD Tablet by Wet Granulation method

GZ TAB 2 D = Gliclazide-HP-β-CD Tablet by Direct compression method.

Formulation Hardness Hardness Hardness Avg. Hardness Std. Dev.

GZ Tab w 5.09(±0.112) 5.69(±0.136) 5.19(±0.128) 5.32 0.32

GZ Tab D 4.76(±0.148) 5.01(±0.187) 4.89(±0.128) 4.88 0.13

GZ Tab1w 4.92(±0.148) 5.03(±0.137) 4.97(±0.176) 4.97 0.06

GZ Tab 2D 4.75(±0.167) 4.90(±0.186) 5.02(±0.138) 4.89 0.14

Table No.29. Hardness Profile of Gliclazide-HP-β-CD Tablets after stability test.

Formulation Time(min) Time(min) Time(min) Avg. Time Std. Dev.

GZ Tabw 5.06 4.98 4.97 5.00 0.05


GZ Tab D
4.62 5.64 4.61 4.95 0.59
GZ Tab1w
4.86 4.83 4.84 4.84 0.02
GZ Tab 2D
4.32 4.32 4.36 4.33 0.02
Table No.30. Disintegration Time Profile of Gliclazide HP-β-CD Tablets after stability
test.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 108 
Formulation Initial wt Weight lost % friability

GZ Tabw 1.532(±0.102) 0.008(±0.147) 0.457

GZ Tab D 1.495(±0.192) 0.004(±0.136) 0.228

GZ Tab1w 1.448(±0.132) 0.004(±0.187) 0.344

GZ Tab 2D 1.423(±0.148) 0.003(±0.128) 0.141

Table No.31. Friability Percentage Profile of Gliclazide-HP-β-CD Tablets after stability

test.

Average %Deviation
Average Deviation Wt from Avg,
Formulation Weight(gm) Wt(±) Wt(±)

GZ Tabw 0.300 ±0.005 ±0.886

GZ Tab D 0.301 ±0.006 ±0.960

GZ Tab1w 0.300 ±0.004 ±0.972

GZ Tab 2D 0.300 ±0.004 ±0.930

Table No.32. Weight Variation Profile of Gliclazide HP-β-CD Tablets after Stability
Study.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 109 
SUMMARY AND CONCLUSION

Low RSD values ensured reproducibility of the method. No interference

from excipients used was observed. Thus method was found suitable for the

estimation of Gliclazide content in dissolution fluids. The calibration curve shown

in fig.no.5 and data table no.10 was used for the purpose.

The melting point of gliclazide, gliclazide-HP-β-CD complexes and

physical mixtures found to be in between 3190c to 3250c.

The solubility of gliclazide is increasing as the concentration of

solvent/surfactant increases. Solubility is more in a blend of water+SLS than the

alcohol and glycerine with water, also showing more solubility in pH 6 then the

other pH. Data is given in table no.11&12.

The phase solubility diagrams for the complex formation between

gliclazide HP β-CD shown in table no.13 and figure no.6. The aqueous solubility

of the gliclazide was increased linearly as a function of the concentration of HP β-

CD. The aqueous solubility diagrams of gliclazide HP β-CD complexes can be

classified as AL type according to Higuchi and Connors. Because the straight line

had a slope < 1 in each case, the increase in solubility was due to the formation of

a 1:1 M complex in solution with HP β-CD.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 110 
The apparent stability constant (Kc) in each case was calculated from the

slope of the corresponding linear plot of the phase solubility diagram according to

the equation, Kc = Slope/ So (1-Slope), where So is the solubility of the drug in the

absence of solubilizer. The estimated Kc values of gliclazide with HP β-CD

complexe are 256.3 M-1. The values of Kc indicated that all the complexes formed

between gliclazide and HP-β-CDs are quite stable.

The Rf values were found to be 0.738 and 0.869 for pure gliclazide,

gliclazide-HP-β-CD complex respectively, indicating that the spots of the sample

were indeed that of the unchanged gliclazide. As the Rf values of gliclazide,

gliclazide HP-β-CD complex coinciding and as no other spots were observed it

can be concluded that gliclazide and HP-β-CD are compatible without any

chemical change or reaction.

The IR spectra of gliclazide, gliclazide-HP-β-CD inclusion complex and

physical mixtures are shown in figure no.7&8 respectively. The IR spectral

observations indicated no interaction between gliclazide and HP-β-CD complexes.

The DSC thermograms of gliclazide HP β-CD inclusion complex and

physical mixtures are shown in figure no.9&10. The DSC curve of HP β-CD

showed broad endothermic peaks. In the thermograms of gliclazide HP β-CD, the

intensity (or height) of the endothermic peak was reduced indicating interaction of

gliclazide with HP-β-CD and absence of crystalline drug and its complete

complexation with HP-β-CD.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 111 
XRD patterns of gliclazide and complexes with HP-β-CD are shown in

figure no.11&12. The diffraction peaks were much reduced in the case of

glicazide HP-β-CD complexes. The disappearance of gliclazide crystalline peaks

confirmed the stronger drug amorphization and entrapment in HP β-CD.

Low C.V. values in the percent drug content ensured uniformity of drug

content in all batches. The coefficient of variation (C.V.) in the percent drug

content was found to be less than 1.0 percent in all the batches prepared, and given

in table no.14&15.

Solid inclusion complexes of Gliclazide-HP-β-CD in 1:0.5, 1:1, 1:1.5, 1:2

and 1:2.5 ratios were prepared by kneading method and physical mixture. The

dissolution rate of gliclazide from CDs complex and physical mixture system was

studied using phosphate buffer solution of pH 7.4 as a dissolution fluid. The result

is given in table no. 16&17and shown figure no.13&14.

The dissolution of gliclazide was higher from all the gliclazide HP-β-

cyclodextrin complexes prepared when compared to gliclazide HP-β-cyclodextrin

physical mixture and pure gliclazide drug. The gliclazide HP-β-cyclodextrin

physical mixture showing more dissolution than the pure gliclazide drug. The

dissolution data were fitted into mathematical models such as zero order and first

order models to assess the kinetics and mechanism of dissolution are given in

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 112 
figures no.15-18. The dissolution data obeyed first order kinetic model. The

correlation coefficient (r2) values observed in the analysis of dissolution data as

per the above models are given in table no.18. All the ‘r2’ values were greater

indicating that the dissolution of gliclazide from all the complexes obeyed first

order model. The corresponding dissolution rate was calculated from the slopes of

the linear plots.

Dissolution efficiency (DE30) values were calculated as per Khan. T50 (time

taken for 50% dissolution), T80 (time taken for 80% dissolution) and T90 (%)

values were recorded from the dissolution profiles. The dissolution parameters are

summarized in table no.19.

The K1 and DE30values were increased as the proportion of CD in the

complex system was increased in each batch. The increase in K1 (no. of folds)

with CD systems is shown in table no.19. Gliclazide HP-β-CD complexes gave

higher enhancement in the dissolution rate and efficiency when compared to

Gliclazide HP-β-CD physical mixture.

The gliclazide tablets prepared with HP-β-CD by wet granulation and direct

compression methods. The physical characteristics like size, shape, thickness and

appearance of all series of tablets prepared were found good.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 113 
The dissolution rate of gliclazide tablets prepared with HP-β-CD by various

methods was studied using phosphate buffer solution of pH 7.4 as the dissolution

fluid. The results are given in table no.20 and shown in figure no.19.

The dissolution of gliclazide tablets prepared with HP-β-CD was higher

when compared with gliclazide tablets prepared without HP-β-CD. The dissolution

data were fitted into various models such as zero order and first order model are

given in figure no.20&21. The correlation coefficient (R2) values observed in the

analysis of dissolution data as per the above models are given in table no.21. All

the R2 values indicating that the dissolution of gliclazide from the tablets prepared

obeyed first order model. The dissolution rates were calculated from the slope of

the linear plots.

Dissolution efficiency (DE30) values are calculated as per Khan. T50, T80

and T90 values were recorded from the dissolution profiles. The dissolution

parameters are given in table no.22. The increase in K1 and K1 no. of folds of all

the tablets of gliclazide prepared with HP-β-CD found when compared with

tablets of glicazide prepared without HP-β-CD and shown in table no.22.

The hardness of the gliclazide tablets is 5.01- 5.41 kg/cm2. The

disintegration time of tablets is in between 4.25- 4.93 min. The friability

percentage and weight variation of all the tablets are found within the standard

limits, the data is given in table no.23-26.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 114 
The tablets prepared in the present investigation were tested for their

stability by storing at 400c and 75% RH for a period of 3 months. The stored

products were evaluated for drug content, dissolution rate, weight variation,

friability, and hardness and disintegration tests. Drug content of all the series

tablets remained unaltered after storage for 3 months are given in table no.27. The

dissolution characteristics of all the series tablets remained unaltered during the

storage period are given in table no 28. And shown in figure no.22. Weight

variation, Friability percent of all series of tablets remains much unaltered after

storage for 3 months; hardness and disintegration of all the tablet remains intact

without much change in their characteristics after storage for 3 months in a

prescribed temperature and humidity are given in table no.29-32.

The present work has been undertaken with an overall objective of studying

the complexation of gliclazide, BCS class II drugs with hydroxypropyl-β-

cyclodextrin to evaluate the feasibility of enhancing their solubility, dissolution

rate, bioavailability and therapeutic efficacy. The feasibility of formulating the CD

complexes into tablets with enhanced dissolution rate characteristics was also

investigated.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 115 
Complexation of gliclazide with hydroxypropyl-β-cyclodextrin was

investigated by solubility in different pH and blend of solvent/surfactants in

different concentration with water and phase solubility study. The effect of

hydroxypropyl-β-cyclodextrin on the solubility of gliclazide was also derived from

phase solubility studies. Solid inclusion complexes of gliclazide-HP-β-CD were

prepared by kneading method and physical mixture employing different ratios of

gliclazide-HP-β-CD. The inclusion complexes prepared by kneading and physical

methods were evaluated and characterized by TLC, IR, XRD and DSC studies.

Kinetics and mechanism of drug dissolution from the inclusion complexes were

evaluated by studying dissolution of solid inclusion complexes. Selected

gliclazide-HP-β-CD complexes were formulated in to tablets by wet granulation

and direct compression methods and the resulting tables were evaluated for

dissolution rate and other physical properties of tablets. Gliclazide-HP-β-CD

tablets were also subjected to stability evaluation as per ICH guidelines.

From the results obtained the following conclusions are drawn.

¾ The phase solubility diagram of gliclazide was of type AL with HP β-CD.

¾ The phase solubility studies indicated the formation of gliclazide- HP β-CD

inclusion complexes at a 1:1 M ratio in solustion. The complexes formed

were quite stable.

¾ The aqueous solubility of gliclazide was increased linearly as a function of

concentration of the HP β-CD.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 116 
¾ Inclusion complex prepared by kneading method exhibited higher

solubilizing efficiency when compared with physical mixture of gliclazide-

HP β-CD.

¾ DSC and XRD indicated better drug inclusion in HP β-CD, and good drug

amorphization and entrapment in HP β-CD.

¾ IR spectral studies indicated no chemical interaction between the

gliclazide- HP β-CD.

¾ The compatibility test between gliclazide- HP β-CD done by TLC method

indicated that gliclazide- HP β-CD are compatible with out any chemical

change or reaction.

¾ Gliclazide- HP β-CD complexes exhibited higher rates of dissolution and

dissolution efficiency values than the uncomplexed gliclazide.

¾ Gliclazide- HP β-CD (1: 2.5) complex exhibited 9.6 fold increases in the

dissolution rate.

¾ Gliclazide- HP β-CD complexes could be formulated in compressed tablets

by wet granulation and direct compression methods. All the Gliclazide-

HP-β-CD tablets prepared fulfilled the official specifications of hardness,

friability and disintegration time and gave rapid dissolution of the contained

drug.

¾ All gliclazide- HP β-CD tablets exhibited higher rates of dissolution and

efficiency values than the tablets prepared without HP β-CD.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 117 
¾ The gliclazide-HP β-CD tablets prepared by direct compression gave a little

increased rate of dissolution and efficiency when compared to the tablets

prepared by wet granulation method.

¾ Cyclodextrin complexation (HP- β-CD) has markedly enhanced the

absorption rate gliclazide.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 118 
BIBILOGRAPHY.

1. Loftsson T, Brewster, ME. Pharmaceutical applications of cyclodextrin.1. Drug

solubilization

and stabilization. J Pharm Sci. 1996; 85:1024-1074.

2. Loftsson, T, Brewster, M. “Cyclodextrin as pharmaceutics excipients” Pharma

Manager. 1997; 5:22-31.

3. Loftsson T, and Brewster, ME. J Pharm Sci. 1996; 85; 1017

4. Rajewski R A, and Stella V.J, J Pharm Sci.1996; 85; 1142

5. Szejtli J, Drug Investigation, 1990; 2(suppl.); 11.

6. Szejtli J, Cyclodextrin and their inclusion complexes, akademiai,Kiado, Budapest,

Hungary,1982; p.13.

7. Gandhi R.B. and Karara A.H, Drug Dev.Ind, Pharm., 1988; 14; 657.

8. Fromming K.H. and Szejtli J., Cyclodextrina in Pharmacy, Kluwer Acad. Publ.,

Dordrecht, 1994.

9. McCandless R, Yalkowsky SH. Effect of hydroxypropyl-beta-cyclodextrin and pH on

the

Solubility of levemopamil HCl. J Pharm Sci. 1998; 87(12): 1639-42.

10. Szejtli J, Bolla-Pusztai E, Tardy-Lengyel M, Szabo P, Ferenczy T. Preparation,

properties and biological activity of beta-cyclodextrin inclusion complex of menadione.

Pharmazie. 1983; 38(3):189-193.

11. Aguiar MM, Rodrigues JM, Silva Cunha A. Encapsulation of insulin-cyclodextrin

complex in PLGA microspheres: a new approach for prolonged pulmonary insulin

delivery. J Microencapsul. 2004; 21(5): 553-64.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 119 
12. Loftsson T, Magnusdottir A, Masson M, Sigurjonsdottir JF. Self-association and

cyclodextrin solubilization of drugs, J Pharm Sci. 2002; 91(11):2307-2316.

13. Loftsson T, Masson M, Brewster ME, Selfassociation of cyclodextrins and

cyclodextrin complexes, J Pharm Sci. 2004; 93(5):2307-2316.

14. Hersey A, Robinson BH, Kelly HC. Mechanism of inclusion compound formation for

binding of organic dyes, ions and surfactants to alpha cyclodextrin studied by kinetic

methods based on competition experiments. J Chem Soc, Faraday Trans 1. 1986;

82:1271Y1287.

15. Cramer F, Saenger W, Satz HC. Inclusion compounds. ΧΙХ. The formation of

inclusion compounds of alpha cyclodextrin in aqueous solutions, thermodynamics and

kinetics. J Am Chem Soc.1967; 89:14Y20.

16 Loftsson T, Brewester M. Pharmaceutical applications of cyclodextrins. 1. Drug

solubilization and stabilization. J Pharm Sci. 1996; 85:1017Y1025.

17. Uekama K, Otagiri M. Cyclodextrins in drug carrier systems. Crit Rev Ther Drug

Carrier Sys. 1987; 3:1Y40.

18. Szejtli J. Medicinal applications of cyclodextrins. Med Res Rev.1994; 14:353Y386.

19. Thomson DO. Cyclodextrins–enabling excipients: their present and future use in

pharmaceuticals. Crit Rev Ther Drug Carr Sys.1997; 14:1Y104.

20. Jayachandra Babu R, Pandit JK. Cyclodextrin inclusion complexes: oral applications.

Eastern Pharmacist. 1995; 38:37Y42.

21. Irie T, Uekama K. Pharmaceutical applications of cyclodextrins.III. Toxicological

issues and safety evaluation. J Pharm Sci.1997; 86:147Y162.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 120 
22. Stella VJ, Rajeswski RA. Cyclodextrins: their future in drug formulation and

delivery. Pharm Res. 1997; 14:556Y567.

23. Uekama K, Hirayama F, Irie T. Cyclodextrin drug carrier systems. Chem Rev. 1998;

98:2045Y2076.

24. Loftsson T, Ólafsson JH. Cyclodextrins: new drug delivery system in dermatology.

Int J Dermatol. 1998; 37:241Y246.

25. Loftsson T. Increasing the cyclodextrin complexation of drugs and drug

bioavailability through addition of water-soluble polymers.

26. Blanchard J, Stefan P. Some important considerations in the use of cyclodextrins.

Pharm Res. 1999; 16:1796Y1798.

27. Muller BW, Brauns U. Hydroxypropyl-beta-cyclodextrin derivatives: influence of

average degree of substitution on complexing ability and surface activity. J Pharm Sci.

1986; 75:571Y572.

28. Szejtli J in controlled Drug Bioavailability, Vol 3, New York: Smiden WF and Ball

LA, Bds Wiley, 1986, p365.

29. Okimoto K, Rajewski RA, Uekama K, Jona JA,Stella VJ. The interaction of charged

and

uncharged drugs with neutral (HP-beta-CD) and anionically charged (SBE7-beta-CD)

betacyclodextrins, Pharm Res. 1996; 13(2):256-264.

30. Kim Y, Oksanen DA, Massefski W, Blake JF, Duffy EM, Chrunyk B. Inclusion

complexation of ziprasidone mesylate with beta-cyclodextrin sulfobutyl ether. J Pharm

Sci. 1998; 87:1560Y1567.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 121 
31. Tros de Ilarduya MC, Martin C, Goni MM, Martinez-Oharriz MC. Solubilization and

interaction of sulindac with beta-cyclodextrin in the solid state and in aqueous solution.

Drug Dev Ind Pharm.1998; 24:301Y306.

32. Veiga F, Fernandes C, Maincent P. Influence of the preparation method on the

physicochemical properties of tolbutamide/cyclodextrin binary systems. Drug Dev Ind

Pharm. 2001; 27:523Y532.

33. Loftsson T, Peterson DS. Cyclodextrin solubilization of ETH-615, a zwitterionic

drug. Drug Dev Ind Pharm. 1998; 24:365Y370.

34. Loftsson T, Guomundsdottir TK, Frioriksdottir H. The influence of water-soluble

polymers and pH on hydroxypropyl-α-cyclodextrin complexation of drugs. Drug Dev Ind

Pharm. 1996; 22:401Y406.

35. Nath BS, Shivkumar HN. A 2(3) Factorial studies on factors influencing Meloxicam

β-cyclodextrin complexation for better solubility. Indian J Pharm Sci. 2000; 62:129Y132.

36. Cappello B, Carmignani C, Iervolino M, La Rotonda MI, Saettone MF. Solubilization

of tropicamide by hydroxypropyl-beta-cyclodextrin and water-soluble polymers: in vitro/

in vivo studies. Int J Pharm. 2001; 213:75Y81.

37. Faucci MT, Mura P. Effect of water-soluble polymers on naproxen complexation

with natural and chemically modified beta-cyclodextrins. Drug Dev Ind Pharm. 2001;

27:909Y917.

38. Granero G, de Bertorello NM, Longhi M. Solubilization of a naphthoquinone

derivative by hydroxypropyl-beta-cyclodextrin (HP-β-CD) and polyvinylpyrrolidone

(PVP-K30). The influence of PVPK30 and pH on solubilizing effect of HP-β-CD. Boll

Chim Farm.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 122 
2002; 141:63Y66.

39. Tokumura T, Nanda M, Tsushima Y, et al. Enhancement of bioavailability of

cinnarizine from its beta-cyclodextrin complex on oral administration with DL-

phenylalanine as a competing agent. J Pharm Sci. 1986; 75:391Y394.

40. Muller BW, Albers E. Effect of hydrotropic substances on the complexation of

sparingly soluble drugs with cyclodextrin derivatives and the influence of cyclodextrin

complexation on the pharmacokinetics of the drugs. J Pharm Sci. 1991; 80:599Y604.

41. Redenti E, Szente L, Szetli J. Drug/cyclodextrin/hydroxy acid multicomponent

systems. Properties and pharmaceutical applications. J Pharm Sci. 2000; 89:1Y8.

42. Li P, Zhao L, Yalkowsky SH. Combined effect of cosolvent and cyclodextrin on

solubilization of nonpolar drugs. J Pharm Sci. 1999; 88:1107Y1111.

43. Miyake K, Irie T, Arima H, et al. Characterization of itraconazole/ 2-hydroxypropyl-

beta-cyclodextrin inclusion complex in aqueous propylene glycol solution. Int J Pharm.

1999; 179:237Y245.

44. Zarzycki PK, Lamparczyk H. The equilibrium constant of β-cyclodextrin-

phenolphtalein complex; influence of temperature and tetrahydrofuran addition. J Pharm

Biomed Anal. 1998; 18:165Y179.

45. Jain AC, Adeyeye MC. Hygroscopicity, phase solubility and dissolution of various

substituted sulfobutylether beta-cyclodextrins (SBE) and danazol-SBE inclusion

complexes. Int J Pharm. 2001; 212:177Y186.

46. Nagase Y, Hirata M, Wada K, et al. Improvement of some pharmaceutical properties

of DY-9760e by sulfobutyl ether beta-cyclodextrin. Int J Pharm. 2001; 229:163Y172.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 123 
47. Mura P, Adragna E, Rabasco AM, et al. Effects of the host cavity size and the

preparation method on the physicochemical properties of ibuproxam-cyclodextrin

systems. Drug Dev Ind Pharm. 1999; 25:279Y287.

48. Chowdary KPR, Nalluri BN. Nimesulide and beta-cyclodextrin inclusion complexes:

physicochemical characterization and dissolution rate studies. Drug Dev Ind Pharm.

2000; 26:1217Y1220.

49. Palmeiri GF, Angeli DG, Giovannnucci G, Martelli S. Inclusion of methoxytropate in

β− and hydroxylpropyl β-cyclodextrins: Comparision of preparation methods. Drug Dev

Ind Pharm. 1997; 23:27Y37.

50. Palmieri GF, Wehrle P, Stamm A. Inclusion of vitamin D2 in β-cyclodextrin:

evaluation of different complexation methods. Drug Dev Ind Pharm. 1993; 19:875Y885.

51. Moyano JR, Arias MJ, Gines JM, Perez JI, Rabasco AM. Dissolution behavior of

oxazepam in the presence of cyclodextrins: evaluation of oxazepam dimeb binary system.

Drug Dev Ind Pharm. 1997; 23:379Y385.

52. Castillo JA, Canales JP, Garcia JJ, Lastres JL, Bolas F, Torrado JJ. Preparation and

characterization of albendazole beta-cyclodextrin complexes. Drug Dev Ind Pharm. 1999;

25:1241Y1248.

53. Diaz D, Escobar Llanos CM, Bernad MJB. Study of the binding in an aqueous

medium of inclusion complexes of several cyclodextrins involving fenoprofen calcium.

Drug Dev Ind Pharm. 1999; 25:107Y110.

54. Pose-Vilarnovo B, Perdomo-Lopez I, Echezarreta-Lopez M, Schroth-Pardo P, Estrada

E, Torres-Labandeira JJ. Improvement of water solubility of sulfamethizole through its

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 124 
complexation with β- and hydroxypropyl-β-cyclodextrin—Characterization of the

interaction in

Solution and in solid state. Eur J Pharm Sci. 2001; 13:325Y331.

55. Veiga F, Fernandes C, Maincent P. Influence of the preparation method on the

physicochemical properties of tolbutamide/cyclodextrin binary systems. Drug Dev Ind

Pharm. 2001; 27:523Y532.

56. Pedersen M. Effect of hydrotropic substances on the complexation of clotrimazole

with beta cyclodextrin. Drug Dev Ind Pharm. 1993; 19:439Y448.

57. Londhe V, Nagarsenker M. Comparision between Hydroxypropyl-β-cyclodextrin and

polyvinyl pyrrolidine as carriers for carbamazepine solid dispersions. Indian J Pharm Sci.

1999; 61:237Y240.

58. Bettinetti G, Gazzaniga A, Mura P, Giordano F, Setti M. Thermal behavior and

dissolution properties of naproxen in combinations with chemically modified beta-

cyclodextrins. Drug Dev Ind Pharm. 1992; 18:39Y53.

59. Becket G, Schep LJ, Tan MY. Improvement of the in vitro dissolution of praziquantal

by complexation with alpha-, betaand gamma-cyclodextrins. Int J Pharm. 1999;

179:65Y71.

60. Ueda H, Ou D, Endo T, Nagase H, Tomono K, Nagai T. Evaluation of a sulfobutyl

ether beta-cyclodextrin as a solubilizing/stabilizing agent for several drugs. Drug Dev Ind

Pharm. 1998; 24:863Y867.

61. Sanghavi NM, Choudhari KB, Matharu RS, Viswanathan L.Inclusion complexation

of Lorazepam with beta-cyclodextrin. Drug Dev Ind Pharm. 1993; 19:701Y712.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 125 
62. Ahn HJ, Kim KM, Choi SJ, Kim CK. Effects of cyclodextrin derivatives on

bioavailability of ketoprofen. Drug Dev Ind Pharm.1997; 23:397Y401.

63. Dhanaraju MD, Santil Kumaran K, Baskaran T, Moorthy MSR. Enhancement of

bioavailability of griseofulvin by its complexationwith beta-cyclodextrin. Drug Dev Ind

Pharm. 1998; 24:583Y587.

64. Veiga MD, Diaz PJ, Ahsan F. Interactions of griseofulvin with cyclodextrins in solid

binary systems. J Pharm Sci. 1998; 87:891Y900.

65. Lotter J, Krieg HM, Keizer K, Breytenbach JC. The influence of beta-cyclodextrin on

the solubility of chlorthalidone and its enantiomers. Drug Dev Ind Pharm. 1999;

25:879Y884.

66. Askrabic JM, Rajic DS, Tasic L, Djuric S, Kasa P, Hodi KP. Etodolac and solid

dispersion with β-cyclodextrin. Drug Dev Ind Pharm. 1997; 23:1123Y1129. AAPS

PharmSciTech 2005; 6 (2) Article 43 (http://www.aapspharmscitech.org).

67. Cavallari C, Abertini B, Rodriguez MLG, Rodriguez L. Improved dissolution

behavior of steam granulated piroxicam. Eur J Pharm Biopharm. 2002; 54:65Y73.

68. Chowdary KPR, Rao SS. Investigation of dissolution enhancement of itraconazole by

complexation with β-, and hydroxypropyl-β-cyclodextrins. Indian J Pharm Sci. 2001;

63:438Y441.

69. Ghorab MK, Adeyeye MC. Enhancement of ibuprofen dissolution via wet granulation

with beta cyclodextrin. Pharm Dev Technol. 2001; 6:305Y314.

70. Arias MJ, Moyano JR, Munoz P, Gines JM, Justo A, Giordano F. Study of

omeprazole-gamma-cyclodextrin complexation in the solid state. Drug Dev Ind Pharm.

2000; 26:253Y259.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 126 
71. Uekama K, Fujinaga T, Hirayama F, et al. Improvement of the oral bioavailability of

digitalis glycosides by cyclodextrin complexation. J Pharm Sci. 1983; 72:1338Y1341.

72. McCandless R, Yalkowsky SH. Effect of hydroxypropyl-betacyclodextrin and pH on

the solubility of levemopamil HCl. J Pharm Sci. 1998; 87:1639Y1642.

73. Trapani G, Latrofa A, Franco M, et al. Complexation of zolpidem with 2-

hydroxypropyl-β-, methyl-β-, 2-hydroxypropyl-γ-cyclodextrins: Effect on aqueous

solubility, dissolution rate and ataxic activity in rats. J Pharm Sci. 2000; 89:1443Y1451.

74. Latrofa A, Trapani G, Franco M, et al. Complexation of phenytoin with some

hydrophilic cyclodextrins: Effect on aqueous solubility, dissolution rate and anti-

covulsant activity in mice. Eur J Pharm Biopharm. 2001; 52:65Y73.

75. Miyake K, Arima H, Hiramaya F, et al. Improvement of solubility and oral

bioavailability of rutin by complexation with 2-hydroxypropylbeta-cyclodextrin. Pharm

Dev Technol. 2000; 5:399Y407.

76. Kang J, Kumar V, Yang D, Chowdhury PR, Hohl RJ. Cyclodextrin complexation:

influence on the solubility, stability, and cytotoxicity of camptothecin, an antineoplastic

agent. Eur J Pharm Sci. 2002; 15:163Y170.

77. Zhao L, Li P, Yalkowsky SH. Solubilization of fluasterone. J Pharm Sci. 1999;

88:967Y969.

78. Kaukonen AM, Lennernas H, Mannermaa JP. Water-soluble Beta cyclodextrin in

paediatric oral solutions of spiranolactone: preclinical evalution of spiranolactone

bioavailability from solutions of beta cyclodextrin derivatives in rats. J Pharm Pharmacol.

1998; 50:611Y619.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 127 
79. Arima H, Yunomae K, Miyake K, Irie T, Hirayama F, Uekama K. Comparative

studies of the enhancing effects of cyclodextrins on the solubility and oral bioavailability

of tacrolimus in rats. J Pharm Sci. 2001; 90:690Y701.

80. Bettinetti G, Mura P, Faucci MT, Sorrenti M, Setti M. Interaction of naproxen with

noncrystalline acetyl beta- and acetyl gammacyclodextrins in the solid and liquid state.

Eur J Pharm Sci. 2002; 15:21Y29.

81. Masson, Thorstein Loftsson, Gisli Masson, Einar Stefansson, Cyclodextrins as

permeation

enhancers: some theoretical evaluations and in vitro testing, J Controlled Rel. 1997;

59:107-118

82. Marttin E, Verhoef JC, Spies F, van der Meulen J, Nagelkerke JF, Koerten HK,

Merkus FWHM. The effect of methylated β-cyclodextrins on the tight junctions of the rat

nasal respiratory epithelium: Electron microscopic and confocal laser scanning

microscopic visualization studies, J Controlled Rel. 1999; 57:205-213.

83. Highuchi. Physical chemical analysis of percutaneous absorption process from

creams and

Ointments J Soc Cosmetic Chemists. 1960; 11:85-97.

84. Hovgaard L, Bronsted H. Drug delivery studies in Caco-2 monolayers. IV.

Absorption enhancer effects of cyclodextrins. Pharm Res.1995; 12(9):1328-1332.

85. Masson, Thorstein Loftsson, Gisli Masson, Einar Stefansson, Cyclodextrins as

permeation

Enhancers: some theoretical evaluations and In Vitro testing. J Controlled Rel. 1997;

59:107-118.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 128 
86. Nakanishi K, Nadai T, Masada M, Miyajima K. Effect of cyclodextrins on biological

membrane. II. Mechanism of enhancement on the intestinal absorption of non-absorbable

drug by cyclodextrins. Chem Pharm Bull. 1992; 40(5):1252-1256.

87. Makita T, Ojima N, Hashimoto Y et al. Chronic oral toxicity study of β-cyclodextrin

in rats. Oyo Yakuri. 1975; 10:449-458.

88. Szejtli J, Sebestyen G. Resorption, metabolism and toxicity studies on the per oral

application of β-cyclodextrin. Starch Starke. 1979; 31:385-389.

89. Szejtli J, Cyclodextrins Properties and Applications, Drug Investigation. 1990; 4:11-

21.

90. French D. The Schardinger dextrins. Advances in Carbohydrate Chemistry. 1957;

12:189-260.

91. Rajewski RA, Stella VJ. Pharmaceutical applications of cyclodextrins.2. In vivo drug

delivery. J Pharm Sci. 1996; 85:1142Y1168.

92. Nicolazzi C, Venard V, Le Faou A, Finance C. In vitro antiviral activity of the

gancyclovir complexed with beta cyclodextrin on human cytomegalovirus strains.

Antiviral Res. 2002; 54:121Y127.

93. Blanchard J, Ugwu SO, Bhardwaj R, Dorr RT. Development and testing of an

improved of phenytoin using 2-hydroxypropyl-betacyclodextrin. Pharm Dev Technol.

2000; 5:333Y338.

94. Scalia S, Villani S, Casolari A. Inclusion complexation of the sunscreening agent 2–

ethyl hexyl–p–dimethyl aminobenzoate with hydroxypropyl-β-cyclodextrin: effect on

photostability. J Pharm Pharmacol. 1999; 51:1367Y1374.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 129 
95. Serni U. Rheumatic diseases—clinical experience with piroxicambeta-cyclodextrin.

Eur J Rheumatol Inflamm. 1993; 12:47Y54.

96. Kim JH, Lee SK, Ki MH, et al. Development of parenteral formulation for a novel

angiogenesis inhibitor, CKD-732 through complexation with hydroxypropyl-β-

cyclodextrin. Int J Pharm. 2004; 272:79Y89.

97. Nagase Y, Arima H, Wada K, et al. Inhibitory effect of sulfobutyl ether beta-

cyclodextrin on DY-9760e-induced cellular damage: In vitro and in vivo studies. J Pharm

Sci. 2003; 92:2466Y2474.

98. Loftssona T, Jarvinen T. Cyclodextrins in ophthalmic drug delivery. Adv Drug Deliv

Rev. 1999; 36:59Y79.

99. Babu R, Pandit JK. Effect of aging on the dissolution stability of glibenclamide/beta-

cyclodextrin complex. Drug Dev Ind Pharm.1999; 25:1215Y1219.

100. Cwiertnia B, Hladon T, Stobiecki M. Stability of Diclofenac sodium in the inclusion

complex in the beta cyclodextrin in the solid state. J Pharm Pharmacol. 1999;

51:1213Y1218.

101. Li J, Guo Y, Zografi G. The solid-state stability of amorphous quinapril in the

presence of beta-cyclodextrins. J Pharm Sci. 2002; 91:229Y243.

102. Brewster ME, Loftsson T, Estes KS, Lin JL, Friðriksdóttir H. Effects of various

cyclodextrins on solution stability and dissolution rate of doxorubicin hydrochloride. Int J

Pharm. 1992; 79:289Y299.

103. Ma DQ, Rajewski RA, Velde DV, Stella VJ. Comparative effects of (SBE) 7m-beta-

CD and HP-beta-CD on the stability of two anti-neoplastic agents, melphalan and

carmustine. J Pharm Sci. 2000; 89:275Y287.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 130 
104. Singla AK, Garg A, Aggarwal D. Paclitaxel and its formulations. Int J Pharm. 2002;

235:179Y192.

105. Jarho P, Vander Velde D, Stella VJ. Cyclodextrin-catalyzed deacetylation of

spironolactone is pH and cyclodextrin dependent. J Pharm Sci. 2000; 89:241Y249.

106. Sortino S, Giuffrida S, De Guldi G, et al. The photochemistry of flutamide and its

inclusion complex with beta-cyclodextrin: Dramatic effect of the microenvironment on

the nature and on the efficiency of the photodegradation pathways. Photochem Photobiol.

2001; 73:6Y13.

107. Matsuda H, Arima H. Cyclodextrins in transdermal and rectal delivery. Adv Drug

Deliv Rev. 1999; 36:81Y99.

108. Mielcarek J. Photochemical stability of the inclusion complexes formed by modified

1, 4-dihydropyridine derivatives with betacyclodextrin. J Pharm Biomed Anal. 1997;

15:681Y686.

109. Lutka A, Koziara J. Interaction of trimeprazine with cyclodextrins in aqueous

solution. Chem Pharm Bull (Tokyo). 2000; 57:369Y374.

110. Lutka A. Investigation of interaction of promethazine with cyclodextrins in aqueous

solution. Acta Pol Pharm. 2002; 59:45Y51.

111. Croyle MA, Cheng X, Wilson JM. Development of formulations that enhance

physical stability of viral vectors for gene therapy. Gene Ther. 2001; 8:1281Y1290.

112. Dwivedi AK, Kulkarni D, Khanna M, Singh S. Effect of cyclodextrins on the

stability of new antimalarial compound N1–31 –Acetyl/ - 41, 51 – Dihydro – 21 Furanyl

– N4–(6–Methoxy, 8-Quinolinyl)–1– 4–Pentane diamine. Ind J Pharm Sci. 1999;

61:175Y177.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 131 
113. Gerloczy A, Fonagy A, Keresztes P, Perlaky L, Szejtli J. Absorption, distribution,

excretion and metabolism of orally administered 14C β- cyclodextrin in rats. Arzneimittel

Forschung. 1985; 35(7):1042-1047.

114. Gerloczy A, Fonagy A, Szejtli J. Absorption and metabolism of β-cyclodextrin by

rats. In Szejtli J (Ed) Proceedings of 1st International Symposium on Cyclodextrins

Reidel, Dordrechi. 1982, p101-108.

115. Higuchi T, Connors KA. Phase-solubility techniques. Adva Anal Chem Instr. 1965;

4:212Y217.

116. Loftsson T, Stefánsson E. Effect of cyclodextrins on topical drug delivery to the eye.

Drug Dev Ind Pharm. 1997; 23:473Y481.

117. Van Dorne H. Interaction between cyclodextrins and ophthalmic drugs. Eur J Pharm

Biopharm. 1993; 39:133Y139.

118. Loftsson T, Másson M, Stefánsson E. Cyclodextrins as permeation enhancers.17th

Pharmaceutical Technology Conference and Exhibition; March 24-26, 1997; Dublin,

Ireland.

119. Loftsson T, Leeves N, Bjornsdottir B, Duffy L, Masson M. Effect of cyclodextrins

and polymers on triclosan availability and substantivity in toothpastes in vivo. J Pharm

Sci. 1999; 88:1254Y1258.

120. Willems L, Geest RV, de Beule K. Itraconazole oral solution and intravenous

formulations: a review of pharmacokinetics and pharmacodynamics. J Clin Pharm Ther.

2001; 26:159Y169.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 132 
121. Yang T, Hussain A, Paulson J, Abbruscato TJ, Ahsan F. Cyclodextrins in nasal

delivery of low-molecular-weight heparins: in vivo and in vitro studies. Pharm Res. 2004;

21:1127Y1136.

122. Watanabe Y, Kiriyama M, Ito R, et al. Pharmacodynamics and pharmacokinetics of

recombinant human granulocyte colony-stimulating factor (rhG-CSF) after

administration of a rectal dosage vehicle. Biol Pharm Bull. 1996; 19:1059Y1063.

123. Duchene D, Dchnreres B, Vaution C. Improvement of drug stability by

cyclodextrins inclusion complexation, STP Pharm Sci. 1987; 1:37.

124. Otero Espinar FJ, Anguiano S, Blanco MJ, Vila JJL. Reduction in the ulcerogenicity

of naproxen by complexation by complexation with β-cyclodextrin. Int J Pharm. 1991;

70:35-41

125. Lin SL, Wouessidjewe D, Poelman MC, Duchene D. In vivo evaluation of

indomethacin ./

Cyclodextrin complexes gastrointestinal tolerance and dermal anti-inflammatory activity.

Int J

Pharm. 1994; 106:63-67.

126. Zuo Z, Kwon G, Stevenson B, Diakur J, Wiebe LI. Flutamide-Hydroxy proyl- β-

cyclodextrin complex: formulation, physical characterization, and absorption studies

using the Caco-2 in vitro model. J Pharm Pharm Sci. 2000; 3:220Y227.

127. Yoo SD, Yoon BM, Lee HS, Lee KC. Increased bioavailability of clomipramine

after sublingual administration in rats. J Pharm Sci.1999; 88:1119Y1121.

128. Choi HG, Lee BJ, Han JH, et al. Terfenadine-beta-Cyclodextrin inclusion complex

with antihistaminic activity enhancement. Drug Dev Ind Pharm. 2001; 27:857Y862.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 133 
129. Veiga F, Fernandes C, Teixeira F. Oral bioavailability and hypoglycaemic activity

of tolbutamide/cyclodextrin inclusion complexes.Int J Pharm. 2000;202:165Y171.

130. Fathy M, Sheha M. In vitro and in vivo evaluation of amylobarbitone/

hydroxypropyl-β-cyclodextrin complex prepared by a freeze-drying method. Pharmazie.

2000; 55:513Y517.

131. Pitha J, Harman SM, Michel ME. Hydrophilic cyclodextrin derivatives enable

effective oral administration of steroidal hormones. J Pharm Sci. 1986; 75:165Y167.

132. Pitha J, Anaissie EJ, Uekama K. Gamma-cyclodextrin: testosterone complex

suitable for sublingual administration. J Pharm Sci. 1987; 76:788Y790.

133. Farag Badawy SI, Ghorab MM, Adeyeye CM. Bioavailability of danazol-

hydroxypropyl-á-cylodextrin complex by different routes of administration. Int J Pharm.

1996; 145:137Y143.

134. Jain AC, Aungust BJ, Adeyeye MC. Development and in vivo evaluation of buccal

tablets prepared using danazol-sulfobutylether 7 beta-cyclodextrin (SBE 7) complexes. J

Pharm Sci.

2002; 91:1659Y1668.

135. Funasaki N, Kawaguchi R, Hada S, Neya S. Ultraviolet spectroscopic estimation of

microenvironments and bitter tastes of oxyphenonium bromide in cyclodextrin solutions.

J Pharm Sci. 1999; 88:759Y762.

136. Okimoto K, Ohike A, Ibuki R, et al. Design and evaluation of an osmotic pump

tablet (OPT) for chlorpromazine using (SBE) 7m-beta-CD. Pharm Res. 1999;

16:549Y554.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 134 
137. Okimoto K, Miyake M, Ohnishi N, et al. Design and evaluation of an osmotic pump

tablet (OPT) for prednisolone, a poorly water soluble drug, using (SBE)7m-beta-CD.

Pharm Res. 1998; 15:1562Y1568.

138. Roger A, Rajewski, Valentino J. Stella. Pharmaceutical applications of cyclodextrin

2. In

vivo drug delivery. J Pharm Sci. 1996; 85(11):1024-1074.

139. Frijlink, HW, Franssen, EJF, Eissens AC, Oosting, R, Lerk CF, Meijer DKF. Effects

of cyclodextrins on the disposition of intravenously injected drugs in the rat. Pharm Res.

199; 8:380-384.

140. Kubota Y, Fukuda M, Muroguchi M, Koizumi K. Absorption, distribution and

excretion of betacyclodextrin and glucosyl-beta-cyclodextrin in rats. Bio Pharm Bull.

1996; 19(8):1068-1072.

141. Usayapant A, Karara A H, Narurkar M M. Effect of 2-hydroxypropyl-beta-

cyclodextrin on the ocular absorption of dexamethasone and dexamethasone acetate.

Pharma Res. 1991; 8:1495-

1499.

142. Loftsson T, Jarvinen T. Cyclodextrin in ophthalmic drug delivery, Adv Drug Del

Rev. 1999; 36:59-79.

143. Haydee Blanco Fluente, Blanca Esteban Fernandez, Jose Blanco Mendez, Francisco

Javier

Otero Espinar. Chem Pharm Bull. 2002; 50(1):40-46.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 135 
144. Blanchard J, Ugwu SO, Bhardwaj R, Dorr T. Anhydrous carbopol polymer gels for

the topical delivery of oxygen/water sensitive compounds. Pharm Dev Technol. 2000;

7:249Y255.

145. Hirayama F, Uekama K. Cyclodextrin-based controlled drug release system. Adv

Drug Deliv Rev. 1999; 36:125Y141.

146. Sinha VR, Nanda A, Kumria R. Cyclodextrins as sustained-release carriers.

Pharmaceutical Technology. 2002. Available at: http://www. Pharmtech.com. Accessed

May 25, 2005.

147. Uekama K, Horikawa T, Yamanaka M, Hirayama F. Peracylated β-cyclodextrins as

novel sustained-release carriers for a water-soluble drug, molsidomine. J Pharm

Pharmacol. 1994; 46:714Y717.

148. Quaglia F, Varricchio G, Miro A, La Rotonda MI, Larobina D, Mensitieri G.

Modulation of drug release from hydrogels by using cyclodextrins: the case of

nicardipine/ beta-cyclodextrin system in crosslinked polyethylenglycol. J Control

Release. 2001; 71:329Y337.

149. Fernandes CM, Teresa Viera M, Veiga FJ. Physicochemical characterization and in

vitro dissolution behavior of nicardipinecyclodextrins inclusion compounds. Eur J Pharm

Sci. 2002; 15:79Y88.

150. Fernandes CM, Ramos P, Falcao AC, Veiga FJ. Hydrophilic and hydrophobic

cyclodextrins in a new sustained release oral formulation of nicardipine: in vitro

evaluation and bioavailability studies in rabbits. J Control Release. 2003; 88:127Y134.

151. Chowdary KPR, Reddy GK. Complexes of nifedipine with β- and hydroxypropyl-β-

cyclodextrin in the design of nifedipine SR tablets. Ind J Pharm Sci. 2002; 64:142Y146.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 136 
152. Burgos AE, Belchior JC, Sinisterra RD. Controlled release of rhodium (II)

carboxylates and their association complexes with cyclodextrins from hydroxyapatite

matrix. Biomaterials. 2002; 23:2519Y2526.

153. Minami K, Hirayama F, Uekama K. Colon-specific drug delivery based on a

cyclodextrin prodrug: release behavior of biphenylylacetic acid from its cyclodextrin

conjugates in rat intestinal tracts after oral administration. J Pharm Sci. 1998;

87:715Y720.

154. Hirayama F, Ogata T, Yano H, et al. Release characteristics of a short-chain fatty

acid, n-butyric acid from its beta-cyclodextrin ester conjugate in rat biological media. J

Pharm Sci. 2000; 89:1486Y1495.

155. Lopez MEV, Reyes LN, Igea SA, Espinar FJO, Mendez JB. Formulation of

triamcinolone acetonide pellets suitable for coating and colon targeting. Int J Pharm.

1999; 79:229Y235.

156. Irie T, Uekama K. Cyclodextrins in peptide and protein delivery. Adv Drug Deliv

Rev. 1999; 36:101Y123.

157. Augustijns PF, Bradshaw TP, Gan LSL, Hendren RW, Thakker DR. Evidence for a

polarized efflux system in caco-2 cells capable of modulating cyclosporin a transport.

Biochem Biophys Res Commun. 1993; 197:360Y365.

158. Sharom FJ, Xiaohong YU. DioDiodato G, Chu JWK. Synthetic hydrophobic

peptides are substrates for P-glycoprotein and stimulate drug transport. Biochem J. 1996;

320:421Y428.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 137 
159. McNally EJ, Park JY. Peptides and Proteins - Oral Absorption. In: Swarbrick J,

Boylan JC, eds. Encyclopedia of Pharmaceutical Technology. 2nd Ed. New York, NY:

Marcell Dekker; 2002:2096Y2113.

160. Jerry N, Anitha Y, Sharma CP, Sony P. In vivo absorption studies of insulin from an

oral delivery system. Drug Deliv. 2001; 8:19Y23.

161. Dass CR. Vehicles for oligonucleotide delivery. J Pharm Pharmacol. 2002; 54:3Y27.

162. Redenti E, Pietra C, Gerlozy A, Szente L. Cyclodextrins in oligonucleotide delivery.

Adv Drug Deliv Rev. 2001; 53:235Y244.

163. Driscoll CO, Darcy R. Cyclodextrin constructs for delivery of genotherapeutic

agents. Business Briefing: Pharmatech2002. Available at:

http://www.bbriefings.com/pdf/17/ACF9C6C.pdf. Accessed May 25, 2005.

164. Hwang SJ, Bellocq NC, Davis ME. Effects of structure of β-cyclodextrin-containing

polymers on gene delivery. Bioconjugate Chem. 2001; 12:280Y290.

165. Pun SH, Davis DE. Development of a nonviral gene delivery vehicle for systemic

application. Bioconjugate Chem. 2002; 13:630Y639.

166. Croyle MA, Roessler BJ, Hsu CP, Sun R, Amidon GL. Beta cyclodextrins enhance

adenoviral-mediated gene delivery to the intestine. Pharm Res. 1998; 15:1348Y1355.

167. Buschmann HJ, Schollmeyer E. Applications of cyclodextrins in cosmetic products:

a review. J Cosmet Sci. 2002; 53:185Y191.

168. Brewster ME, Loftsson T. The use of chemically modified cyclodextrins in the

development of formulations for chemical delivery systems. Pharmazie. 2002;

57:94Y101.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 138 
169. Wu WM, Wu J, Bodor N. Effect of 2-hydroxypropyl-betacyclodextrin on the

solubility, stability, and pharmacological activity of the chemical delivery system of TRH

analogs. Pharmazie. 2002; 57:130Y134.

170. Arima H, Yunomae K, Hirayama F, Uekama K. Contribution of P-glycoprotein to

the enhancing effects of dimethyl-β-cyclodextrin on oral bioavailability of Tacrolimus. J

Pharm Exp Therapeutics. 2001; 297:547Y555.

171. McCormack B, Gregoriadis G. Entrapment of cyclodextrin-drug complexes into

liposomes: potential advantages in drug delivery. J Drug Target. 1994; 2:449Y454.

172. McCormack B, Gregoriadis G. Drugs-in-cyclodextrins-in-liposomes: an approach to

controlling the fate of water insoluble drugs in vivo. Int J Pharm. 1998; 162:59Y69.

173. McCormack B, Gregoriadis G. Drugs-in-cyclodextrins-in liposomes: a novel

concept in drug delivery. Int J Pharm. 1994; 112:249Y258.

174. Duchene D, Ponchel G, Wouessidjewe D. Cyclodextrins in targeting. Application to

nanoparticles. Adv Drug Del Rev. 1999; 36:29Y40.

175. McCormack B, Gregoriadis G. Comparative studies of the fate of free and liposome-

entrapped hydroxypropyl-/3-cyclodextrin/drug complexes after intravenous injection into

rats: implications in drug delivery. Biochim Biophys Acta. 1996; 1291:237Y244.

176. Bibby DC, Davies NM, Tucker IG. Investigations into the structure and composition

of beta-cyclodextrin/poly (acrylic acid) microspheres. Int J Pharm. 1999; 180:161Y168.

177. Memisoglu E, Bochot A, Sen M, Duchene D, Hıncal AA. Non-surfactant

nanospheres of progesterone inclusion complexes with amphiphilic β-cyclodextrins. Int J

Pharm. 2003; 251:143Y153.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 139 
178. Monza da Silveira A, Ponchel G, Puisieux F, Duchene D. Combined poly

(isobutylcyanoacrylate) and cyclodextrins nanoparticles for enhancing the encapsulation

of lipophilic drugs. Pharm Res. 1998; 15:1051Y1055.

179. Duchene D, Ponchel G, Wouessidjewe D. Cyclodextrins in targeting Application to

nanoparticles. Adv Drug Deliv Rev. 1999; 36:29Y40.

180. Boudad H, Legrand P, Lebas G, Cheron M, Duchene D, Ponchel G. Combined

hydroxypropyl-beta-cyclodextrin and poly (alkylcyanoacrylate) nanoparticles intended

for oral administration of saquinavir. Int J Pharm. 2001; 218:113Y124.

181. Memisoglu E, Bochot A, Sen M, Charon D, Duchene D, Hincal AA. Amphiphilic

beta-cyclodextrins modified on the primary face: synthesis, characterization, and

evaluation of their potential as novel excipients in the preparation of nanocapsules. J

Pharm Sci. 2002; 91:1214Y1224.

182. Memisoglu E, Bochot A, Ozalp M, Sen M, Duchene D, Hincal AA. Direct

formation of nanospheres from amphiphilic beta-cyclodextrin inclusion complexes.

Pharm Res. 2003; 20:117Y125.

183. Gèze A, Aous S, Baussanne I, Putaux JL, Defaye J, Wouessidjewe D. Influence of

chemical structure of amphiphilic β-cyclodextrins on their ability to form stable

nanoparticles. Int J Pharm. 2002; 242:301Y305.

184. Shangraw RF, Pande GS, Gala P. Charactarisation of the tabletting properties of

beta cyclodextrin: the effects of processing variableson the inclusion complex formation,

compactability and dissolution. Drug Dev Ind Pharm. 1992; 18:1831Y1851.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 140 
185. Tsai T, Wu JS, Ho HO, Sheu MT. Modification of physical characteristics of

microcrystalline cellulose by codrying with betacyclodextrins. J Pharm Sci. 1998;

87:117Y122.

186. Li WD, Huang JC, Corke H. Effect of beta-cyclodextrin on pasting properties of

wheat starch. Nahrung. 2000; 44:164Y167.

187. Gazzaniga A, Sangalli ME, Bruni G, Zema L, Vecchio C, Giordano F. The use of

beta-cyclodextrin as a pelletization agent in the extrusion/spheronization process. Drug

Dev Ind Pha

188. Proniuk S, Blanchard J. Influence of degree of substitution of cyclodextrins on their

colligative properties in solution. J Pharm Sci. 2001; 90:1086Y1090.

189. Loftsson T, Stefansdottir O, Friariksdottir H, Guomundsson O. Interaction between

preservatives and 2-hydroxypropyl β-cyclodextrin. Drug Dev Ind Pharm. 1992;

18:1477Y1484.

rm. 1998; 24:869Y873.

190. Cramer F, Saenger W, and Spatz H., J.Am.Chem.S0c., 1967;89;14.

191. Bekers O. et al., Drug Dev.Ind. Pharm., 1991; 17; 1503.

192. Higuchi T. and Connors K A, Phase-solubility Techniques, In: Advances in

Analytical Chemistry and Instrumentation, Reilly, C.N., Ed.., Wiley-Interscience. New

York. 1965; 4; 117.

193. Patel V P, Patel N M, Chaudhari B G. Effect of water soluble polymers on

dissolution profile of glipizide cyclodextrins complex. Indian Drugs. 45(1), Jan 2008, 31-

36.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 141 
194. Doijad RC, Kanakal MM, Manvi FV. Effect of processing variables on dissolution

and solubility of piroxicam: Hydroxy-propyl-β-cyclodextrin inclusion complexes. Ind.J.

Pharm. Sci. March-April 2007, 323-326.

195. Seoung Wook Jun, Min-Soo Kim, Jeong-Soo Kim., et.al. Preparation and

characterization of simvastatin/HP-β-CD inclusion complex using supercritical

antisolvent (SAS) process. European Journal of Pharmaceutics and Biopharmaceutics. 66,

2007, 413-421.

196. Deshmukh SS, Potnis VV, Shelar DB. Studies on inclusion complexes of

ziprasidone hydrochloride with β-CD and HP-β-CD. Indian drugs. 44(9), 2007, 677-682.

197. Tayede PT, Vavia PR. Inclusion complexes of ketoprofen with β- Cyclodextrins:

Oral Pharmacokinetics of ketoprofen in human. Indian J. Pharm. Sci. 68(2), 2006, 164-

170.

198.Chowdary KPR, Vijay Srinivas. Effect of polyvinyl pyrrolidone on complexation

and dissolution rate of β and hydroxypropyl-β-cyclodextrins complexes of celecoxib.

Ind.J. Pharm. Sci. Sept-oct. 2006, 631-634.

199. Derle Deelip, Boddu SS, Magar M. Studies on the preparation, characterization and

solubility of β-cyclodextrin–Satranidazole Inclusion complexes. Indian J. Pharm. Educ.

Res. 40 (4), Oct-Dec 2006, 232-236.

200. Monique WJ Den Brok, Sabien C.van der Schoot., et.al. 2-HP-β-CD extracts 2-

phenylphenol from silicone tubing. International journal of pharmaceutics. 278, 2004,

303-309

2001. Nagesh Bandi, William Wei, Christopher B Roberts, et.al. Preparation of

budesonide and indomethacin-hydroxypropyl-β-cyclodextrin complexes using a single

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 142 
step, organic solvent free super-critical fluid process. Eur.J.Pharm.Sci. 23, 2004, 159-

168.

202 Sunil Jambhekar, Casella R, Maher T. The physicochemical characteristics and

bioavailability of indomethacin from β-cyclodextrins, hydroxyl-ethyl β-cyclodextrin and

hydroxypropyl β-cyclodextrins complexes. Inter. J. Pharm. 270, 2004, 149-166.

203.Indap MA, Sunita C Bhosle, Tayade PT and Vavia PR. Evaluation of toxicity and

antitumor effect of a hydroxypropyl β-cyclodextrins inclusion complex of quercetin. Ind.

J. of Pharm. Sci. 64(4), Aug 2002, 349-353.

204.Mourya V K, Saini T R, Jadhav G S. et al. Molecular inclusion of sparfloxacin and

hydroxypropyl β-cyclodextrin. Indian J. of Pharm. Sci. 64 (6), 2002, 568-572.

205 Tayade NG, Nagarsenker MS. Preparation, characterization and evaluation of

hydroxypropyl β-cyclodextrins inclusion complexes of antimalarial drug. Scientific

Abstract 54th IPC Pune, Sept. 2002, 156.

206. Raida S Al-Kassas, Omaimah MN, Al-Gohary, et.al. Controlling of systemic

absorption of Gliclazide through incorporation into alginate beads. Int. J. Of Pharm. 341,

2007, 230-237.

207. Biswal S, Sahoo J, Murthy PN. Characterization of Gliclazide - PEG 8000 Solid

dispersions. Tropical J. of Pharm. Research. 8 (5), Oct 2009, 417-424.

208. Hussien Allaboun, Khouloud A Alkhamis, Wafa Y Almomani. The application of

the convective diffusion model and the film equilibrium model to surfactant facilated

dissolution of Gliclazide. Eur. J. of Pharm. Sci. 19, 2003, 231-236.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 143 
209. Roya Talari, Jaleh Varshosaz, Ali Nakhodchi. Dissolution enhancement of

Gliclazide using pH change approach in presence of twelve stabilizers with various

physic-chemical properties. J. Pharm. Pharmaceut. Sci. 12 (3), 2009, 250-265.

210. Gopal V S, Averineni R K, Yogendra N U., et.al. Enhanced dissolution and

bioavailability of Gliclazide using solid dispersion technique. Int. J. of Drug Delivery. 2,

2010, 49-57.

Department of Industrial Pharmacy, KRES’s KCP, BIDAR  Page 144 
ERRATA

PAGE NO. PRINTED AS READ AS

Das könnte Ihnen auch gefallen