You are on page 1of 212

 

FORMULATION AND EVALUATION OF FAST DISSOLVING


TABLETS OF KETOPROFEN
Submitted by

MAHAJAN ANIL ARUN

Reg. No: 09PU178

Dissertation submitted to
Rajiv Gandhi University of Health Sciences,
Karnataka, Bangalore.

In partial fulfillment of
the requirements for the award of the degree of

MASTER OF PHARMACY
IN
PHARMACEUTICS

Under the guidance of

Mr. R. RAMESH
M.Pharm.
Professor & Head of the Department

DEPARTMENT OF PHARMACEUTICS
Dr.H.L.T.COLLEGE OF PHARMACY
KENGAL, CHANNAPATANA.
2011
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,

KARNATAKA, BANGALORE.

DECLARATION BY THE CANDIDATE

I hereby declare that this dissertation entitled “FORMULATION AND

EVALUATION OF FAST DISSOLVING TABLETS OF KETOPROFEN” is a

bonafide and genuine research work carried out by me under the guidance of

Mr. R. RAMESH, Professor.

Date: Signature of the candidate

Place: Channapatna MAHAJAN ANIL ARUN

ii 

 
Dr. H.L.T. COLLEGE OF PHARMACY
KENGAL, CHANNAPATNA - 571502

CERTIFICATE BY THE GUIDE

This is to certify that the dissertation entitled “FORMULATION AND

EVALUATION OF FAST DISSOLVING TABLETS OF KETOPROFEN” is a

bonafide research work done by MAHAJAN ANIL ARUN in partial fulfillment

of the requirement for the degree of Master of Pharmacy in Pharmaceutics.

Date: Mr. R. RAMESH


M. Pharm.
Place: Channapatana Department of Pharmaceutics
Dr. H.L.T. College of pharmacy
Kengal, Channapatana.

iii 

 
Dr. H.L.T. COLLEGE OF PHARMACY
KENGAL, CHANNAPATNA - 571502

ENDORSEMENT BY THE H.O.D., PRINCIPAL/HEAD OF


THE INSTITUTION

This is to certify that the dissertation entitled “FORMULATION AND


EVALUATION OF FAST DISSOLVING TABLETS OF KETOPROFEN” is a
bonafide research work done by MAHAJAN ANIL ARUN under the guidance of
Mr. R. RAMESH, Professor, Department of Pharmaceutics, Dr. H.L.T. College
of Pharmacy, Channapatna.
 

Mr. R.RAMESH Mr. R.RAMESH


M. Pharm. M. Pharm.
Head of the Department Professor and Principal,
Department of Pharmaceutics Dr. H.L.T College of Pharmacy
Dr. H.L.T College of Pharmacy Kengal, Channapatna
Kengal, Channapatna

Date: Date:

Place : Place:

iv 

 
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
KARNATAKA, BANGALORE.

Copyright

Declaration by the Candidate

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

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

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

purpose.

Date: Signature of the candidate


Place: Channapatna MAHAJAN ANIL ARUN

© Rajiv Gandhi University of Health Sciences, Karnataka.

 
ACKNOWLEDGEMENT

I offer my adoration to God Almighty who created me, gave me the strength
and courage to complete my dissertation and given me the opportunity to thanks all
those people through whom this Grace was delivered to me.

I take it as a privilege to sincerely express my deep sense of gratitude and


thank my guide, Mr. R. Ramesh, Professor & Principal Department of
Pharmaceutics, Dr. H.L.T. College of Pharmacy, whose meticulous guidance,
valuable suggestions and constant motivation enabled me to complete this
dissertation.

I express my heartfelt thank to Mr. S. Murlidhar, Associate Professor,


Department of Pharmaceutics, Dr. H.L.T. College of Pharmacy, whose inspiring
support, valuable suggestions, scholarly remarks and guidance made me to complete
this project successfully.

It is my pleasure to thank Dr.T.V.Narayana, Chairman of Dr. H.L.T. College


of Pharmacy dynamic approach towards students made us easy feeling during the
course of study.

I take this opportunity to thanks Dr. Ashok Kumar D., Professor, Department
of Pharmacognosy, and Mr. Gurumurthy M., Associate Professor, Mr. Syed Azhar
Nizami, Associate Professor and Mr. Kumara Prasad S. A., Assistant Professor,
Department of Pharmaceutical Chemistry, Dr. H.L.T. College of Pharmacy for their
valuable guidance, suggestions and support during my M.Pharm course.

It’s a great pleasure to utilize this unique opportunity to express my deep


sense of gratitude and offer my most sincere and humble regards to Dr.P.P.Thampi &
Mrs. Sarala Thampi Professors, for their continuous encouragement and support in
completion of my Course and Dissertation successfully.

I would like to express my sincere and heartfelt thanks to Mrs. Benny Beby,
Mrs. Usha balkeshav Lecturers, K.C.P. College of Pharmacy, Bangalore, for their
kind help who were, and still are my guiding light and support during my course of
Pharmacy education and profession.

I would like to take this opportunity to express my heartfelt gratitude and


thank sincerely Mr. Vishwanath Pawar senior scientist, Zydus Cadila, Ahmadabad,

vi 
 
for his valuable suggestions and constant guidance in the formulation aspects, which
gave a proper shape to my research work.

I would like to thanks Mr. Sandip for providing me gift sample of Ketoprofen
and Mr. Ajani for providing me gift samples of excipients.

I am thankful for utilizing the services of Library and Information Centre of


our college. I am thankful to Mrs. Susheela Librarian and Mr. Umesh, Asst.
Librarian for their help in all the work starting from book references to literature
survey.

I am also thankful Mr. Gangadhar, Mr. Srinivas, Mrs. B. Pramila and to


other non-teaching staff for their kind co-operation. Further, I heartily thankful to
Mr. Adinarayan, & Parmesh Lab. Technician, Mr. Shankar and Mr. Prakash for
timely help.

I would like to thank my seniors, my juniors Nawaz, Kranthiteja, Pragnesh,


Kiran and others for their co-operation and help.

My special thanks to my classmates Naresh, Rakesh, Aakif, Sachin, Jaypal,


Balvenkat Reddy Mamata, Vidyasagar, Anil, Geethanjali, Rajarao, and Sarath for
their understanding, cooperation and encouragement in all my endeavors.

I would also like to thank my friends Naresh, Amol, Vinod, Sandeep, Yogesh,
Shaila, Savitri, Dhanajay, Sunil, Imran, Mangesh, Pravin, Jaydeep, Mahesh,
Tushar, Ajay and others who are directly or indirectly involved in motivating,
encouraging and supporting me all through my career.

I am blessed indeed to have such caring and loving parents, my mother


Smt. Kamal Mahajan, my father Sri. Arun Mahajan, my grandmother
Smt. Tulasabai Mahajan and elders throughout my get-up-and-go and it is those to
whom I like to dedicate this thesis.

Words will fall short to express my feelings for my father who is a very simple,
down to earth and a faultless person. He taught me the biggest lesson of life that is
hard work, honesty and self confidence. He has been the first person to tell me all I
needed to know about the value of higher education in becoming a better person by
his simple eloquence. Education commences at the mother’s knee, my Mamma is the
soul of my energy. Her blessings have always led me to tag along the right path. I

vii 
 
cannot repay her silent patience and constant encouragement. She stood by me
whenever I felt let down, pepped me up when my spirit dampened.

With all my love and affection, I am indebted to my parents, and all my family
members, my sisters Mrs. Pratibha, Mrs. Manisha and my jiju Mr. Ravindra Patil,
Mr. Dipak Choudhari and my uncle Mr.Gajanan Sonawane for their constant
encouragement during my entire career.

I also express my hearty thanks to Central Bank of India, Erondol branch for
providing me education loan for my study.

I again thank all those who were involved in my accomplishments directly or


indirectly.

Date:

Place: (MAHAJAN ANIL ARUN)

viii 
 
 

 
ABBREVIATIONS USED
Abbreviation Full Form

CA : Citric Acid
cm : Centimeter (Unit of length)
ρ : Density
0
C : Degree Centigrade (Unit of Temperature)
DSC : Differential Scanning Calorimetry
e.g. : For example
FDT : Fast Dissolving Tablets
FT-IR : Fourier Transform Infrared
g : Gram (Unit of weight)
h : Hour (Unit of time)
IP : Indian Pharmacopoeia
IR : Infrared
kg : Kilogram (Unit of weight)
et al. : Latin et alii (and others)
M : Mass
µg : Microgram
mg : Milligram (Unit of weight)
min : Minute (Unit of time)
ml : Milliliter (Unit of volume)
PM : Physical Mixture
% : Percentage
Sec : Second (Unit of time)
SBC : Sodium Bicarbonate
SD : Solid Dispersion
SEM : Scanning Electron Microscopy
SSG : Sodium Starch Glycolate
USP : United States Pharmacopoeia
UV : Ultraviolet
V : Volume
w/v : Weight by Volume
w/w : Weight by Weight
XRD : X-Ray Diffraction

ix 
ABSTRACT:

Ketoprofen is non-steroidal anti-inflammatory drug; mainly used for osteoarthritis

and rheumatoid arthritis. The major problem with this drug is its very low solubility in

biological fluids; which results in poor solubility after oral administration. Therefore solid

dispersion of Ketoprofen with PEG-6000 and PVP K30 in different weight ratios (1:1, 1:2,

1:3) were prepared with a view to increase its water solubility. The solid dispersions were

evaluated by solubility study, drug content, in-vitro drug release study, dissolution efficiency

and characterized by FT-IR, DSC, XRD and surface morphology by SEM. The Ketoprofen

SD with PVP K30 (1:3) ratio showed maximum amount of drug release hence it selected for

Fast Dissolving Tablets formulation. The Fast Dissolving Tablets of Ketoprofen was prepared

by direct compression technique by addition of superdisintigrant like Sodium starch glycolate,

Crosscarmalose sodium, and Crospovidone in different concentration (1-5% w/w) and by

effervescence technology by using combination of (2:3 ratio) Citric acid and sodium

bicarbonate in different concentration (1-5% w/w) with a view to enhance the patient

compliance. Directly compressible dextrose was used to enhance the mouth feel. The

prepared batches of tablets was evaluated for hardness, friability, disintegration time, wetting

time, dispersion time, drug content uniformity and in-vitro drug release in 6.8 pH Sorenson’s

buffer measured at 260 nm. Among all formulations, F15 containing 5% w/w of

Crospovidone is best having least disintegration time 25.68 seconds and release 99.55% of

drug in 20 minutes. The formulation F4, F10, F18 and best formulation F15 was selected for

accelerated stability study at 400 C (75% RH) for period of 3 months. Stability study confirms

there is no significantly change in hardness, friability, disintegration time, drug content and

in-vitro drug release pattern.

Keywords: Ketoprofen, Solid dispersion, Direct compression, Fast dissolving Tablets,

Superdisintigrants, Effervescent method, Stability Study.

 
CONTENTS

S. NO. PARTICULARS PAGE NO.

1 INTRODUCTION 1-44

2 AIM AND OBJECTIVES 45-47

3 REVIEW OF LITERATURE 48-100

4 METHODOLOGY 101-122

5 RESULTS 123-165

6 DISCUSSION 166-176

7 CONCLUSIONS 177-179

8 SUMMARY 180-181

9 REFERENCES 182-194

10 ANNEXURES 195-196

xi
LIST OF FIGURES

Figure No. Title Page No.


A schematic representation of the bioavailability
1 enhancement of a poorly water-soluble drug by solid 4
dispersion compared with conventional tablet or capsule
2 Disintegration of tablet by wicking and swelling 31
3 Disintegration by deformation and repulsion 33
Various Technologies Used to Manufacture Fast Dissolving
4 34
Tablet
5 Step Involved In Sublimation Process 37
6 Mechanism of Action of Ketoprofen 69
7 Modified Device Used to Determine Disintegration Time 116
8 In vitro Wetting Property 117
9 In Vitro Disintegration Property 117
10 IR Spectra (A) Ketoprofen IP Spectra 124
11 IR Spectra (B) Sample of Ketoprofen 124
12 Differential Scanning Calorimetry Curve of Ketoprofen 125
13 Scan Graph of Ketoprofen 126
Calibration Curve of Ketoprofen in Sorenson’s Buffer (pH
14 128
6.8)
15 IR Spectra of Croscarmallose sodium 129
16 IR Spectra of mixture of Drug and Croscarmallose sodium 129
17 IR Spectra of Sodium Starch Glycolate 129
18 IR Spectra of mixture of Drug and Sodium Starch Glycolate 130
19 IR Spectra of Crospovidone 130
20 IR Spectra of mixture of Drug and Crospovidone 130
Cumulative Percent Release of Ketoprofen from Physical
21 Mixtures of Ketoprofen-PVP K-30 and Ketoprofen-PEG- 135
6000 Systems
Cumulative Percent Release of Ketoprofen from Solid
22 Dispersions of Ketoprofen-PVP K-30 and Ketoprofen-PEG- 135
6000 Systems
23 FT-IR Spectra of PVP K 30. 136
24 FT-IR Spectra of Ketoprofen and PVP K 30. 137
25 FT-IR Spectra of PEG-6000 137
26 FT-IR Spectra of Ketoprofen and PEG-6000. 138
27 DSC of PEG-6000 139
28 DSC of PVP K-30 139
29 DSC of KPVP-3 140
30 DSC of KPEG-3 140

xiv 
31 X-Ray Diffraction Curves of Ketoprofen 141
32 X-Ray Diffraction Curves of PVP K-30 141
33 X-Ray Diffraction Curves of PEG-6000 142
34 X-Ray Diffraction Curves of KPVP3 142
35 X-Ray Diffraction Curves of KPEG3 143
36 Scanning Electron Photomicrograph of Ketoprofen 143
37 Scanning Electron Photomicrograph of PVP K30 144
38 Scanning Electron Photomicrograph of KPVP3 144
Effect of Concentration of Superdisintegrant and
39 148
Effervescent Agent on Disintegration Time
Effect of Concentration of Superdisintegrant and
40 148
Effervescent Agent on Dispersion Time
Effect of Concentration of Superdisintegrant and
41 148
Effervescent Agent on Wetting Time
Zero Order Dissolution Release Profile of Ketoprofen from
42 152
F1-F5
Zero Order Dissolution Release Profile of Ketoprofen from
43 152
F6-F10
Zero Order Dissolution Release Profile of Ketoprofen from
44 153
F11-F15
Zero Order Dissolution Release Profile of Ketoprofen from
45 153
F15-F20
46 First Order Release Profile of Ketoprofen from F1-F5 156
47 First Order Release Profile of Ketoprofen from F6-F10 156
48 First Order Release Profile of Ketoprofen from F11-F15 157
49 First Order Release Profile of Ketoprofen from F15-F20 157
In-vitro release profile of F4 during Stability studies at
50 163
(40°C/75% RH)
In-vitro release profile of F10 during Stability studies at
51 164
(40°C/75% RH)
In-vitro release profile of F15 during Stability studies at
52 164
(40°C/75% RH)
In-vitro release profile of F18 during Stability studies at
53 165
(40°C/75% RH)

xv 
LIST OF TABLES

Table
Title Page No.
No.
1. Materials used as carrier for solid dispersion 11
Various commercially available superdisintegrants along with
2. 39
their properties.
3. Comparison of some patented technologies for FDT. 44
4. Typical Properties of Croscarmellose Sodium 73
5. Typical Properties of Sodium Starch Glycolate 75
6. Typical Properties of Crosspovidone 76
7. List of Materials 101
8. List of Equipment Used 102
9. Composition of Ketoprofen-PVP K-30 Physical Mixture 105
10. Composition of Ketoprofen-PEG-6000 Physical Mixture 105
11. Composition of Ketoprofen-PVP K-30 Solid Dispersions 106
12. Composition of Ketoprofen-PEG-6000 Solid Dispersions 106
Formulation of Fast Dissolving Tablet Using Croscarmllose
13. 109
sodium
Formulation of Fast Dissolving Tablet Using Sodium Starch
14. 110
Glycolate
15. Formulation of Fast Dissolving Tablet Using Crospovidone 110
Formulation of Fast Dissolving Drug Free Tablet Using
16. 111
Effervescent Agents
Compressibility Index as an Indication of Powder Flow
17. 112
Properties
18. Angle of Repose as an Indication of Powder Flow Properties 113
19. Weight Variation Limits for Tablets as per IP 114
20 Storage conditions according to ICH guidelines 121
21 Melting point of Ketoprofen 123
22 Solubility of Ketoprofen in Different Solvents 127
23 Solubility of Ketoprofen in Various Buffer Solutions 127
Calibration curve data of Ketoprofen in Sorenson’s Buffer (pH
24 128
6.8)
25 Interpretation of drug polymer interaction study. 131
Solubility data of Ketoprofen, physical mixture and solid
26 132
dispersion in Sorenson’s buffer ph 6.8 at 25oc and 370c
Percent Drug Content of Ketoprofen-PVP K-30 & Ketoprofen-
27. 133
PEG-6000 Physical Mixtures and Solid Dispersions
Dissolution Release Profile of Ketoprofen from Physical
28 134
Mixture

xii 
29 Dissolution release profile of Ketoprofen from solid dispersion 134

30 Dissolution Efficiency of Ketoprofen-PVP K30 &/ Ketoprofen 136


-PEG 6000 Physical Mixtures and Solid Dispersions.
31 Interpretation of drug polymer interaction study 138
32 Characterization of Blends 145
33 Characterization of Fast Dissolving Tablets 146
34 Characterization of Fast Dissolving Tablets 147
35 Drug content in the Fast Dissolving Tablets of Ketoprofen 149
Zero Order Dissolution Release Profile of Ketoprofen from
36 150
F1-F5
Zero Order Dissolution Release Profile of Ketoprofen from
37 150
F6-F10
Zero Order Dissolution Release Profile of Ketoprofen from
38 151
F11-F15
Zero Order Dissolution Release Profile of Ketoprofen from
39 151
F15-F20
40 First Order Release Profile of Ketoprofen from F1-F5 154
41 First Order Release Profile of Ketoprofen from F6-F10 154
42 First Order Release Profile of Ketoprofen from F11-F15 155
43 First Order Release Profile of Ketoprofen from F16-F20 155
Fit of various kinetics Models for fast dissolving Tablets of
44 158
Ketoprofen
Effect of Storage Condition (40°C/75%RH) on Weight of the
45 159
Ketoprofen Tablets
Effect of Storage Condition(40°C/75%RH) on Hardness of
46 159
the Ketoprofen Tablets
Effect of Storage Condition(40°C/75%RH) on Friability of
47 160
the Ketoprofen Tablets
Effect of Storage Condition(40°C/75%RH) on Disintegration
48 160
Time of the Ketoprofen Tablets
Effect of Storage Condition(40°C/75%RH) on Drug Content
49 161
of the Ketoprofen Tablets
In-vitro release profile of F18 during Stability studies at
50 161
(40°C/75%RH)
In-vitro release profile of F18 during Stability studies at
51 162
(40°C/75%RH)
In-vitro release profile of F18 during Stability studies at
52 162
(40°C/75%RH)
In-vitro release profile of F18 during Stability studies at
53 163
(40°C/75%RH)

xiii 
INTRODUCTION

CHAPTER-1.

INTRODUCTION

Oral route has been one of the most popular routes of drug delivery due to its

ease of administration, patient compliance, least sterility constraints and flexible

design of dosage forms.

For many decades treatment of an acute disease or chronic illness has mostly

accomplished by delivery of drugs to patients using conventional drug delivery

system. Even today these conventional drug delivery systems are the primary

pharmaceutical products commonly seen in the prescription. Conventional oral drug

products are formulated to release the active principle immediately after oral

administration to obtain rapid and complete systemic drug absorption.

Drug absorption is defined as the process of movement of unchanged drug

from the site of administration to systemic circulation1.

Systemic drug absorption from a drug product consists of a succession of rate

process for solid oral, immediate release drug products.

The rate process include

¾ Dissolution of the drug in an aqueous environment.

¾ Absorption across cell membranes into systemic circulation.

For drugs that have very poor aqueous solubility, the rate at which the drug

dissolves (dissolution) is often the slowest step and therefore exhibits a rate limiting

effect on drug bioavailability. In contrast, for a drug that has a high aqueous solubility

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 1 


 
INTRODUCTION

the dissolution rate is rapid the rate at which the drug crosses or permeates cell

membrane is the slowest or rate limiting step.2, 3.

Together with the permeability, the solubility behavior of a drug is a key

determinant of its oral bioavailability. They have always been certain drugs for which

solubility has presented a challenge to the development of a suitable formulation for

oral administration. Examples such as griseofulvin, digoxin, phenytoin, sulphathiazole

& chloramphenicol come immediately to mind. With the recent advent of high

through put screening of potential therapeutic agents, the number of poorly soluble

drug candidates has risen sharply and the formulation of poorly soluble compounds

for oral delivery now presents one of the most frequent and greatest challenges to

formulation scientists in the pharmaceutical industry.

Consideration of the modified Noyes – Whitney equation provides some hints

as to how the dissolution rate of even very poorly soluble compounds might be

improved to minimize the limitations to oral availability.

dc AD ( C S − C )
=
dt h

Where dc/dt = rate of dissolution

A = Surface area available for dissolution.

D = Diffusion coefficient of the compound

Cs = Solubility of the compound in the dissolution medium.

C = Concentration of drug in the medium at time t

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 2 


 
INTRODUCTION

h = thickness of the diffusion boundary layer adjacent to

surface of the dissolving compound.

The main possibilities for improving dissolution according to this analysis are

to increase the surface area available for dissolution by decreasing the particle size of

the solid compound and/or by optimizing the wetting characteristics of the compound

to decrease the boundary layer thickness, to ensure sink conditions for dissolution

and, last but definitely not least, to improve the apparent solubility of the drug under

physiologically relevant conditions.

1.1. INTRODUCTION TO SOLID DISPERSION TECHNOLOGY:

The enhancement of oral bioavailability of poorly water soluble drugs remains

one of the most challenging aspects of drug development. Although salt formation,

solubilization and particle size reduction have commonly been used to increase

dissolution rate and thereby oral absorption and bioavailability of such drugs, there

are practical limitations of these techniques. The salt formation is not feasible for

neutral compounds and the synthesis of appropriate salt forms of drugs that are

weakly acidic or weakly basic may often not be practical. Even when salts can be

prepared, an increased dissolution rate in the GIT may not be achieved in many cases

because of the reconversion of salts into aggregates of their respective acid or base

forms. The solubilization of drugs in organic solvents or in aqueous media by the use

of surfactants and cosolvents leads to liquid formulations that are usually undesirable

from the viewpoints of patient acceptability and commercialization. Although particle

size reduction is commonly used to increase dissolution rate, there is a practical limit

to how much size reduction can be achieved by such commonly used methods as

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 3 


 
INTRODUCTION

controlled crystallization, grinding, etc. The use of very fine powders in a dosage

form may also be problematic because of handling difficulties and poor wettability.

In 1961, Sekiguchi and Obi developed a practical method whereby many of

the limitations with the bioavailability enhancement of poorly water-soluble drugs can

be overcome, which was termed as “Solid Dispersion”.

Figure-1: A schematic representation of the bioavailability enhancement of a

poorly water-soluble drug by solid dispersion compared with conventional tablet

or capsule

The advantage of solid dispersion compared with conventional capsule or

tablet formulations is shown schematically in figure-1. From conventional capsules

and tablets, the dissolution rate is limited by the size of the primary particles formed

after the disintegration of dosage forms. In this case, an average particle size of 5µm

is usually the lower limit, although higher particle sizes are preferred for ease of

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 4 


 
INTRODUCTION

handling, formulation and manufacturing. On the other hand, if a solid dispersion or a

solid solution is used, a portion of the drug dissolves immediately to saturate the

gastrointestinal fluid, and the excess drug precipitates out as fine colloidal particle or

oily globules of submicron size. Because of such easily promises in the bioavailability

enhancement of poorly water-soluble drugs, solid dispersion has become one of the

most active areas of research in the pharmaceutical field.

1.2. HISTORICAL BACKGROUND:

The effect of the particle size of the drugs on their dissolution rates and

biological availability was reviewed comprehensively by Fincher. For drugs whose

gastrointestinal absorption is rate limited by dissolution, reduction of the particle size

generally increases the rate of absorption and or total bioavailability. This commonly

occurs for drugs with poor water-solubility. For example, the therapeutic dose of

griseofulvin was reduced to 50% by micronization and it also produced a more

constant and reliable blood level. The commercial dose of spironolactone was also

decreased to half by just a slight reduction of particle size. Such enhancement of drug

absorption could further be increased several fold if a micronized product was used.

In 1961, a unique approach of solid dispersion to reduce the particle size and increase

rates of dissolution and absorption was first demonstrated by Sekiguchi and Obi. They

proposed the formation of a eutectic mixture of a poorly soluble drug such as

sulfathiazole with a physiologically inert, easily soluble carrier such as urea. The

eutectic mixture was prepared by melting the physical mixture of the drug and the

carrier, followed by a rapid solidification process. Upon exposure to aqueous fluids,

the active drug was expected to be released into the fluids as fine, dispersed particles

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 5 


 
INTRODUCTION

because of the fine dispersion of the drug in the solid eutectic mixture and the rapid

dissolution of the soluble matrix.

Levy and Kanig subsequently noted the possibility of using a solid solution

approach in which a drug is dispersed molecularly in a soluble carrier. In a series of

reports in 1965-66, Goldberg et al presented a detailed experimental and theoretical

discussion of advantages of solid solution over the eutectic mixture.

In 1965, Tachibana and Nakamaru reported a novel method for preparing

aqueous colloidal dispersions of β-carotene by using water-soluble polymers such as

polyvinyl pyrrolidone. They dissolved the drug and the polymer carrier in a common

solvent and then evaporated the solvent completely. A colloidal dispersion was

obtained when the coprecipitate was exposed to water.

In 1966, Mayersohn and Gibaldi demonstrated that the dissolution rate of

griseofulvin could be markedly enhanced when dispersed in polyvinyl pyrrolidone by

the same solvent method.

Chiou and Riegelman recently advocated the application of glass solution to

increase dissolution rates. They used PEG 6000 as a dispersion carrier. It is believed

that this relatively new field of pharmaceutical technique and principles will play an

important role in increasing dissolution, absorption and therapeutic efficacy of drugs

in future dosage forms. Therefore, a thorough understanding of its fast release

principles, methods of preparation, selection of suitable carriers, determination of

physical properties, limitations and disadvantages will be essential in the practical and

effective application of this approach.

In addition to absorption enhancement, the solid dispersion technique may

have numerous pharmaceutical applications which remain to be further explored. It is

possible that such a technique can be used to obtain a homogeneous distribution of a

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 6 


 
INTRODUCTION

small amount of drug at solid state, to stabilize unstable drugs, to dispense liquid or

gaseous compounds, to formulate a fast release priming dose in a sustained release

dosage form, and to formulate sustained release regimens of soluble drugs by using

poorly soluble or insoluble carriers.

1.3. DEFINITION AND TYPES OF SOLID DISPERSIONS: 4

1.3.1. Definition:

Solid dispersion technology is the science of dispersing one or more active

ingredients in an inert matrix in the solid stage in order to achieve increased

dissolution rate, sustained release of drugs, altered solid state properties, enhanced

release of drugs from ointment and suppository bases, and improved solubility and

stability.

Advantages:

¾ Rapid dissolution rate that result in an increase in the rate and extent of the

absorption of the drug and a reduction in pre-systemic metabolism. This latter

advantage may occur due to saturation of the enzyme responsible for

biotransformation of the drug as in the case of 17-β-estradiol or inhibition of the

enzyme by the carrier as in the case of morphine-tristearin dispersion. Both can lead

to lower doses of the drug.

¾ Transformation of the liquid form of the drug into a solid from. For

eg: clofibrate and benzylbenzoate can be incorporated into PEG 6000 to give a solid.

¾ Avoidance of polymorphic changes and there by bioavailability problems as in

the case of Nabilone and PVP dispersion.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 7 


 
INTRODUCTION

¾ Protection of certain drugs by PEGs. Eg: cardiac glycosides against

decomposition by saliva to allow buccal absorption.

Disadvantages:

1. Solid dispersions have stability problems.

2. Changes in crystallinity and a decrease in dissolution rate with aging.

1.3.2. Types of solid dispersions: 5

a) Simple eutectic mixture:

An eutectic mixture of a sparingly water soluble drug and a highly water

soluble carrier may be regarded thermodynamically as an intimately blended physical

mixture of its two crystalline component. The increase in surface area is mainly

responsible for increased rate of dissolution. This led to a conclusion that the increase

in dissolution was mainly due to decreased particle size.

b) Solid solutions:

Solid solutions consist of a solid solute dissolved in a solid solvent. A mixed

crystal is formed because the two components crystallize together in a homogenous

one-phase system. Hence, this system would be expected to yield much higher rates

of dissolution than simple eutectic systems.

c) Glass solution of suspension:

A glass solution is a homogenous system in which a glassy or a vitreous of the

carrier solubilizer drug molecules in its matrix. PVP dissolved in organic solvents

undergoes a transition to a glassy state upon evaporation of the solvent.

d) Compound or complex formation:

This system is characterized by complexation of two components in a binary

system during solid dispersion preparation. The availability of the drug from the

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 8 


 
INTRODUCTION

complex is dependent on the solubility dissociation constant and the intrinsic

absorption rate of the complex.

e) Amorphous precipitation:

Amorphous precipitation occurs when drug precipitates as an amorphous form

in the inert carrier. The higher energy state of the drug in this system generally

produces much greater dissolution rates than the corresponding crystalline forms of

the drug.

1.4. MECHANISM OF INCREASED DISSOLUTION RATE: 6

The enhancement in dissolution rate as a result of solid dispersion formulation,

relative to pure drug varies from as high as 400 folds to less than two-fold. Corrigan

reviewed the current understanding of the mechanism of release from solid dispersion.

The increase in dissolution rate for solid dispersion can be attributed to a number of

factors. It is very difficult to show experimentally that any one particular factor is

more important than another. The main reasons postulated for the observed

improvements in dissolution of these systems are as follows:

a) Reduction of particle size:

In case of glass, solid solution and amorphous dispersions, particle size is reduced

to a minimum level. This can result in an enhanced dissolution rate due to an increase

in both the surface area solubilization.

b) Solubilization effect:

The carrier material, as it dissolves may have a solubilization effect on the drug.

This was shown to be the case for acetaminophen and chlorpropamide in urea as well

as for numerous other drugs.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 9 


 
INTRODUCTION

c) Wettability and dispersibility:

The carrier material may also have an enhancing effect on the wettability and

dispersibility of the drug in the dissolution media. This should retard any

agglomeration or aggregation of the particles, which can slow the dissolution process.

d) Metastable Forms:

Formation of metastable dispersions with reduced lattice energy would result in

faster dissolution rates. It was found that the activation energies for dissolution for

furosemide was 17 K Cal per mol, whereas that for 1:2 furosemide: PVP coprecipitate

was only 7.3 K Cal per mol.

1.5. SELECTION OF A CARRIER:

The properties of the carrier have a major influence on the dissolution

characteristics of the dispersed drug. A carrier should meet the following criteria to be

suitable for increasing the dissolution rate of a drug-

1. Be freely water-soluble with intrinsic rapid dissolution properties.

2. Be non-toxic and pharmacologically inert.

3. Be heat stable with a low melting point for the melt method.

4. Be soluble in a variety of solvents and pass through a vitreous state upon solvent

evaporation for the solvent method.

5. Be able to preferably increase the aqueous solubility of the drug and

6. Be chemically compatible with the drug and not form a strongly bonded complex

with the drug.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 10 


 
INTRODUCTION

Table-1: Materials used as carrier for solid dispersion

1 Sugars Dextrose, Sucrose, Galactose, Sorbitol, Maltose,

. Xylitol, Mannitol, Lactose.

2 Acids Citric acid, Succinic acid.

3 Polymeric materials Povidone (PVP), Polyethylene glycol (PEG),

Hydroxypropyl methyl cellulose, Methyl cellulose,

Hydroxy ethyl cellulose, Cyclodextrin, Pectin,

Galactomannan, Hydroxy propyl cellulose.

4 Insoluble or enteric Hydroxy propyl methyl cellulose phthalate, Eudragit

Polymer L100, Eudragit S100, Eudragit RL, Eudragit RS.

5 Surfactants Polyoxyethylene stearate, renex, Poloxamer 188,

Texafor AIP, Deoxycholic acid, Tweens, Spans.

6 Miscellaneous Pentaerythritol, Pentaerythrityl tetraacetate, Urea,

Urethane, Hydroxy alkyl xanthins.

1.6. POLYMERS USED IN SOLID DISPERSIONS: 5

Polymers used in solid dispersions are as follows:

a) Polyethylene glycols (PEG):

The term polyethylene glycol refers to compounds that are obtained by reacting

ethylene glycol with ethylene oxide. PEGs whose molecular weight is above 300000

are commonly termed as polyethylene oxides.

Effect of PEG molecular weight:

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 11 


 
INTRODUCTION

The dissolution rate of pure PEG decreases with increasing molecular weight. The

dissolution rate of the drug in solid dispersion can be increased with an increase in

molecular weight of PEG. In these cases, the rate at which the polymer dissolved

dictated the rate at which the drug dissolved. Lower molecular weight PEGs melt at

37ºC in the dissolution medium prior to dissolution, further increasing the rate of

dissolution. In some drug-PEG solid dispersion systems, the rate dissolution decreases

with molecular weight upto a certain composition of the drug above which the trend

becomes irregular.

b) Polyvinyl pyrrolidone (PVP):

PVP has a molecular weight ranging from 10,000 to 700,000. It is soluble in

solvents like water, ethanol, chloroform and isopropyl alcohol. PVP is not suitable for

preparation of solid dispersions prepared by melt method because of it melts at a very

high temperature above 275ºC, where it becomes decomposed.

Effect of PVP molecular weight:

The effect of molecular weight of PVP on the rate of dissolution of a drug is more

consistent than for PEG. An increase in molecular weight of PVP will decrease the

dissolution rate of most drugs. An increase in viscosity of PVP solution due to an

increase in molecular weight decreases diffusion of drug molecules from the surface

of viscous material into the dissolution medium. Lower molecular weight PVP has a

short swelling time prior to dissolution resulting in an increase in dissolution rate of

the polymer and drug.

c) Polymers and surface active agent combinations:

The addition of surfactants to dissolution medium lowers the interfacial tension

between the drug and the dissolution medium and promote the wetting of the drug

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 12 


 
INTRODUCTION

thereby they enhance the solubility and dissolution of drugs. Ternary dispersion

systems have higher dissolution rates than binary dispersion systems.

d) Cyclodextrins:

Cyclodextrins are primarily used to enhance solubility, chemical protection, taste

masking and improved handling by the conversion of liquids into solids by

entrapment.

Oral administration of cyclodextrins:

Cyclodextrins play an important role in the bioavailability of poorly water

soluble drugs by increasing the rate and extent of dissolution of drug.

Cyclodextrins also have the advantage of:

9 Increasing the stability of the drug.

9 Release profile during gastrointestinal transit through modification of drug

release site and time profile.

9 Decreasing local tissue irritation.

9 Masking unpleasant taste

e) Phospholipids:

Phospholipids are major structural components of cell membranes.

Phosphotidylcholine was first isolated from egg yolk and brain. Its chemical name is

1,2-diacyl-in-glycero-3-phosphocholine. In phosphatidyl ethanolamine and

phosphatidyl serine, the choline moiety is replaced by ethanolamine and serine

respectively. Other phospholipids that occur in tissues include phosphotidyl

ethanolamide (PE), phosphotidyl serine (PS), and phosphotidyl glycerol (PG).

Naturally occuring lecithins contain both a saturated fatty acid and an unsaturated

fatty acids with some exceptions.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 13 


 
INTRODUCTION

1.7. METHODS OF PREPARING SOLID DISPERSIONS: 4, 5, 7.

The basic methods used to prepare solid dispersions are

1. Melting method or fusion method

2. Solvent evaporation method

3. Kneading method

4. Supercritical Fluid Process

1. Melting method

In this method physical mixture of an active agent and a water soluble carrier is

heated until it is melted. The melt is solidified rapidly in an ice bath under vigorous

stirring, pulverizing and sieving.

Rapid congealing is desirable because it results in supersaturation of the drug as a

result of entrapment of solute molecules in the solvent matrix by instantaneous

solidification. The solidification process can be achieved on stainless steel plates

attached to a cooling system to favour rapid heat loss.

Advantages

9 This method is simple and economical.

Disadvantages

9 This method is not suitable for drugs which are unstable at the fusion

temperature or evaporates at high temperatures.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 14 


 
INTRODUCTION

9 This method may produce tacky and intractable nature of the resulting

solidified melt and irregular crystallization owing to the presence of a

miscibility gap on the phase diagram for a given carrier system.

2. Solvent Evaporation Method

Tachibani and Nakumara were the first to dissolve both the drug and carrier in a

common solvent and then evaporate the solvent under vacuum to produce a solid

solution. This enabled them to produce a solid solution of the highly lipophilic β-

carotene in the highly water soluble carrier PVP.

The evaporation method was then taken up by Mayersohn and Gibaldi. By

dissolving both griseofulvin and PVP in chloroform, and then evaporating the solvent,

they were able to achieve a solid dispersion. The release rate of griseofulvin from the

solid dispersion was 5 to 11 times higher than that of micronized drug depending on

the drug/carrier ratio.

An important requisite for the manufacture of a solid dispersion using the solvent

method is that both the drug and the carrier are sufficiently soluble in the solvent. The

solvent can be removed by any one of a number of methods. Temperatures used for

solvent evaporation usually lie in the range 23-650C. The solvent can also be removed

by freeze drying or by spray drying.

It must be remembered that when an organic solvent is to be removed, small

variations in the conditions used can lead to quite large changes in product

performance. Another point to consider is the importance of thoroughly removing all

of the solvents, since most of the organic solvents used have toxicity issues.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 15 


 
INTRODUCTION

Advantages

9 The thermal decomposition of drugs and carriers avoided which can be

associated with the fusion method.

Disadvantages

9 This method has higher cost of preparation.

9 In this there is a use of large quantities of solvent.

3. Kneading method

In this method both drug and polymer was taken in a glass mortar and triturated

by using a small volume of organic solvent to give a thick paste which was kneaded

up to 30 minutes and then kept for air dry. Then the dried mass was scratched and

pulverized and sifted through mesh # 80.

4. Supercritical Fluid Process:

Supercritical CO2 is a good solvent for water insoluble as well as water soluble

compounds under suitable conditions of temperature and pressure. Therefore,

supercritical CO2 has potential as an alternative for conventional organic solvents

used in solvent based processes for forming solid dispersions due to its favourable

properties of being nontoxic and inexpensive. The process developed by Ferro

Corporation consists of the following steps:

™ Charging the bioactive material and suitable polymer into the autoclave.

™ Addition of supercritical CO2 under precise conditions of temperature and

pressure, that causes polymer to swell;

™ Mechanical stirring in the autoclave; and

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 16 


 
INTRODUCTION

™ Rapid depressurization of the autoclave vessel through a computer controlled

orifice to obtain desire particle size. The temperature conditions used in this process

are fairly mild (35–75°C), which allows handling of heat sensitive biomolecules, such

as enzymes and proteins.

™ Solid dispersion of cabamazepine-PEG8000 has been obtained by this method.

1.8 EVALUATION OF SOLID DISPERSIONS: 5

Various methods are available which can give information regarding the

physical nature of solid dispersion system. The commonly used methods are the

following-

1. Thermal Methods:

This method is most commonly used to observe the Physico-chemical

interactions of two or more compounds. It utilizes the principle of change of thermal

energy as a function of temperature and can be performed by the following

techniques.

• Cooling Curve Method:

Physical mixtures of components in various properties are heated to a

homogenous melt. During the cooling process temperature of each mixture is plotted

as a function of time. Critical temperatures are noted and plotted against composition

to provide the phase diagram.

The method is time consuming, requires relatively larger amounts of samples

and is not applicable to samples that decompose after melting. More over changes in

slope can be mixed, especially if cooling takes place rapidly.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 17 


 
INTRODUCTION

• Thaw Melt Method:

A solidified sample in a capillary tube is heated gradually and the thaw and

melting points are noted by visual observation. Since the method depends on visual

observation the results are not reproducible.

• Thermo Microscopic Method:

Physical mixtures of drug and carrier placed in a slide covered with a cover

slip and scaled with silicone grease to prevent sublimation. The mixture is heated until

it completely liquefies. After cooling, the mixture is reheated. The hae and melting

points are noted and a phase diagram is constructed.

• Differential Thermal Analysis (DTA), differential scanning calorimetry

(DSC):

It is effective for studying phase equilibrium of pure compounds as well as

their mixtures. This method is limited to compounds with high thermal stability and

low volatility. In addition to thaw and melting points, polymorphic transitions,

evaporation, sublimation, dissolvation and other types of decomposition can also be

detected by thermal analysis. The greatest advantage lies in constructing phase

diagrams of high reproducibility.

2. X-ray diffraction method (XRD):

It is very important and efficient tool in studying physical nature of solid

dispersions. In simple eutectic systems, diffraction peaks of each crystalline

compound can be found in the diffraction spectra. In substitutions solid solutions, the

lattice parameter of the solvent crystal is either increased, remains unchanged or

decreased depending on the relative size of the solute atom or molecule. In continuous

solid solutions, there is a shift from the positions of the peaks in one pure component

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 18 


 
INTRODUCTION

to those in other. In interstitial solid solutions the diffraction pattern of solvent

component may or may not change while that of the solute component disappears.

The X-ray diffraction method can be applicable in detecting compound or

complex formation. Since the spectra of lattice parameters of a complex are different

from those of pure compound.

3. Dissolution Rate Determination:

This method can be used to study degree or crystallinity in solid-solid

equilibria. This method involves comparison of in-vitro dissolution rates of solute

component from a constant surface tablet with the physical mixture of same

composition. The technique is simple to perform except that in some binary systems,

the tablet may not be constant due to the leaching of particulars into dissolution

media. It has been shown to be applicable to simulated systems of indomethacin-

PEG-6000 and sulphathiazole-urea.

4. Electro Microscopy:

Often used to get primary information of the systems and to detect amorphous

and crystalline structures.

5. Thermodynamic Methods:

The phase diagrams of eutectic and solid solution systems can be evaluated by

phase thermodynamic parameters. The knowledge of heats of fusion, entropies and

partial pressure at various compositions enables the determination of the solubility

gap below the solid-liquid equilibrium temperature.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 19 


 
INTRODUCTION

1.9. FUTURE PROSPECTS:

Despite many advantages of solid dispersion, issues related to preparation,

reproducibility, formulation, scale up and stability limited its use in commercial

dosage forms for poorly water-soluble drugs. Successful developments of solid

dispersion systems for preclinical, clinical and commercial use have been feasible in

recent years due to the availability of surface-active and self-emulsifying carriers with

relatively low melting points. The preparation of dosage forms involves the dissolving

of drug in melted carriers and the filling of the hot solutions into hard gelatin capsules

because of the simplicity of manufacturing and scale up processes, the physico-

chemical properties and, as a result, the bioavailability of solid dispersions are not

expected to change significantly during the scale up. For this reason, the popularity of

the solid dispersion system to solve difficult bioavailability issues with respect to

poorly water-soluble drugs will grow rapidly. Because the dosage form can be

developed and prepared using small amounts of drug substances in early stages of the

drug development process, the system might have an advantage over such other

commonly used bioavailability enhancement techniques as micronization of drugs and

soft gelatin encapsulation.

One major focus of future research will be the identification of new

surfaceactive and self-emulsifying carriers for solid dispersion. Only a small number

of such carriers are currently available for oral use. Some carriers that are used for

topical application of drug only may be qualified for oral use by conducting

appropriate toxicological testing. One limitation in the development of solid

dispersion systems may the inadequate drug solubility in carrier, so a wider choice of

carriers will increase the success of dosage form development. Research should also

be directed toward identification of vehicles or excipients that would retard or prevent

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 20 


 
INTRODUCTION

crystallization of drugs from super-saturated systems. Attention must be given to any

physiological and pharmacological effects of carriers used. Many of the surfaceactive

and self-emulsifying carriers are lipidic in nature, so potential roles of such carriers on

drug absorption, especially on their inhibitory effects on CYP-3 based drug

metabolism and p-glycoprotein-mediated drug efflux will require careful

consideration.

In addition to bioavailability enhancement, much recent research on solid

dispersion systems was directed toward the development of extended-release dosage

forms. Physical and chemical stability of both the drug and the carrier in a solid

dispersion are major developmental issues, an exemplified by the recent withdrawal

of ritonavir capsules from the market, so future research needs to be directed to

address various stability issues. The semisolid and waxy nature of solid dispersions

poses unique stability problems that might not be seen in other types of solid dosage

forms. Predictive methods will be necessary for the investigation of any potential

crystallization of drugs and its impact on dissolution and bioavailability, possible

drug-carrier interactions must also be investigated.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 21 


 
INTRODUCTION

1.10. INTRODUCTION FAST DISSOLVING TABLETS

Recent advances in Novel Drug Delivery System (NDDS) aims to enhance

safety and efficacy of already used drug molecule by formulating a convenient dosage

forms for administration and to achieve better patient compliance. To develop a

chemical entity, a lot of money, hard work and time are required. So focus is rather

being laid on the development of new drug delivery systems for already existing

drugs, with enhanced efficacy and bioavailability, thus reducing the dose and dosing

frequency to minimize the side effects. 8

The oral route of administration is the most preferred route due to its many

advantages like ease of administration, accurate dosage, self-medication, pain

avoidance, versatility and patient compliance. 9 The most popular dosage forms being

tablets and capsules, one important drawback of these dosage forms however is the

difficulty to swallow. 10

It is estimated that 50% of the population is affected by this problem which

results in a high incidence of non-compliance and ineffective therapy. The difficulty

is experienced in particular by pediatric and geriatric patients, but it also applies to

people who are ill in bed and to those active working patients who are busy or

traveling, especially those who have no access to water and also in following

conditions like: Parkinsonism, Motion sickness, Unconsciousness and Mentally

disabled persons. 11

To fulfill these medical needs, the pharmaceutical technologists have

developed a novel type of dosage form for oral administration, the Fast Dissolving

Tablets (FDT), tablets that disintegrate and dissolve rapidly in saliva without water.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 22 


 
INTRODUCTION

1.11. FAST DISSOLVING TABLETS:

The fast dissolving tablets usually dissolve in the oral cavity within 15 seconds

to 3 minutes. In another words a fast-dissolving tablet is tablet that dissolves or

disintegrates in the oral cavity without the need of water or chewing.

Fast dissolving tablets are also called as Orodispersible tablets, Quick

disintegrating tablets, Mouth dissolving tablets, Oral rapid disintegrating tablets,

Rapid dissolving tablets, Porous tablets and Rapimelts. However, of all the above

terms, United States Pharmacopoeia (USP) approved those dosage forms as Orally

Disintegrating Tablets (ODTs). Recently European Pharmacopoeia has used the term

Orodispersible tablet for tablets that disperses readily and within three minutes in

mouth before swallowing.

United States Food and Drug Administration (USFDA) define ODT as “A

solid dosage form containing medicinal substances or an active ingredient which

disintegrates rapidly usually within a matter of seconds when placed upon tongue”.

The disintegration time for fast dissolving tablets generally ranges from several

seconds to about a minute.12

1.12. ADVANTAGES OF FAST DISSOLVING DRUG DELIVERY SYSTEM:


13, 14, 15, 16, 17.

• Ease of administration to pediatric, geriatric patients and psychiatric patients.

• Free of the risk of suffocation due to physical obstruction when swallowed,

thus offering improved safety.

• Convenience of administrate accurate dose as compared to liquids.

• Having good mouths feel property.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 23 


 
INTRODUCTION

• No need of water to swallow the dosage from.

• Rapid dissolution of drug and absorption, which may produce rapid onset of

action from the mouth, pharynx and esophagus.

• Pregastric absorption can result in improved bioavailability, reduced dose and

improved clinical performance by reducing side effects.

• New business opportunities: product differentiation, line extension and life-cycle

management, exclusivity of product promotion and patent-life extension.

1.13. LIMITATIONS OF FAST DISSOLVING TABLETS: 18.

• Drugs with relatively larger doses are difficult to formulate into FDT e.g.

antibiotics like ciprofloxacin with adult dose tablet containing about 500 mg of the

drug.

• Patients who concurrently take anticholinergic medications may not be the

best candidates for FDT. Similarly patients with Sjögren's syndrome or dryness of the

mouth due to decreased saliva production may not be good candidates for these tablet

formulations.

• The tablets usually have insufficient mechanical strength. Hence, careful

handling is required.

• The tablets may leave unplesant taste and/or grittiness in mouth if not

formulated properly.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 24 


 
INTRODUCTION

1.14. CHALLENGES TO DEVELOP FAST DISSOLVING TABLET: 19, 20.

I) Mechanical strength and disintegration time-

ODTs are formulated to obtain disintegration time usually less than a minute.

While doing so, maintaining a good mechanical strength is a prime challenge. Many

ODTs are fragile and there are many chances that such fragile tablet will break during

packing, transport or handling by the patients. Tablets based on technologies like

Zydis need special type of packaging. It is very natural that increasing the mechanical

strength will delay the disintegration time. So a good compromise between these two

parameters is always essential.

II) Taste masking-

Many drugs are bitter in taste. A tablet of bitter drug dissolving/ disintegration

in mouth will seriously affect patient compliance and acceptance for the dosage form.

So effective taste masking of the bitter drugs must be done so that the taste of the drug

is not felt in the oral cavity.

III) Mouth feel-

The ODT should not disintegrate into larger particles in the oral cavity. The

particles generated after disintegration of the ODT should be as small as possible.

ODT should leave minimal or no residue in mouth after oral administration. Morever

addition of flavours and cooling agents like menthol improve the mouth feel.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 25 


 
INTRODUCTION

IV) Sensitivity to environmental conditions-

ODT generally should exhibit low sensitivity to environment conditions such

as humidity and temperature as most of the materials used in an ODT are meant to

dissolve in minimum quantity of water.

V) Amount of drug:

For lyophilized dosage forms, the drug dose must be lower than 400 mg for

insoluble drugs and less than 60 mg for soluble drugs.

VI) Aqueous solubility:

Water-soluble drugs form eutectic mixtures, which result in freezing-point

depression and the formation of a glassy solid that may collapse upon drying because

of loss of supporting structure during the sublimation process.

VII) Size of tablet:

It has been reported that the easiest size of tablet to swallow is 7-8 mm while

the easiest size to handle was larger than 8 mm. Therefore, the tablet size that is both

easy to take and easy to handle is difficult to achieve.

VIII) Cost-

The technology used for an ODT should be acceptable in terms of cost of the

final product. Methods like Zydis and Orasolv that require special technologies and

specific packaging increase the cost to a remarkable extent8.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 26 


 
INTRODUCTION

1.15. SELECTION OF DRUGS: 21

The ideal characteristics of a drug to be selected.

™ No bitter taste.

™ Dose lower than 20mg.

™ Small to moderate molecular weight.

™ Good stability in water and saliva.

™ Partially non ionized at the oral cavities pH.

™ Ability to diffuse and partition into the epithelium of the upper GIT (log p>1,

or preferably>2).

™ Ability to permeate oral mucosal tissue.

1.16. EXCIPIENTS USED IN FAST DISSOLVING TABLET:

• Super disintegrants:

Crosspovidone, Microcrystalline cellulose, sodium starch glycollate, sodium

carboxy methyl cellulose, pregelatinzed starch, calcium carboxy methyl cellulose, and

modified corn starch. Sodium starch glycollate has good flowability than

crosscarmellose sodium. Cross povidone is fibrous nature and highly compactable.

• Flavours:

Peppermint flavor, cooling flavor, flavor oils and flavoring aromatic oil,

peppermint oil, clove oil, bay oil, anise oil, Cardamom flavor, eucalyptus oil thyme

oil, oil of bitter almonds. Flavoring agents include, vanilla, citus oils, fruit essences

• Sweeteners’:

Aspartame, Sugars derivatives

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 27 


 
INTRODUCTION

• Fillers:

Directly compressible spray dried Mannitol, Lactose, Dextrose, Sorbitol, xylitol,

calcium carbonate, magnesium carbonate, calcium phosphate, calcium sulfate,

pregelatinized starch, magnesium trisilicate, aluminium hydroxide.

• Surface active agents:

Sodiumdoecylsulfate, sodiumlaurylsulfate, polyoxyethylene sorbitan fatty acid

esters (Tweens), sorbitan fatty acid esters (Spans), polyoxyethylene stearates.

• Lubricants:

Stearic acid, Magnesium stearate, Zinc stearate, calcium stearate, talc,

polyethylene glycol, liquid paraffin, magnesium lauryl sulfate, colloidal silicon

dioxide.

1.17. SUPERDISINTEGRANTS: 22

Disintegrants are substances routinely included in tablet formulations and in

some hard shell capsule formulations to promote moisture penetration and dispersion

of the matrix of dosage form in dissolution fluids. An oral solid dosage form should

ideally disperse into the primary particles from which it was prepared.

Superdisintegrants are generally used at a low concentration, typically 1-10%

by weight relative to total weight of dosage unit. Generally employed

superdisintegrants are croscarmellose sodium (Ac-Di-Sol), crospovidone (CP),

sodium starch glycolate (SSG) etc. which represent example of cross-linked cellulose,

cross-linked polymer and cross-linked starch respectively.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 28 


 
INTRODUCTION

Selection of appropriate formulation excipients and manufacturing technology is

necessary for obtaining the optimized design features of orally disintegrating dosage

forms. Ideally, superdisintegrants should cause the tablet to disrupt, not only into the

granules from which it was compressed but also into powder particles from which the

granules were prepared

1.18. SELECTION OF SUPERDISINTEGRANTS: 22

Although superdisintegrants primarily affect the rate of disintegration, but

when used at high levels they can also affect mouth feel, tablet hardness and friability.

Hence, various ideal factors to be considered while selecting an appropriate

superdisintegrants for a particular formulation should:

¾ Produce rapid disintegration, when tablet comes in contact with saliva in the

mouth/oral cavity.

¾ Be compactable enough to produce less friable tablets.

¾ Produce good mouth feel to the patients. Thus, small particle size is preferred

to achieve patient compliance.

¾ Have good flow, since it improves the flow characteristics of total blend.

1.19. MECHANISM OF ACTION OF DISINTEGRANT: 22

Various mechanisms proposed in this concern include water wicking,

swelling, deformation recovery, repulsion and heat of wetting. It seems likely that no

single mechanism can explain the complex behavior of the disintegrants. However,

each of these proposed mechanisms provides some understanding of different aspects

of disintegrant action.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 29 


 
INTRODUCTION

I. Water wicking

The ability of disintegrant to draw water into the porous network of tablet is

essential for effective disintegration. On keeping the tablet into suitable aqueous

medium, the medium enters into tablet and replaces the air adsorbed on the particles

which weakens the intermolecular bonds and breaks the tablet into fine particles.

Water uptake by tablet depends upon hydrophilicity of the drug/excipients and on

tableting conditions. Unlike swelling, which is mainly a measure of volume expansion

with accompanying force generation, water wicking is not necessarily accompanied

by a volume increase. The ability of a system to draw water can be summarized by

Washburn’s equation:

L2 = (γ Cosθ/2η) × rt

The Washburn equation is too simplistic to apply to a dynamic tablet-

disintegration process, but it does show that any change in the surface tension (γ),

pore size (r), solid-liquid contact angle (θ) or liquid viscosity (η) could change the

water wicking efficiency. L is the length of water penetration in the capillary and t is

the time. This process is also considered as capillary action method.

II. Swelling

Although water penetration is a necessary first step for disintegration, swelling

is probably the most widely accepted mechanism of action for tablet disintegrants. For

swelling to be effective as a mechanism of disintegration, there must be a

superstructure against which disintegrant swells. Figure 2 represents the disintegration

of tablet by wicking and swelling. Swelling of the disintegrant against the matrix

leads to development of a swelling force. A large internal porosity in the dosage form

in which much of the swelling can be accommodated reduces the effectiveness of the

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 30 


 
INTRODUCTION

disintegrant. On the other hand, sufficient swelling force is exerted in the tablet with

low porosity. It is worthwhile to note that if packing fraction is very high, fluid is

unable to penetrate in the tablet and disintegration is again slowed down.

Figure 2: Disintegration of tablet by wicking and swelling

III. Heat of wetting

When disintegrants with exothermic properties get wetted, localized stress is

created due to capillary air expansion, which aids in disintegration of tablet. This

explanation, however, is limited to only a few types of disintegrants and cannot

describe the action of most modern disintegrating agents.

IV. Due to release of gases

Carbon dioxide gets released within tablets on wetting due to interaction

between bicarbonate and carbonate with citric acid or tartaric acid. The tablet

disintegrates due to generation of pressure within the tablet. This effervescent mixture

is used when pharmacist needs to formulate very rapidly dissolving tablets or fast

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 31 


 
INTRODUCTION

disintegrating tablet. As these disintegrants are highly sensitive to small changes in

humidity level and temperature, strict control of environment is required during

preparation of the tablets. The effervescent blend is either added immediately prior to

compression or can be added into two separate fractions of formulation.

V. Particle repulsive forces

This is another mechanism of disintegration that attempts to explain the

swelling of tablet made with non-swellable disintegrants. Guyot-Hermann proposed a

particle-particle repulsion theory to explain the observation that particles which do not

swell extensively such as starch, could still disintegrates tablets. According to this

theory, water penetrates into tablet through hydrophilic pores and a continuous starch

network is created that can convey water from one particle to the next, imparting a

significant hydrostatic pressure. The water then penetrates between starch grains

because of its affinity for starch surfaces, thereby breaking hydrogen bonds and other

forces holding the tablet together. The electric repulsive forces between particles are

the mechanism of disintegration and water is required for it.

VI. Deformation recovery

Deformation recovery theory implies that the shape of disintegrant particles is

distorted during compression and the particles return to their precompression shape

upon wetting, thereby causing the tablet to break apart. Such a phenomenon may be

an important aspect of the mechanism of action of disintegrants such as crospovidone

and starch that exhibit little or no swelling. Disintegration of tablet by deformation as

well as repulsion is illustrated in Figure 3.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 32 


 
INTRODUCTION

Figure 3: Disintegration by deformation and repulsion

VII. By enzymatic reaction

Enzymes present in the body also act as disintegrants. These enzymes dearth

the binding action of binder and helps in disintegration. Due to swelling, pressure is

exerted in the outer direction that causes the tablet to burst or the accelerated

absorption of water leads to an enormous increase in the volume of granules to

promote disintegration.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 33 


 
INTRODUCTION

1.20. TECHNOLOGIES USED TO MANUFACTURE FAST DISSOLVING


23-29.
TABLET:

The technologies used to manufacture fast dissolving tablets can be classified as:

TECHNOLOGIES

CONVENTIONAL PATENTED
TECHNOLOGIES TECHNOLOGIES

i. Freeze Drying i. Zydis Technology

ii. Cotton Candy Process ii. Orasolv Technology

iii. Tablet Molding iii. Durasolv Technology


iv. Nanocrystal Technology
iv Sublimation
v. Dispersible Tablet Technology
v. Spray Drying vi. Wowtab Technology
vii. Flashtab Technology
vi. Mass extrusion
viii. Lyoc Technology
vii. Direct Compression
ix. Pharmaburst Technology
x. Frosta Technology
xi. Ziplets/advatab Technology
xii. Oraquick Technology

Figure-4: Various Technologies Used to Manufacture Fast Dissolving Tablet

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 34 


 
INTRODUCTION

3.10.1. CONVENTIONAL TECHNIQUES:

™ Lyophilization or Freeze Drying

Formation of porous product in freeze-drying process is exploited in

formulating Fast dissolving tablets (FDT). Lyophilization is a process, which includes

the removal of solvent from a frozen suspension or solution of drug with structure-

forming additives. Freeze-drying of drug along with additives imparts glossy

amorphous structure resulting in highly porous and light weight product. The resulting

tablet has rapid disintegration and dissolution when placed on the tongue and the

freeze-dried unit dissolves instantly to release the drug. However, the FDTs formed

by lyophilization have low mechanical strength, poor stability at higher temperature,

and humidity. Along with above complications and its expensive equipment for

freeze-drying is observed to be limitation of this technology.

™ Cotton Candy Process

This process is so named as it utilizes a unique spinning mechanism to

produce floss-like crystalline structure, which mimic cotton candy. Cotton candy

process involves formation of matrix of polysaccharides or saccharides by

simultaneous action of flash melting and spinning. The matrix formed is partially

recrystallized to have improved flow properties and compressibility. This candy floss

matrix is then milled and blended with active ingredients subsequently compressed to

Fast dissolving tablets. This process can accommodate high doses of drug and offers

improved mechanical strength. However, high-process temperature limits the use of

this process.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 35 


 
INTRODUCTION

™ Molding

Molding process includes moistening, dissolving or dispersing the drug with a

solvent then molding the moist mixture into tablets (compression molding with lower

pressure than conventional tablet compression), evaporating the solvent from drug

solution, or suspension at ambient pressure (no vacuum lyophilization), respectively.

The molded tablets formed by compression molding are air-dried. As the compression

force employed is lower than conventional tablets, the molded tablet results in highly

porous structure, which increases the disintegration and dissolution rate of the

product. However, to further improve dissolution rate of the product powder mixture

should be sieved through very fine screen. As molding process is employed usually

with soluble ingredients (saccharides) which offers improved mouth feel and

disintegration of tablets. However, molded tablets have low mechanical strength,

which results in erosion and breakage during handling.

™ Sublimation

The presence of a highly porous structure in the tablet matrix is the key factor

for rapid disintegration of Fast dissolving tablets. Even though the conventional

tablets contain highly water-soluble ingredients, they often fail to disintegrate rapidly

because of low porosity. To improve the porosity, volatile substances such as

camphor can be used in tableting process, which sublimated from the formed tablets,

Koizumi et al. developed Fast dissolving tablet (FDT) utilizing camphor; a subliming

material that is removed from compressed tablets prepared using a mixture of

mannitol and camphor. Camphor was sublimated in vacuum at 80° for 30 minutes

after preparation of tablets.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 36 


 
INTRODUCTION

Figure5: Step Involved In Sublimation Process

™ Spray-Drying

Highly porous, fine powders are obtained by this method. Allen et al. utilized

this process for preparing Fast dissolving tablets. The Fast dissolving tablet

formulations consisted of hydrolyzed/unhydrolyzed gelatin as supporting agents for

matrix, mannitol as bulking agent, and sodium starch glycolate or croscarmellose

sodium as disintegrating agent. Disintegration and dissolution were further improved

by adding effervescent components, i.e. citric acid (an acid) and sodium bicarbonate

(an alkali). The formulation was spray dried to yield a porous powder. The fast

dissolving tablets made from this method disintegrated within a minute.

™ Mass-Extrusion

This technology involves softening the active blend using the solvent mixture

of water-soluble polyethylene glycol and methanol and subsequent expulsion of

softened mass through the extruder or syringe to get a cylinder of the product into

even segments using heated blade to form tablets.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 37 


 
INTRODUCTION

™ Direct Compression

Easiest way to manufacture tablets is direct compression. Low manufacturing

cost, conventional equipment’s and limited number of processing steps led this

technique to be a preferable one. However disintegration and dissolution of directly

compressed tablets depend on single or combined effect of disintegrant, water soluble

excipients and effervescing agents.

It is essential to choose a suitable and an optimum concentration of

disintegrant to ensure quick disintegration and dissolution. Superdisintegrants are

newer substances which are more effective at lower concentrations with greater

disintegrating efficiency and mechanical strength. On contact with water the

superdisintegrants swell, hydrate, change volume or form and produce a disruptive

change in the tablet. Effective superdisintegrants provide improved compressibility,

compatibility and have no negative impact on the mechanical strength of formulations

containing high dose drugs. The type of disintegrants and its proportion are of prime

importance. Also factors to be considered are particle size distribution, contact angle,

pore size distribution and water absorption capacity. Studies revealed that the water

insoluble superdisintegrants like sodium starch glycolate and Croscarmellose sodium

show better disintegration property than the slightly water soluble agents like

Crospovidone, since they do not have a tendency to swell. Superdisintegrants that

tend to swell show slight retardation of the disintegration property due to formation of

viscous barrier. There is no particular upper limit regarding the amount of

superdisintegrant as long as the mechanical properties of the tablet are compatible

with its intended use. The superdisintegrant may be used alone or in combination with

other superdisintegrants. Commercially available superdisintegrants which are widely

used are listed in the Table 1.1.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 38 


 
INTRODUCTION

Table 2: Various commercially available superdisintegrants along with their

properties.

Sr. no. Name Type Properties Brand name

1. Crospovidone Polyvinyl- Crossed linked Polyplasdone XL,

pyrrolidone Polyvinlypyrrolidone Rapidly Kollidon CL

disperses and swells in water

2. Croscarmellose Modified Cross linked sodium carboxy Ac-di-sol,

Sodium. cellulose methylcellulose. Excellent Primellose,

swelling and water wicking Solutab.

properties.

3. Sodium starch Modified Sodium salt of carboxy methyl Primogel,

Glycolate Starch ether of starch. High swelling Explotab,

capacity and rapid water Glycolys.

uptake

4. Indion 414 Ion exchange Cross linked polyacrylic with Indion 414.

Resin a -COO- functional group and

K+ ionic form. High water

intake facility.

Other superdisintegrants which are rarely used are Gellan gum, Xanthan gum and

Soya polysaccharide.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 39 


 
INTRODUCTION

3.10.2. PATENTED TECHNOLOGIES:

¾ Zydis Technology

This technology includes physical trapping of the drug in a matrix composed

of a saccharide and a polymer. Polymers generally employed are partially hydrolyzed

gelatin, hydrolyzed dextran, dextrin, alginates, polyvinyl alcohol, polyvinyl

pyrrolidine, acacia and mixture of these. The methodology involves solution or

dispersion of components is prepared and filled in to blister cavities, which are frozen

in a liquid nitrogen environment. The frozen solvent is removed or sublimed to

produce porous wafers. Peelable backing foil is used to pack Zydis units. Zydis

formulation is sensitive to moisture and may degrade at humidity greater than 65%

RH.

¾ Durasolv Technology

The tablets produced by this technology utilize the conventional tableting

equipment. Tablets in this are formulated by using drug, nondirect compression fillers

and lubricants. Nondirect compressible fillers are dextrose, mannitol, sorbitol, lactose

and sucrose, which have advantages of quick dissolution and avoid gritty texture,

which is generally present in direct compressible sugar. The tablets obtained are

strong and can be packed in conventional packing in bottles and blisters. Nondirect

compressible fillers generally used in the range of 60-95%, lubricant in 1-2.5%.

¾ Orasolv Technology

This includes use of effervescent disintegrating agents compressed with low

pressure to produce the Fast dissolving tablets (FDT). The evolution of carbon

dioxide from the tablet produces fizzing sensation, which is a positive organoleptic

property. Concentration of effervescent mixture usually employed is 20-25% of tablet

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 40 


 
INTRODUCTION

weight. As tablets are prepared at low compression force, they are soft and fragile in

nature. This initiated to develop Paksolv a special packaging to protect tablets from

breaking during storage and transport. Paksolv is a dome-shaped blister package,

which prevents vertical movement of tablet with in the depression. Paksolv offers

moisture, light and child resistance packing.

¾ Nanocrystal Technology

Nanocrystal technology includes lyophilization of colloidal dispersions of

drug substance and water-soluble ingredients filled in to blister pockets. This method

avoids manufacturing process such as granulation, blending, and tableting, which is

more advantageous for highly potent and hazardous drugs. As manufacturing losses

are negligible, this process is useful for small quantities of drug.

¾ Dispersible tablet Technology

It offers development of Fast dissolving tablets with improved dissolution rate

by incorporating 8-10% of organic acids and disintegrating agents. Disintegrating

agents facilitates rapid swelling and good wetting capabilities to the tablets that results

in quick disintegration. Disintegrants include starch, modified starches,

microcrystalline cellulose, alginic acid, cross-linked sodium carboxy methyl cellulose

and cyclodextrins. Combination of disintegrants improved disintegration of tablets

usually less than 1 minute.

¾ Wowtab Technology

“WOW” means without water. This technology utilizes conventional

granulation and tableting methods to produce fast dissolving tablets employing low

and high moldability saccharides. Low moldability saccharides are lactose mannitol,

glucose, sucrose and xylitol. High-moldability saccharides are maltose, maltitol,

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 41 


 
INTRODUCTION

sorbitol and oligosaccharides. When these low and high moldable saccharides used

alone tablets obtained do not have desired properties of rapid disintegration and

hardness, so combinations are used. This technology involves granulation of low

moldable saccharides with high moldable saccharides as a binder and compressing

into tablets followed by moisture treatment. Thus tablets obtained showed adequate

hardness and rapid disintegration.

¾ Flashtab Technology

This technology includes granulation of excipients by wet or dry granulation

method and followed by compressing into tablets. Excipients used in this technology

are of two types. Disintegrating agents include reticulated polyvinylpyrrolidine or

carboxy methylcellulose. Swelling agents include carboxymethylcellulose, starch,

modified starch, microcrystalline cellulose, carboxy methylated starch, etc. These

tablets have satisfactory physical resistance. Disintegration time is within 1 minute.

¾ Lyoc Technology

Oil in water emulsion is prepared and placed directly into blister cavities

followed by freeze-drying. Nonhomogeneity during freezedrying is avoided by

incorporating inert filler to increase the viscosity finally the sedimentation. High

proportion of filler reduces porosity of tablets due to which disintegration is lowered.

¾ Frosta Technology

It utilizes the concept of formulating plastic granules and compressing at low

pressure to produce strong tablets with high porosity. Plastic granules composed of:

• Porous and plastic material,

• Water penetration enhancer, and

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 42 


 
INTRODUCTION

• Binder.

The process involves usually mixing the porous plastic material with water

penetration enhancer and followed by granulating with binder. The tablets obtained

have excellent hardness and rapid disintegration time ranging from 15 to 30 s

depending on size of tablet.

¾ OraQuick

The OraQuick fast-dissolving/disintegrating tablet formulation utilizes a

patented taste masking technology. KV Pharmaceutical claims its microsphere

technology known as MicroMask, has superior mouth feel over taste-masking

alternatives. The taste masking process does not utilize solvents of any kind and

therefore leads to faster and more efficient production. Also, lower heat of production

than alternative fast-dissolving/disintegrating technologies makes OraQuick

appropriate for heat-sensitive drugs. KV Pharmaceutical also claims that the matrix

that surrounds and protects the drug powder in microencapsulated particles is more

pliable, meaning tablets can be compressed to achieve significant mechanical strength

without disrupting taste-masking. OraQuick claims quick dissolution in a matter of

seconds, with good taste-masking. There are no products using the OraQuick

technology currently on the market, but KV Pharmaceutical has products in

development such as analgesics, scheduled drugs, cough and cold, psychotropics, and

anti-infectives.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 43 


 
INTRODUCTION

Table 3: Comparison of some patented technologies for fast dissolving tablets

Drug release /
Technology Novelty Handling/storage
bioavailability

Zydis First to market. Do not push tablet through Dissolves in 2 to 10

Freeze Dried foil. Do not use dosage form seconds. May allow for
(R.P. Scherer, Inc.)
from damaged package. pre-gastric absorption

Sensitive to degradation at leading to enhanced

humidities >65% bioavailability

Orasolv Unique taste Packaged in patented foil Disintegrates in 5 to 45

masking. Lightly packs seconds depending upon


(Cima Labs, Inc.)
compressed the size of the tablet. No

significant change in drug

bioavailability

Durasolv Similar to Orasolv, Packaged in foil or bottles. If Disintegrates in 5 to 45

but with better packaged in bottles, avoid seconds depending upon


(Cima Labs, Inc.)
mechanical exposure to moisture or the size of the tablet. No

strength humidity significant change in drug

bioavailability

Wowtab Compressed dosage Package in bottles. Avoid Disintegrates in 5 to 45

(Yamanouchi form. Proprietary exposure to moisture or seconds depending upon

Pharma taste masking. humidity the size of the tablet. No

Technologies, Inc.) Smooth melt action significant change in drug

gives superior bioavailability

mouth feel

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 44 


 
AIM AND OBJECTIVE

CHAPTER-2.

OBJECTIVES OF THE STUDY

The aim of present research work was to formulate FDT of already used

therapeutic molecule to enhance effectiveness, and to avoid side effects (gastric

irritation) of the drug. For FDT most promising NSAID’s candidate Ketoprofen is

selected for the present study.

Thus the objectives of the work were:

™ To enhance the solubility and bioavailability of Ketoprofen by Solid

Dispersions technique

™ Evaluate the potential of Polyvinyl pyrolidone K 30, Polyethylene glycol-6000

as suitable drug carrier systems for delivery of Ketoprofen

™ Determine the effect of change in polymer and polymer composition and

Drug-polymer ratio on solubility of Ketoprofen.

™ Study of in-vitro dissolution kinetics of Ketoprofen from the formulated Solid

dispersion systems.

™ To formulate and evaluate fast dissolving tablets of Ketoprofen, having

adequate mechanical strength, rapid disintegration and fast action.

2.2 RATIONALE BEHIND THE SELECTION OF DRUG:

• Ketoprofen belongs to class II drug under BCS classification i.e. low solubility

and high permeability.

• Ketoprofen is non-steroidal anti-inflammatory drug, which is used in the

treatment of rheumatoid arthritis and osteoarthritis.

DEPT. OF PHARMACEUTICS, Dr.H.L.T.COLLEGE OF PHARMACY.  Page 45 


AIM AND OBJECTIVE

• One of the major problems with this drug is its low solubility in biological

fluids, which results into poor bioavailability after oral administration.

• The solubility of Ketoprofen in aqueous medium was very low i.e. 0.016

mg/ml in water.

• Absolute bioavailability of the Ketoprofen was 99% and biological half-life is

only 2±0.3 hours that results into poor bioavailability after oral administration.

• Conjugation of Ketoprofen with the different types of carriers to increases the

solubility and dissolution rate of Ketoprofen.

• By increasing the solubility of Ketoprofen, its dissolution rate and

consequently bioavailability is increased.

2.3 PLAN OF WORK:

Based upon the literature survey, the application of solid dispersions results in

increasing the solubility of many poorly soluble drugs, the objective of the present

study, investigated to improve the solubility and consequently bioavailability of

Ketoprofen by using different carriers.

Scheme of proposed work is as follows:

• Analytical method development for analysis using UV Spectrophotometer

method.

o Scanning of λmax of Ketoprofen

o Standard plot of Ketoprofen

• Preparation of Ketoprofen solid dispersions.

DEPT. OF PHARMACEUTICS, Dr.H.L.T.COLLEGE OF PHARMACY.  Page 46 


AIM AND OBJECTIVE

• Evaluation of Ketoprofen solid dispersions by Solubility, Drug content,

Dissolution efficiency, FT-IR, DSC, XRD, SEM, in-vitro dissolution studies.

• Formulation of Ketoprofen tablets from selected Ketoprofen solid dispersions

• Evaluation of prepared Ketoprofen tablets.

• To evaluate the stability studies of best formulations as per ICH guidelines.

DEPT. OF PHARMACEUTICS, Dr.H.L.T.COLLEGE OF PHARMACY.  Page 47 


REVIEW OF LITERATURE

CHAPTER-3.

REVIEW OF LITERATURE

3.1. REVIEW OF LITERATURE

1. Late SG, et.al. (2009), Were investigated effects of calcium silicate

(disintegration-promoting agent) and various lubricants on an optimized

cyclodextrin-based fast-disintegrating tablet formulation. Effects of moisture

treatment were also evaluated at 75, 85 and 95% relative humidities. A two

factor, three levels (32) full factorial design was used to optimize

concentrations of calcium silicate and lubricant. Magnesium stearate, being

commonly used lubricant, was used to optimize lubricant concentration in

optimization study. Results of multiple linear regression analysis revealed that

concentration of calcium silicate had no effect; however concentration of

lubricant was found to be important for tablet disintegration and hardness.30

2. Ahmed I. S, et.al. (2006), Were developed Ketoprofen tablets which dissolve-

rapidly in the mouth. The solubility and dissolution rate of poorly water-

soluble Ketoprofen was improved by preparing a lyophilized tablet (LT) of

Ketoprofen using freeze-drying technique.31

3. Maria Victoria Margarit et.al, (1994), Were prepared SD of Ketoprofen by

using PEG-6000 and compared the dissolution kinetics of the dispersions with

physical mixtures and pure drug. The results of dissolution kinetics studies

showed that PEG-6000, when used as a carrier for solid dispersions, increased

the dissolution rate of Ketoprofen. The t 80% of dissolution for pure drug

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 48 


REVIEW OF LITERATURE

(88.85% min) decreased to 1.9, 4 and 22.5 min, respectively, in solid

dispersion containing 10:90, 50:50, and 90:10 proportions of Ketoprofen/PEG-

6000.32

4. Piera Di Martino et.al., (2004), Were studied Ketoprofen-

polyvinylpyrrolidone physical interaction, amorphous solid solutions were

obtained in different proportions by dissolving Ketoprofen and

Polyvinylpyrrolidone K-30 in methanol and by evaporating then under

reduced pressure. The result indicated that in the Ket-PVP co-precipitates, the

ket molecules, interacting with their carboxylic group through hydrogen

bonding with the PVP moieties, are molecularly and irregularly dispersed

within the amorphous solvendum that acts as mechanical substratum for

amorphous stabilization.33

5. R. Jachowicz et.al, (2000), Were prepared ternary solid dispersion of

Ketoprofen with macrogol and kollagen hydrolizate as carriers by different

method (melting, solvent method, different cooling) in different concentrations

of drug/carriers. The best solid dispersion consisted of Ketoprofen-Macrogol

6000-KLHT (1+8.9+0.1) was chosen to formulate the pellets on the basis of

the pharmaceutical availability of Ketoprofen from solid dispersion and the

physical chemical studies. The increase in the amount of released Ketoprofen

from solid dispersion pellets was 3.8 times greater than from the pellets

containing the drug alone.34

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 49 


REVIEW OF LITERATURE

6. Vasanthkumar S. et.al., (2008), Were prepared immediate release tablets of

Telmisartan using superdisintegrant like polyplasdone XL-10 at intragranular,

extragranular and partly intra and extragranular level of addition to increase

the rate of drug release from dosage form to increase the dissolution rate and

hence its bioavailability. It was concluded that the immediate release tablets

with proper hardness, disintegration time and with increase rate of dissolution

can be made using polyplasdone XL-10. Formulation F 10 was the best

formulation having disintegration time 4.11 sec.35

7. Melleswara rao et.al, (2008), Were prepared solid dispersion of Meloxicam

by using PVA, PVP, PEG-4000 and PEG-6000 in 1:1, 1:2 and 1:4 ratios by

solvent evaporation method. In-vitro release profile of all SDs (F1 to F12)

were comparatively evaluated and also studied against pure meloxicam. Faster

dissolution was exhibited by SD containing 1:2 ratio of drug: PEG-6000 due

to increase in wettability, hydrophilic nature of the carrier and also due to

reduction in drug crystallinity.36

8. P G Bhole et.al., (2009), Were prepared solid dispersion of Felodipine by

using PEG-6000 and PVA in different ratio by solvent evaporation

method.DSC and XRD analysis demonstrated the conversion of felodipine to

amorphous form with both physical mixture and SD. SD with PVA released

95% of the drug in 85 min as compared with 89%of drug released in 90 min

by SD with PEG-6000. Thus SD with both polymers increased drug release,

particularly grater in case of PVA than PEG-6000.37

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 50 


REVIEW OF LITERATURE

9. Dario Leonardi et.al, (2007), Were prepared fast release tablets of Prednisone

by using SD technique. SD was prepared by using PEG-6000 by solvent

evaporation method. As demonstrated by both XRD and SEM, a decreased

crystallinity of both prednisone and the surface morphology of the polymeric

particles explained this improved dissolution rate. The tablets containing those

SD particles had dissolution profiles that were better than those of

conventional tablets without PEG-6000.38

10. M.M. Patel et.al., (2006), Were prepared fast dissolving Valdecoxib tablets

containing SD of Valdecoxib by using mannitol (melt solvent) PEG-6000 and

PVP K-12 solvent evaporation method in different ratio 1:1, 1:2, 1:4, 1:6, 1:8,

1:10. SD with PVP K-12 showed maximum drug release the tablets were

prepared and F1 formulation shows better dissolution than other formulation

and conventional marketed tablets which released only 44.3% drug and

Valdecoxib in β-cyclodextrin , which released 53.4% drug in 20 min, while F1

exhibited almost 100% drug release in 20 min.39

11. D. Nagendrakumar et.al., (2009), Were prepared fast dissolving tablets of

fexofenadine Hcl by effervescent method by using crospovidone,

croscarmalose sodium and sodium starch glycolate along with sodium

bicarbonate and anhydrous citric acid in different ratios. the formulation ECP3

containing 8% w/w of crospovidone and mixture of 24% w/w sodium

bicarbonate, 18% w/w of anhydrous citric acid emerged as the best (t 50% 4

min) based on the in vitro drug release characteristics compared to

conventional commercial tablet formulation (t 50% 15 min).40

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 51 


REVIEW OF LITERATURE

12. S.B.Shirsand et.al., (2008), Were prepared fast disintegrating tablets of

Prochlorperazine maleate by effervescence method by using sodium

bicarbonate and anhydrous citric acid in different ratios along with

crospovidone (2-10% w/w), croscarmalose sodium (2-10% w/w), were used as

superdisintegrants. Among the all formulation , the formulation containing

10% w/w of crospovidone and mixture of 20% w/w sodium bicarbonate, 15%

w/w of anhydrous citric acid emerged as the best (t 50% 6 min) based on the in

vitro drug release characteristics compared to conventional commercial tablet

formulation (t 50% 17.4 min).41

13. Bhalerao AV, et.al. (2009), Were developed fast disintegrating tablets of

Clonazepam using superdisintigrants and solid dispersion technique. The drug

is poorly water soluble therefore to enhance the solubility and release of drug,

solid dispersion of drug and PVP K30was prepared by solvent evaporation

method. Different combinations of superdisintegrants such as croscarmellose

sodium, sodium starch glycolate, crospovidone were used. The tablets were

prepared. All the tablets had hardness 3-3.5 Kg/cm2 and friability was less than

1. Among all formulations, formulation F10 prepared by drug:PVPK30 (1:4)

ratio and combination of 5%w/w crosscarmellose sodium and 5% w/w of

sodium starch glycolate showed best dispersion time of 8 sec and faster

dissolution 42

14. Singh J, et.al, (2009), Were formulated and optimized orodispersible

formulation of Meloxicam using a 22 factorial design for enhanced

bioavailiabity. The tablets were made by non-aqueous wet granulation using

crospovidone and mannitol. A22 factorial design was used to investigate the

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 52 


REVIEW OF LITERATURE

amount of crospovidone and taste masking, smoothening hydrophilic agent

(manifold), as independent variables, and disintegration time as dependent

response. Formulated orodispersible tablets were evaluated for weight

variation, friability, disintegration time, drug content, wetting time, water

absorption ratio and in vitro drug release.43

15. Radke RS, et.al., (2009), Were formulated orodispersible tablets of Baclofen

using various concentrations of superdisintegrant agents like AC-Di-Sol,

crospovidone, sodium starch glycolate by direct compression method. These

tablets were evaluated for drug content weight variation, friability, hardness,

wetting time and invitro disintegration time. It was concluded that

superdisintergtrants addition technique is a useful method for preparing

orodispersible tablets by direct compression method.44

16. Gangane P.S., et.al, (2009), Were formulated rapid disintegrating tablet of

Gatifloxacin Sesquihydrate using Indion 204, Indion 214, Indion 234, Tulsion

335 (ion exchange resin) as a taste masking agent. The tablet was prepared by

wet granulation technique with three superdisintegrants e.g. sodium starch

glycolate, crosscarmellose sodium and crospovidone. The granules were

examined for angle of repose, bulk density, tapped density and Hauser’s ratio.

The tablets were evaluated for hardness, drug content and friability and

disintegration time. The disintegration in oral cavity was also tested and was
45
found to be 22 sec.

17. Venkatesh D. P., et.al, (2008), Were formulated the taste masked oral –

dispersible tablets of Ambroxol hydrochloride. Cation exchange resins like

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 53 


REVIEW OF LITERATURE

Indion-204 and Indion-234 were utilized for the sorption of drug. Drug-

resinates was prepared in drug to ratio of 1:5 and 1:6. Tablets with both the

resins have shown quick disintegrating features, i.e., within 20 sec which is

very characteristics of orodispersible tablets. Almost more than 90% of drug

was released from the both the formulation within 1 hour .46

18. Patel D M, et.al, (2008), Were prepared Etoricoxib fast dissolving tablets.

Granules containing etoricoxib, Menthol, crospovidone aspartame and

mannitol were prepared by wet granulation technique. Menthol was sublimed

from the granules by exposing the granules to vacuum. The porous granules

were then compressed in to tablets. Alternatively, tables were first prepared

and later exposed to vacuum. The tablets were evaluated for percentage

friability and disintegration time. A32 full factorial design was applied to

investigate the combined effect of to formulation variables: amount of menthol

and crospovidone. The results of multiple regression analysis indicated that for

obtaining fast dissolving tablets; optimum amount of menthol and higher

percentage of crospovidone should be used.47

19. Furtado S, et.al, (2008), Were developed orodispersible tablets of Famotidine

containing a subliming agent. Mannitol as a diluent, sodium saccharin as

sweetening agent, alcoholic solution of PVP (10%w/v) as binder and

magnesium stearate with talc as a flow promoter. Addition of camphor in the

formulation improved the tablet properties with respect to wetting time and in

vitro dispersion time. The present study demonstrated potentials for rapid

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 54 


REVIEW OF LITERATURE

absorption, improved bioavailability, effective therapy and patient

compliance.48

20. Mizumotu T, et.al, (2008), Were formulated fast disintegrating dosage form

containing taste- masked particles of Famotidine. Partial granulations was

found to be an effective and practical way to address content uniformity,

however, oral disintegration time tended to become longer as content

uniformity improved. The disintegration time was improved considerably by

controlling ambient humidity during the compression process (>50%RH).

Furthermore, since the new fast-disintegrating technology made it possible to

use low compression force, there was no change in the structure or dissolution

rate of the taste-masked particles after compression. Therefore, this system can

produce a taste-masked fast-disintegrating tablet with satisfactory attributes.49

21. Jha SK, et.al, (2008), Were formulated melt-in- mouth tablets of Haloperidol

which is widely used neuroleptic. Though haloperidol is well absorbed after

oral dosing, there is a first pass metabolism leading to a reduced

bioavailability of the drug (60-70%) Therefore, the present investigation is

concerned with the development of melt-in-mouth tablets of haloperidol.

Various formulations were prepared incorporating a combination of

superdisintegrants, croscarmellose sodium, sodium starch glycolate, and

crospovidone by direct compression method. The formulated melt-in-mouth

tables were evaluated for various physicochemical parameters, disintegration

time and in vitro drug release. All the formulations had disintegration time less

than 30s and release maximum amount of drug by 12 min.50

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 55 


REVIEW OF LITERATURE

22. Prabhu Namita B., Rao Leena et.al, (2007), Studied the taste masking of

Drotaverine hydrochloride. The drug was treated with cationic exchange

resins, which, by complexation technique masked the bitterness of the drug.

The loading process was optimized for taste masking and drug: resin ratio.

The resinate was evaluated for bulk density, tap density and taste. The

complex was characterized using DSC. The taste masked Drotaverine complex

was incorporated into palatable melt in mouth tablets.51

23. Mukesh C. Gohel, Rajesh K. et.al, (2007), Were prepared and assement of

Novel coprocessed Superdsisintegrant consisting of Crospovidone and Sodium

Starch Glycolate. Corprocessing is defined as combining 2 or more established

excipients by an appropriate process. Coprocessed superdisintegrant consisting

of crospovidone and SSG exhibited good flow and compression

characteristics. Cefixime trihydrate and ibuprofen tablets containing

coprocessed superdisintegrant exhibited quick disintegration and improved

drug dissolution.52

24. Jacob. S, Shirwalkar. A. et.al., (2007) Studied novel co-processed excipients

of Mannitol and Microcrystalline Cellulose for preparing Fast Dissolving

Tablets of Glipizide.Co-processed particles of microcrystalline cellulose and

mannitol were fabricated by spray drying technique to be used as a direct

compression excipient in fast dissolving tablet formulation. Microcrystalline

cellulose passed through sieve no.80, having a volumetric mean diameter (d50)

of 28.35 pm, was used to form composite particles with powdered mannitol

which was previously passed through sieve no. 80, in various mixing ratios

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 56 


REVIEW OF LITERATURE

distributed in the microcrystalline matrix in spray dried form compared to

physical mixture of the same combination. The fast disintegration may be due

to the partial amorphization and formation of submicron particles of mannitol.

Fast dissolving tablets of glipizide were prepared by blending with other

excipients and compressed into tablets.53

25. Dina Nath Mishra, Madhu Bindal et.al, (2006) Studied spray dried

excipients based Technique for the Formulation of Orally Disintegrating

Tablets of Valdecoxib (low aqueous solubility) and Metoclopramide (high

aqueous solubility). Spray dried excipient base was prepared using Scientech

spray drier. Super disintegrants (such as Ac-Di-Sol, Kollidon CL, sodium

starch glycolate.) diluent (mannitol) along with sweeting agent (aspartame)

were used in the formulation of tablets. The tablets were evaluated for

hardness, friability, water absorption ratio, disintegration time (DT) and in

vitro drug release.54

26. Jinichi Fukami, Etsuo Yonemochi et.al, (2006) Evaluated of rapidly

disintegrating tablets containing glycine and carboxymethylcellullose. Effect

of glycine as disintegrant on the disintegration behavior of the tablet in the

oral cavity was evaluated. Wetting time prepared from

carboxymethylecellulose (NS-300) having the hardness of 4kg was 3s. Tablet

containing NS-300 showed fastest disintegration compared to other

formulations. Glycine, one of an amino acid, presents excellent wetting nature

and suitable for the fast disintegrating formulation. Ethenzamide and ascorbic

acid were added to the formulation. And their disintegration behaviour was

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 57 


REVIEW OF LITERATURE

evaluated. Ethenzamide did not affect the disintegration property; however,

ascorbic acid prolonged disintegration time.55

27. Chaudhari P.D., Kolhe S. R., et.al, (2006) Studied the use of cation

exchange resins to optimize the taste masking of bitter drug Rizatriptan

benzoate. The taste should be masked in order to reduce its bitterness to

increase its palatability and also to improve patient compliance. So attempts

were undertaken to mask the bitter taste by complexation technique using ion

exchange resins. Drug release from the complex is obtained at salivary and

gastric pH.56

28. Gohel. M. C. et.al, (2005) Studied a review of co-processed directly

compressible excipients. The present review out-lines the importance of the

functionality of the directly compressible adjuvants in the formulation of

tablets. The co-processing is the most widely explored method for the

preparation of directly compressible adjuvants because it is cost effective and

can be prepared in-house based on the functionality required. Hence, the

present review focuses on the properties of the co-processed directly

compressible adjuvants available in the market.57

29. Na Zhao and Larry L. Augsburger et.al, (2005) Studied functionality

comparison of 3 classes of superdisintegrants in promoting Aspirin tablet

disintegration and dissolution by Ac-Di-Sol, Primojel, and Polyplasdone

XL10. Ac-Di-Sol was found to disintegrate tablets rapidly into apparently

primary particles; Primojel also apparently disintegrated tables into primary

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 58 


REVIEW OF LITERATURE

particles but more slowly; Polyplasdone XL10 disintegrated tablets rapidly but

into larger masses of aggregated particles. The aspirin tablet matrix is

proposed as a model formulation for disintegrate efficiency comparison and

performance consistency testing for quality control purpose.58

30. Abdelbary G., et.al, (2004) Were prepared the orally disintegrating tablets

using a hydrophilic waxy binder (Superpolystate, PEG-6 stearate).

Superpolystate is a waxy material with a melting point of 33-370C and an

HLB value of 9, so it will not only act as a binder and increase the physical

resistance of tables but will also help the disintegration of the tablets as it

melts in the mouth and solubilizes rapidly leaving no residues. The

incorporation of Superpolystate in the formulation of RDT was realized by

means of two different granulation methods: wet granulation by using an

emulsion of this waxy binder as granulating liquid and melt granulation where

the molten form of the binder was used. The potential of the intracranial

addition of croscarmellose sodium as a disintegrating agent was also

evaluated.59

31. Damrongsak Faroongsarng et.al, (2003) Studied the thermal porosity

analysis of croscarmellose sodium and sodium starch glycolate by differential

scanning calorimetry. Croscarmellose sodium (CCS) and sodium starch

glycolate (SSG) were allowed to sorb moisture in 85%, 90%, 95% and 100%

relative humidity (RH) at 400C for 24 hours. The pretreated samples were then

subjected to DSC running temperature ranging from 250C to – 500C at a

cooling rate of 100C/min. The porosity analysis of CCS and SSG was also

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 59 


REVIEW OF LITERATURE

done using nitrogen adsorption as a reference method. The nonfreezable

moisture content was refereed to tightly bound, plasticizing water, whereas the

frozen one may be attributed to loosely bound water condensation in pore

structure of CCS and SSG surfaces.60

32. Toshihiro Shimizu et.al, (2003) Studied the formulation of Lansoprazole

Fast-disintegrating Tablet. III. Design of Rapidly Disintegrating tablets. In the

design of the inactive granules, mannitol was used as a basic excipient,

Microcrystalline cellulose, low-substituted hydroxypropyl cellulose (L-HPC),

and crospovidone were used as binders and disintegrants. A new grade of L-

HPC (L-HPC-33) with a hydroxypropoxy group content of 5.0-6.9% was

developed and it has no rough texture due to decrease in water absorption. It

was clarified that L-HPC 33 could be useful as a binder and disintegrant in

rapidly disintegrating tablets. LFDT contain enteric-coated microgranules in

tablet form.61

33. Toshihiro Shimizu et.al, (2003) Studied the formulation of Lansoprazole fast-

disintegrating tablet-I. Effect of Compression on Dissolution Behaviour.

Lansoprazole is an antiulcer agent and is unstable under acidic conditions; we

have developed LFDT as an orally disintegrating tablet containing enteric-

coated microgranules. The effect of compression on dissolution behavior was

investigated, as compression affected cleavage and crushing of the enteric

layer. To decrease cleavage and crushing of the enteric layer, the effects of the

combined ratio of methacrylic acid copolymer dispersion to ethyl acrylate-

methyl methacrylate copolymer dispersion and the concentration of triethly

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 60 


REVIEW OF LITERATURE

citrate on the dissolution in the acid stage and the dissolution in the buffer

stage were evaluated.62

34. Yutaka Morita et.al, (2002) Evaluated the disintegration time of rapidly

disintegrating tablets via a novel method Utilizing a CCD Camera. Ordinary

disintegration testing, such as the Japanese Pharmacopoeia (JP) method, faces

limitations with respect to the evaluation of rapid disintegration due to strong

agitation. Therefore, we have developed a novel apparatus and method to

determine the dissolution of the RDT. The novel device consists of a

disintegrating bath and CCD camera interfaced with a personal computer

agitation. Therefore, we have developed a novel apparatus and method to

determine the dissolution of the RDT. The novel device consists of a

disintegrating bath and CCD camera interfaced with a personal computer

equipped with motion capture and image analysis software. Simultaneously,

were also able to detect qualitative information.63

35. Hector Fausett et.al., (2000) Evaluated the quick disintegrating Calcium

carbonate tablets by direct compression and compare it with commercially

available calcium tablets, CC tablets were formulated on a Carver press using

3 different forms of CC direct compressed granules ( Cal-Carb 4450, Cal-Carb

4457, and Cal-Carb 4462 ). The dissolution studies showed that all

formulations released 100% of the elemental calcium in simulated gastric fluid

in less than 20 minutes. In summary, this study clearly demonstrated that

quick disintegrating CC tablets can be formulated without expensive

effervescence technology.64

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 61 


REVIEW OF LITERATURE

36. Chaulang G., et.al, (2009), Were prepared solid dispersion of Furosemide in

SSG in ratios of 1:1 and 1:2 by kneading method. The FTIR, DSC, and XRD

showed a change in crystal structure towards an amorphous from of

furosemide. Dissolution data indicated that furosemide dissolution was

enhanced. The sold dispersion formulated in 1:2 ratio showed a 5.40 fold

increase in dissolution and also exhibited superior dissolution characteristics

to commercial furosemide tablets .65

37. Madan J, et.al, (2009), Were prepared fast dissolving tablets of the

nutraceutical, freeze-dried Aloe vera gel by dry granulation method. The

tablets were evaluated for crushing strength, disintegration time, wetting time,

friability, drug content and drug release. A 32 full factorial design was applied

to investigate the combined effect of two formulation variables - amounts of

microcrystalline cellulose and mannitol. The results of multiple regression

analysis revealed that in order to obtain a fast dissolving tablet of the Aloe

vera gel, an optimum concentration of mannitol and a higher content of

microcrystalline cellulose should be used.66

38. Mohapatra A, et.al, (2008), Were prepared the tablets of Metformin using

starch RX1500 and microcrystalline cellulose by direct compression. The

tablets showed erosion behavior rather than disintegration. Then lactose was

incorporated which created pores to cause burst release of drug. But these

tablets did not give good mouth feel. Thus, Pearlitol SD 200 (spray dried

mannitol) was used to prepare tablets by wet granulation (10%

polyvinylpyrrolidone in Isopropyl alcohol as binder). The optimized batches

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 62 


REVIEW OF LITERATURE

of tablets (LMCT3 and MP13) not only exhibited desired mouth feel but also

disintegration time, In-vitro dispersion time, water absorption ratio, and In-

vitro drug release. All the batches contained 15% starch 1500 and 4% of

croscarmellose sodium. The optimized batches prepared by direct compression

and wet granulation showed 85% drug release at 4 min and 8 min.67

39. Mohammad BJ, et.al, (2007), Were prepared Carbamazepine solid

dispersions by the cogrinding technique using an insoluble but highly

hydrophilic crospovidone and soluble hydroxypropylmethylcellulose (HPMC)

as the carriers. The ratios of drug to carrier were 1:1, 1:5 and 1:10.

Comparison of the dissolution of the drug from its cogrounds with that of the

unground drug, its ground form and the corresponding physical mixtures

revealed considerable differences. The percentage of drug dissolved during the

first 30 min, (%D30), for the ground and coground drug was 75-95, whereas

the %D30 for unground drug and its physical mixtures ranged from 41-62.68

40. Fars KA, et.al, (2007), Were formulated Metoclopramide FDT with sufficient

mechanical strength and fast disintegration from bases prepared by both spray

and freeze drying techniques. Different disintegration accelerators were

utilized to prepare the proper FDT using various super-disintegrants (Ac-Di-

Sol, Kollidon and sodium starch glycolate), a volatilizing solvent (ethanol) and

an amino acid (glycine).69

41. Malke S, et.al, (2007), Were prepared fast dissolving tablets of

Oxycarbazepine containing Avicel PH 102 as a diluent and Ac-Di-Sol as a

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 63 


REVIEW OF LITERATURE

superdisintegrant by wet granulation process. All the formulations were

evaluated for characteristics such as hardness, friability, weight variation,

wetting ability, and disintegration time and dissolution rate. The disintegration

time was found to be 28±5 sec and drug release of not less than 90% within 30

min.70

42. Modi A, et.al, (2006), Were investigated enhancement of the dissolution

profile of Valdecoxib using solid dispersion with polyvinylpyrrolidine. They

also described the preparation of fast-dissolving tablets of Valdecoxib by

using high amount of superdisintegrants. A phase solubility method was used

to evaluate the effect of various water-soluble polymers on aqueous solubility

of valdecoxib. The dissolution of valdecoxib improved significantly in solid

dispersion products ( 85% in 5 min). The tablets containing solid dispersion

exhibited better dissolution profile than commercial tablets.71

43. Shirwaikar AA, et.al, (2004), Were formulated Atenolol as fast disintegrating

tablet using three superdisintegrants, croscarmellose sodium (Ac-Di-Sol),

crospovidone (Polyplasdone XL) and sodium starch glycolate (Explotab). All

the superdisintegrants were used at different concentration levels to assess

their efficiency and critical concentration level.72

44. Ravi Kumar et.al, (2009), Were prepared ODTs of Aceclofenac by wet

granulation technique using camphor as subliming agent and sodium starch

glycolate together with crosscarmellose sodium as superdisintegrants. The

camphor was sublime and forms porous granules. All the formulation showed

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 64 


REVIEW OF LITERATURE

low weight variation with dispersion time less than 50 seconds and rapid in-

vitro dissolution (97.4-99.1%). The result revealed that the tablets containing

subliming agent had a good dissolution profile.73

45. D.M.Patel et.al., (2004), Was studies in formulation of ODTs of Rafecoxib by

using three different superdisintegrants croscarmellose sodium, crospovidone

and sodium starch glycolate by wet granulation method, from result it can be

concluded that the tablets containing crospovidone exhibit quick disintegration

time 6 seconds and wetting time 5 seconds.74

46. Manivannan Rangasamy et.al, (2009), Was designed and evaluated the fast

dissolving tablet of Terbutaline sulphate by incorporating superdisintegrats

such as Explotab, Ac-di-sol, and Polyplasdone XL, by direct compression

method. Among all ,the formulation F9 (containing 5 % w/w conc. of

polyplasdone XL) was considered to be the best formulation, which release up

to 99.33% of the drug in 10 min.,and dispersion time 9 seconds.75

47. Sarasija Suresh et.al, (2007), Were prepared mouth dissolving tablet of

Salbutomol sulphate by wet granulation process using sublimable components

camphor and sodium bicarbonate. Evaluation of the tablets showed that all the

tablets were found to be within official limits and disintegration time from 5-

40 seconds. Among all, the formulation containing microcrystalline cellulose

and sodium bicarbonate showed the least disintegration time of 5 seconds.76

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 65 


REVIEW OF LITERATURE

48. H.S.Mahajan et.al, (2004), Have worked on the mouth dissolved tablets of

Sumatriptan succinate were prepared using superdisintegrants sodium starch

glycolate, carboxymethylcellulose, sodium and treated agar by direct

compression method. The tablet disintegrates invitro and in vivo within 10 to

16 second and 12 to 18 seconds respectively. Almost 90% of drug was release

from all the formulations within 10 minutes.77

49. Shailesh Sharma et.al, (2009), Were prepared Fast dissolving tablets of

Promethazine Theoclate by direct compression method using Ac-Di-Sol,

sodium starch glycolate and crospovidone as superdisintegrants in different

concentration. The tablet containing Ac-Di-Sol showed superior organoleptic

properties along with excellent in-vitro disintegration time (52 sec.) and drug

release (72.57%) as compare to other formulations.78

50. A. S. Mundada et.al., (2008), Were formulated dispersible taste masked

tablets of Roxithromycin by using weak cation exchange resins Indion 214

and Amberlite IRP64, polymer Carbapol 934P.The loading process was

optimized for the pH of loading solution and resin or polymer : drug ratio. In-

vitro drug release studies showed more than 80% drug release from the

optimized formulation within 30 min. Amberlite IRP64 was found to be better

complexing agent for masking the bitter taste of Roxithromycin.79

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 66 


REVIEW OF LITERATURE

3.2. DRUG REVIEW

KETOPROFEN 80-85.

Structure

Chemical Name 2-(3-benzoylphenyl)-propionic acid, 3-Benzoyl-α-

methylbenzene acetic acid

Molecular formula C16H14O3

Molecular weight 254.29

Description White or off white, odorless, tasteless, nonhygroscopic,

fine to granular powder.

Melting point 94-97°

Category Anti-inflammatory; analgesic.

Solubility Freely soluble in ethanol, chloroform, acetone, ether

and soluble in benzene and strong alkali. Practically

insoluble in water at 200 C.

Identification:

The infra-red absorption spectrum is concordant with the reference spectrum

of ketoprofen. The light absorption in the range 230 to 360 nm of a 0.002 % w/v

solution in methanol (75%) exhibits a maximum only at about 258 nm; absorbance at

about 258 nm is about 1.3.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 67 


REVIEW OF LITERATURE

Loss on drying:

When dried to constant weight at 600C at a pressure not exceeding 0.7 kPa,

loses not more than 0.5% of its weight. Use 1 g.

Residue on ignition:

Not more than 0.2%

Sulphated ash:

Not more than 0.1%

Assay:

Weigh accurately about 0.5 gm., dissolve in 25 ml of ethanol (96%)

previously neutralized to phenolphthalein solution, add 25 ml of water and titrate with

0.1M sodium hydroxide using phenolphthalein solution as indicator. Each ml of 0.1M

sodium hydroxide is equivalent to 0.02543 gm. of C16H14O3.

Storage:

Store in well-closed container.

Pharmacological profile:

Ketoprofen is an aryl propionic acid derivatives, it is an NSAID with

pronounced analgesic, anti-inflammatory and anti-pyretic action. It may offer

significant advantages over Aspirin and Indomethacin for many patients, since they

usually are better tolerated. Nevertheless, propionic acid derivatives share all the

detrimental features of the entire class of drugs. The pharmacodynamics properties of

the propionic acid derivatives do not differ significantly. All are effective COX

inhibitors, although there is considerable variation in potency.

Mechanism of action:

Ketoprofen is a non-steroidal anti-inflammatory drug with analgesic and

antipyretic properties. During inflammation, pain and fever, arachidonic acid is

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 68 


REVIEW OF LITERATURE

liberated from phospholipid fraction of the cell membrane; arachidonic acid is then

converted via cyclo-oxygenase (COX-1 and 2) pathways to prostaglandins (PGs),

bradykinins, leukotrienes etc, which are the chemical mediators for the above

conditions. Ketoprofen mainly acts by inhibition of both cyclooxygenase-1 and

cyclooxygenase-2 which leads to the inhibition of prostaglandin synthesis and

leukotriene synthesis, to have antibradykinin activity, as well as to have lysosomal

membrane stabilization action. Antipyretic effects may be due to action on the

hypothalamus, resulting in an increased peripheral blood flow, vasodilation and

subsequent heat dissipation.

Figure-6: Mechanism of Action of Ketoprofen

Pharmacokinetic data:-

Oral availability (%): 100%

Metabolism: Hepatic

Excretion (%): Renal 50 - 90%, feces 1 -8 %

Plasma clearance: 1 – 2 ml/min/kg

Volume of distribution (Vd): 0.1 – 0.2 L/kg

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 69 


REVIEW OF LITERATURE

Peak time (h): 0.5 – 2 h.

Peak plasma concentration (mcg/ml): 4.1 mcg/ml

Protein binding (%): 99%

Half-life (h): 2.0 ± 0.3 h

Onset: 30 min

Clinical Pharmacology:

Ketoprofen is a nonsteroidal anti-inflammatory drug with analgesic and

antipyretic properties. In anti-inflammatory models Ketoprofen has been shown to

have inhibitory effects on prostaglandin and leukotriene synthesis, to have antibrady-

kinin activity, as well as to have lysosomal membrane-stabilizing action. However, its

mode of action, like that of other nonsteroidal anti-inflammatory drugs, is not fully

understood.

Ketoprofen is a racemate with only the S enantiomer possessing

pharmacological activity. The enantiomers have similar concentration time curves and

do not appear to interact with one another. An analgesic effect-concentration

relationship for ketoprofen was established in an oral surgery pain study with Orudis.

The effect-site rate constant was estimated to be 0.9 hour (95% confidence limits: 0 to

2.1), and the concentration (Ceg of Ketoprofen that produced one-half the maximum

PID (pain intensity difference) was 0.3 µg/mLs (9596 confidence limits: 0.1 to 0.5).

Thirty-three (33) to 68% of patients had an onset of action (as measured by reporting

some pain relief) within 30 minutes following a single oral dose in postoperative pain

and dysmenorrhea studies. Pain relief (as measured by remedication) persisted for up

to 6 hours in 26 to 7296 of patients in these studies.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 70 


REVIEW OF LITERATURE

Indications:

Ketoprofen is indicated for the management of the signs and symptoms of

rheumatoid arthritis and osteoarthritis.

Adverse effects:

Ketoprofen causes gastrointestinal disturbances such as intestinal bleeding,

epigastric pain, nausea, sensation of fullness in stomach and heartburns. Less

frequently, they may cause CNS symptoms such as headache, dizziness, blurred

vision, tinnitus, urticaria and insomnia. In few cases fluid retention and edema occurs.

Jaundice, impairment of renal function and thrombocytopenia occurs rarely.

Administration and Dose:

Ketoprofen administered orally 50-100 mg twice daily, with food. Ketoprofen

suppository 100 mg once a day.

Contraindication:

• Allergy to aspirin or other NSAID’s.

• History of ulcer disorder.

• Severe impairment of kidney function.

• Bleeding or blood cell disorder.

• Combination of ketoprofen with warfarin is not recommended.

• Combination of ketoprofen with probencid is not recommended.

Uses:

Ketoprofen is used in treatment of rheumatoid arthritis, osteoarthritis,

dysmenorrhea and management of pain.

Brand name:

Orudis, Oruvail, Profenid, Fastum, Ketopron, Kefinid.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 71 


REVIEW OF LITERATURE

3.3. EXCIPIENTS REVIEW

3.3.1. CROSCARMELLOSE SODIUM 86

Synonyms

Ac-Di-Sol; cross-linked carboxymethylcellulose sodium; Explocel.

Chemical Name and CAS Registry Number

Cellulose, carboxymethyl ether, sodium salt, cross-linked [74811-65-7]

Empirical Formula and Molecular Weight

The USP 28 describes carboxymethylcellulose sodium as the sodium salt of a

polycarboxymethyl ether of cellulose. Typical molecular weight is 90 000–700 000.

Applications in Pharmaceutical Formulation or Technology

Croscarmellose sodium is used in oral pharmaceutical formulations as a

disintegrant for capsules, tablets, and granules. In tablet formulations, croscarmellose

sodium may be used in both direct-compression and wet-granulation processes. When

used in wet granulations, the croscarmellose sodium should be added in both the wet

and dry stages of the process (intra- and extragranularly) so that the wicking and

swelling ability of the disintegrant is best utilized. Croscarmellose sodium at

concentrations up to 5% w/w may be used as a tablet disintegrant, although normally

2% w/w is used in tablets prepared by direct compression and 3% w/w in tablets

prepared by a wet-granulation process.

Description

Croscarmellose sodium occurs as an odorless, white or grayish-white powder.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 72 


REVIEW OF LITERATURE

Table-4: Typical Properties of Croscarmellose Sodium.

S. No. Properties Value

1 Density (Bulk) 0.529 g/cm3

2 Density (Tapped) 0.819 g/cm3

3 Density (True) 1.543 g/cm3

4 Solubility Insoluble in water but rapidly swells up to 4-8 times


its original volume and also insoluble acetone,
ethanol and toluene

Stability and Storage Conditions

A model tablet formulation prepared by direct compression, with

croscarmellose sodium as a disintegrant, showed no significant difference in drug

dissolution after storage at 30°C for 14 months. Croscarmellose sodium should be

stored in a well-closed container in a cool, dry place.

Incompatibilities

Croscarmellose sodium is not compatible with strong acids or with soluble

salts of iron and some other metals such as aluminum, mercury, and zinc.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 73 


REVIEW OF LITERATURE

3.3.2. SODIUM STARCH GLYCOLATE 87

Synonyms

Carboxymethyl starch, sodium salt; Explosol; Glycolys.

Chemical Name and CAS Registry Number

Sodium carboxymethyl starch [9063-38-1]

Applications in Pharmaceutical Formulation or Technology

It is widely used in oral pharmaceuticals as a disintegrant in capsule and tablet

formulations. It is commonly used in tablets prepared by either direct-compression or

wet-granulation processes. The usual concentration employed in a formulation is

between 2% and 8%, with the optimum concentration about 4%, although in many

cases 2% is sufficient. Disintegration occurs by rapid uptake of water followed by

rapid and enormous swelling. Although the effectiveness of many disintegrants is

affected by the presence of hydrophobic excipients such as lubricants, the disintegrant

efficiency of sodium starch glycolate is unimpaired. Increasing the tablet compression

pressure also appears to have no effect on disintegration time. Sodium starch

glycolate has also been investigated for use as a suspending vehicle.

Description

Sodium starch glycolate is a white to off-white, odorless, tasteless, free-

flowing powder. The PhEur 2005 states that it consists of oval or spherical granules,

30–100 μm in diameter, with some less-spherical granules ranging from 10–35 μm in

diameter.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 74 


REVIEW OF LITERATURE

Table-5: Typical Properties of Sodium Starch Glycolate

S. No. Properties Value

1 Density (Bulk) 0.756g/cm3

2 Density (Tapped) 0.945 g/cm3

3 Density (True) 1.443 g/cm3

4 Solubility Sparingly soluble in ethanol;

Practically insoluble in water.

Stability and Storage Conditions

Tablets prepared with sodium starch glycolate have good storage properties.

Sodium starch glycolate is stable and should be stored in a well-closed container in

order to protect it from wide variations of humidity and temperature, which may cause

caking. The physical properties of sodium starch glycolate remain unchanged for up

to 3–5 years if it is stored at moderate temperatures and humidity.

Incompatibilities

Sodium starch glycolate is incompatible with ascorbic acid.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 75 


REVIEW OF LITERATURE

3.3.3. CROSPOVIDONE88

Synonyms

Cross-linked povidone; Kollidon CL; Polyplasdone XL.

Chemical Name and CAS Registry Number

1-Ethenyl-2-pyrrolidinone homopolymer [9003-39-8]

Applications in Pharmaceutical Formulation or Technology

Crospovidone is a water-insoluble tablet disintegrant and dissolution agent

used at 1–5% concentration in tablets prepared by direct-compression or wet- and dry-

granulation methods. It rapidly exhibits high capillary activity and pronounced

hydration capacity, with little tendency to form gels. Studies suggest that the particle

size of crospovidone strongly influences disintegration of analgesic tablets. Larger

particles provide a faster disintegration than smaller particles. Crospovidone can also

be used as a solubility enhancer.

Description

Crospovidone is a white to creamy-white, finely divided, free-flowing,

practically tasteless, odorless or nearly odorless, hygroscopic powder.

Table-6: Typical Properties of Crosspovidone

S. No. Properties Value

1 Density 1.22 g/cm3

2 Solubility Insoluble in water and most


common organic solvents

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 76 


REVIEW OF LITERATURE

Stability and Storage Conditions

Since crospovidone is hygroscopic, it should be stored in an airtight container

in a cool, dry place.

Incompatibilities

Crospovidone is compatible with most organic and inorganic pharmaceutical

ingredients. When exposed to a high water level, crospovidone may form molecular

adduct with some materials.

3.3.4. SODIUM BICARBONATE 89

Synonyms

Baking soda; sodium acid carbonate; sodium hydrogen carbonate.

Chemical Name and CAS Registry Number

Carbonic acid monosodium salt [144-55-8]

Empirical Formula and Molecular Weight

NaHCO3 84.01

Structural Formula

NaHCO3

Functional Category

Alkalizing agent; therapeutic agent.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 77 


REVIEW OF LITERATURE

Applications in Pharmaceutical Formulation or Technology

Sodium bicarbonate is generally used in pharmaceutical formulations as a

source of carbon dioxide in effervescent tablets and granules. It is also widely

used to produce or maintain an alkaline pH in a preparation. In effervescent

tablets and granules, sodium bicarbonate is usually formulated with citric and/or

tartaric acid; combinations of citric and tartaric acid are often preferred in

formulations as citric acid alone produces a sticky mixture that is difficult to

granulate, while if tartaric acid is used alone, granules lose firmness. When the

tablets or granules come into contact with water, a chemical reaction occurs,

carbon dioxide is evolved, and the product disintegrates. Tablets may also be

prepared with sodium bicarbonate alone since the acid of gastric fluid is

sufficient to cause effervescence and disintegration. Sodium bicarbonate is also

used in tablet formulations to buffer drug molecules that are weak acids, thereby

increasing the rate of tablet dissolution and reducing gastric irritation.

Description

Sodium bicarbonate occurs as an odorless, white, crystalline powder with

a saline, slightly alkaline taste. The crystal structure is monoclinic prisms.

Grades with different particle sizes, from a fine powder to free-flowing uniform

granules, are commercially available.

Typical Properties

Acidity/alkalinity:

pH = 8.3 for a freshly prepared 0.1 M aqueous solution at 25°C;

alkalinity increases on standing, agitation, or heating.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 78 


REVIEW OF LITERATURE

Density (bulk): 0.869 g/cm3

Density (tapped): 1.369 g/cm3

Density (true): 2.173 g/cm3

Solubility

Practically soluble in water and most common organic solvents.

Stability and Storage Conditions

When heated to about 50°C, sodium bicarbonate begins to dissociate into

carbon dioxide, sodium carbonate, and water; on heating to 250–300°C, for a

short time, sodium bicarbonate is completely converted into anhydrous sodium

carbonate. However, the process is both time- and temperature-dependent, with

conversion 90% complete within 75 minutes at 93°C. The reaction proceeds via

surface-controlled kinetics; when sodium bicarbonate crystals are heated for a

short period of time, very fine needle-shaped crystals of anhydrous sodium

carbonate are formed on the sodium bicarbonate surface.

Incompatibilities

Sodium bicarbonate reacts with acids, acidic salts, and many alkaloidal

salts, with the evolution of carbon dioxide. Sodium bicarbonate can also

intensify the darkening of salicylates. In powder mixtures, atmospheric moisture

or water of crystallization from another ingredient is sufficient for sodium

bicarbonate to react with compounds such as boric acid or alum. In liquid

mixtures containing bismuth subnitrate, sodium bicarbonate reacts with the acid

formed by hydrolysis of the bismuth salt.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 79 


REVIEW OF LITERATURE

3.3.5. CITRIC ACID MONOHYDRATE 90

Synonyms

E330; 2-hydroxypropane-1, 2, 3-tricarboxylic acid monohydrate.

Chemical Name and CAS Registry Number

2-Hydroxy-1, 2, 3-propanetricarboxylic acid monohydrate [5949-29-1]

Empirical Formula and Molecular Weight

C6H8O7·H2O 210.14

Structural Formula

Functional Category

Acidifying agent; antioxidant; buffering agent; chelating agent; flavor

enhancer.

Applications in Pharmaceutical Formulation or Technology

Citric acid (as either the monohydrate or anhydrous material) is widely

used in pharmaceutical formulations and food products, primarily to adjust the

pH of solutions. It has also been used experimentally to adjust the pH of tablet

matrices in enteric-coated formulations for colon-specific drug delivery. Citric

acid monohydrate is used in the preparation of effervescent granules, while

anhydrous citric acid is widely used in the preparation of effervescent tablets.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 80 


REVIEW OF LITERATURE

Citric acid has also been shown to improve the stability of spray-dried insulin

powder in inhalation formulations.

In food products, citric acid is used as a flavor enhancer for its tart, acidic

taste. Citric acid monohydrate is used as a sequestering agent and antioxidant

synergist.

It is also a component of anticoagulant citrate solutions. Therapeutically,

preparations containing citric acid have been used to dissolve renal calculi.

Description

Citric acid monohydrate occurs as colorless or translucent crystals, or as a

white crystalline, efflorescent powder. It is odorless and has a strong acidic taste.

The crystal structure is orthorhombic.

Typical Properties

Acidity/alkalinity: pH = 2.2 (1% w/v aqueous solution)

Density: 1.542 g/cm3

Solubility:

Soluble 1 in 1.5 parts of ethanol (95%) and 1 in less than 1 part of water

sparingly soluble in ether.

Stability and Storage Conditions

Citric acid monohydrate loses water of crystallization in dry air or when

heated to about 40°C. It is slightly deliquescent in moist air. Dilute aqueous

solutions of citric acid may ferment on standing.

The bulk monohydrate or anhydrous material should be stored in airtight

containers in a cool, dry place.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 81 


REVIEW OF LITERATURE

Incompatibilities

Citric acid is incompatible with potassium tartrate, alkali and alkaline

earth carbonates and bicarbonates, acetates, and sulfides. Incompatibilities also

include oxidizing agents, bases, reducing agents, and nitrates. It is potentially

explosive in combination with metal nitrates. On storage, sucrose may crystallize

from syrups in the presence of citric acid.

3.3.6. PEG – 6000 91

Synonyms:

Carbowax; Carbowax Sentry; Lipoxol; Lutrol E; PEG; Pluriol E;

polyoxyethylene glycol.

Chemical Name and CAS Registry Number:

a-Hydro-o-hydroxypoly (oxy-1,2-ethanediyl)

Empirical Formula and Molecular Weight:

HOCH2(CH2OCH2)mCH2OH where m represents the average number of

oxyethylene groups.

Structural Formula:

Functional Category:

Ointment base; plasticizer; solvent; suppository base; tablet and capsule

lubricant.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 82 


REVIEW OF LITERATURE

Applications in Pharmaceutical Formulation or Technology:

Polyethylene glycols can also be used to enhance the aqueous solubility or

dissolution characteristics of poorly soluble compounds by making solid dispersions

with an appropriate polyethylene glycol. Polyethylene glycol grades with molecular

weights of 6000 and above can be used as lubricants, particularly for soluble tablet. In

solid-dosage formulations, higher-molecular-weight polyethylene glycols can enhance

the effectiveness of tablet binders and impart plasticity to granules.(4) However, they

have only limited binding action when used alone, and can prolong disintegration if

present in concentrations greater than 5% w/w. When used for thermoplastic

granulations, (5–7) a mixture of the powdered constituents with 10–15% w/w PEG

6000 is heated to 70–758C. The mass becomes paste like and forms granules if stirred

while cooling. This technique is useful for the preparation of dosage forms such as

lozenges when prolonged disintegration is required.

Description:

The USPNF 23 describes polyethylene glycol as being an addition polymer of

ethylene oxide and water. Polyethylene glycol grades 200–600 are liquids; grades

1000 and above are solids at ambient temperatures. Solid grades (PEG>1000) are

white or off-white in color, and range in consistency from pastes to waxy flakes. They

have a faint, sweet odor. Grades of PEG 6000 and above are available as free-flowing

milled powders.

Typical Properties:

Density:

1.11–1.14 g/cm3 at 258C for liquid PEGs;

1.15–1.21 g/cm3 at 258C for solid PEGs.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 83 


REVIEW OF LITERATURE

Melting point:

55–630C for PEG 6000;

Solubility:

All grades of polyethylene glycol are soluble in water and miscible in all

proportions with other polyethylene glycols (after melting, if necessary) Solid

polyethylene glycols are soluble in acetone, dichloromethane, ethanol (95%),and

methanol; they are slightly soluble in aliphatic hydrocarbons and ether, but insoluble

in fats, fixed oils, and mineral oil.

Stability and Storage Conditions:

Polyethylene glycols are chemically stable in air and in solution, although

grades with a molecular weight less than 2000 are hygroscopic. Polyethylene glycols

do not support microbial growth, and they do not become rancid. Polyethylene glycols

should be stored in well-closed containers in a cool, dry place

Incompatibilities:

All grades can exhibit some oxidizing activity owing to the presence of

peroxide impurities and secondary products formed by autoxidation. Liquid and solid

polyethylene glycol grades may be incompatible with some coloring agents.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 84 


REVIEW OF LITERATURE

3.3.7. PVP K30 92

Synonyms:

E1201; Kollidon; Plasdone; poly[1-(2-oxo-1-pyrrolidinyl)ethylene];

polyvidone; polyvinylpyrrolidone; PVP; 1-vinyl-2-pyrrolidinone polymer.

Chemical Name and CAS Registry Number:

1-Ethenyl-2-pyrrolidinone homopolymer (9003-39-8)

Empirical Formula and Molecular Weight:

(C6H9NO)n 2500–3000000

PVP K 30: 50000

Structural Formula:

Functional Category:

Disintegrant; dissolution aid; suspending agent; tablet binder.

Applications in Pharmaceutical Formulation or Technology:

Although povidone is used in a variety of pharmaceutical formulations, it is

primarily used in solid-dosage forms. In tableting, povidone solutions are used as

binders in wet granulation processes.(2,3) Povidone is also added to powder blends in

the dry form and granulated in situ by the addition of water, alcohol, or

hydroalcoholic solutions. Povidone is used as a solubilizer in oral and parenteral

formulations and has been shown to enhance dissolution of poorly soluble drugs from

solid-dosage forms.(4–6) Povidone solutions may also be used as coating agents.

Povidone is additionally used as a suspending, stabilizing, or viscosity-increasing

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 85 


REVIEW OF LITERATURE

agent in a number of topical and oral suspensions and solutions. The solubility of a

number of poorly soluble active drugs may be increased by mixing with povidone.

Description:

Povidone occurs as a fine, white to creamy-white colored, odorless or almost

odorless, hygroscopic powder. Povidones with K-values equal to or lower than 30 are

manufactured by spray-drying and occur as spheres. Povidone K-90 and higher K-

value povidones are manufactured by drum drying and occur as plates.

Typical Properties:

Acidity/alkalinity: pH = 3.0–7.0 (5% w/v aqueous solution).

Density (bulk): 0.29–0.39 g/cm3 for Plasdone.

Density (tapped): 0.39–0.54 g/cm3 for Plasdone.

Density (true): 1.180 g/cm3

Flowability: 20 g/s for povidone K-15;

16 g/s for povidone K-29/32.

Melting point:

Softens at 1500C.

Moisture content:

Povidone is very hygroscopic, significant amounts of moisture being absorbed

at low relative humidities.

Solubility:

Freely soluble in acids, chloroform, ethanol (95%), ketones, methanol, and

water; practically insoluble in ether, hydrocarbons, and mineral oil. In water, the

concentration of a solution is limited only by the viscosity of the resulting solution,

which is a function of the K-value.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 86 


REVIEW OF LITERATURE

Viscosity (dynamic):

The viscosity of aqueous povidone solutions depends on both the

concentration and the molecular weight of the polymer employed. PVP K 30

Viscosity (dynamic) is 5.5–8.5

Stability and Storage Conditions

Povidone darkens to some extent on heating at 1508C, with a reduction in

aqueous solubility. It is stable to a short cycle of heat exposure around 110–1308C.

Povidone may be stored under ordinary conditions without undergoing decomposition

or degradation. However, since the powder is hygroscopic, it should be stored in an

airtight container in a cool, dry place.

Incompatibilities

Povidone is compatible in solution with a wide range of inorganic salts,

natural and synthetic resins, and other chemicals. It forms molecular adducts in

solution with sulfathiazole, sodium salicylate, salicylic acid, phenobarbital, tannin,

and other compounds. The efficacy of some preservatives, e.g. thimerosal, may be

adversely affected by the formation of complexes with povidone.

3.3.8. CELLULOSE, MICROCRYSTALLINE 93

Synonyms

Avicel PH; Celex; cellulose gel; Celphere; Ceolus KG; crystalline cellulose;

E460; Emcocel; Ethispheres; Fibrocel; Pharmacel; Tabulose; Vivapur.

Chemical Name and CAS Registry Number

Cellulose [9004-34-6]

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 87 


REVIEW OF LITERATURE

Empirical Formula and Molecular Weight

(C6H10O5)n = 36000.

Where n = 220.

Structural Formula

Functional Category

Adsorbent; suspending agent; tablet and capsule diluent; tablet disintegrant.

Applications in Pharmaceutical Formulation or Technology

Microcrystalline cellulose is widely used in pharmaceuticals, primarily as a

binder/diluent in oral tablet and capsule formulations where it is used in both wet-

granulation and direct-compression processes. In addition to its use as a

binder/diluent, microcrystalline cellulose also has some lubricant and disintegrant

properties that make it useful in tableting.

Microcrystalline cellulose is also used in cosmetics and food products.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 88 


REVIEW OF LITERATURE

Description

Microcrystalline cellulose is purified, partially depolymerized cellulose that

occurs as a white, odorless, tasteless, crystalline powder composed of porous

particles. It is commercially available in different particle sizes and moisture grades

that have different properties and applications.

Typical Properties

Angle of repose:

• 49° for Ceolus KG;

• 34.4° for Emcocel 90M.

Density (bulk):

3
• 0.337 g/cm ;

3
• 0.32 g/cm for Avicel PH-101;

3
• 0.29 g/cm for Emcocel 90M;

3
• 0.29 g/cm for VivaPur 101.

Density (tapped):

3
• 0.478 g/cm ;

3
• 0.45 g/cm for Avicel PH-101;

3
• 0.35 g/cm for Emcocel 90M.

Density (true):

1.512–1.668 g/cm3

Flowability:

1.41 g/s for Emcocel 90M.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 89 


REVIEW OF LITERATURE

Solubility:

Slightly soluble in 5% w/v sodium hydroxide solution; practically insoluble in

water, dilute acids, and most organic solvents.

Stability and Storage Conditions

Microcrystalline cellulose is a stable though hygroscopic material. The bulk

material should be stored in a well-closed container in a cool, dry place.

Incompatibilities

Microcrystalline cellulose is incompatible with strong oxidizing agents.

3.3.9. ANHYDROUS LACTOSE 94

Synonyms

Pharmatose DCL 22; saccharum lactis; Super-Tab Anhydrous.

Chemical Name and CAS Registry Number

O-β-D-galactopyranosyl-(1→4)-β-D-glucopyranose [63-42-3]

Empirical Formula and Molecular Weight

C12H22O11 , 342.30

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 90 


REVIEW OF LITERATURE

Structural Formula

The PhEur 2005 describes anhydrous lactose as O-β-D-galactopyranosyl-

(1→4)-β-D-glucopyranose; or a mixture of O-β-D-galactopyranosyl-(1→4)-α-D-

glucopyranose and O-β-D-galactopyranosyl-(1→4)-β-D-glucopyranose. The

USPNF 23 describes anhydrous lactose as being primarily β-lactose or a mixture

of α- and β-lactose. The JP 2001 describes anhydrous lactose as β-lactose or a

mixture of β-lactose and α-lactose.

Functional Category

Binding agent; directly compressible tableting excipient; lyophilization

aid; tablet and capsule filler.

Applications in Pharmaceutical Formulation or Technology

Anhydrous lactose is widely used in direct compression tableting

applications and as a tablet and capsule filler and binder. Anhydrous lactose can

be used with moisture-sensitive drugs due to its low moisture content.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 91 


REVIEW OF LITERATURE

Description

Lactose occurs as white to off-white crystalline particles or powder.

Several different brands of anhydrous lactose are commercially available which

contain anhydrous β-lactose and anhydrous α-lactose. Anhydrous lactose

typically contains 70–80% anhydrous β-lactose and 20–30% anhydrous α-

lactose.

Typical Properties

Density (true): 1.589 g/cm3 for anhydrous β-lactose;

Density (bulk):0.68 g/cm3 for Pharmatose DCL 21;

Density (tapped):0.88 g/cm3 for Pharmatose DCL 21;

Solubility

Soluble in water; sparingly soluble in ethanol (95%) and ether.

Stability and Storage Conditions

Mold growth may occur under humid conditions (80% RH and above).

Lactose may develop a brown coloration on storage, the reaction being

accelerated by warm, damp conditions. At 80°C and 80% RH, tablets containing

anhydrous lactose have been shown to expand 1.2 times after one day.

Lactose anhydrous should be stored in a well-closed container in a cool, dry

place.

Incompatibilities

Lactose anhydrous is incompatible with strong oxidizers. When mixtures

containing a hydrophobic leukotriene antagonist and anhydrous lactose or lactose

monohydrate were stored for six weeks at 40°C and 75% RH, the mixture

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 92 


REVIEW OF LITERATURE

containing anhydrous lactose showed greater moisture uptake and drug

degradation.

Studies have also shown that in blends of roxifiban acetate (DMP-754)

and lactose anhydrous, the presence of lactose anhydrous accelerated the

hydrolysis of the ester and amidine groups.

3.3.10 DEXTROSE 95

Synonyms

Corn sugar; D-(+)-glucopyranose monohydrate; grape sugar;

Chemical Name and CAS Registry Number

D-(+)-Glucose monohydrate [5996-10-1]

Empirical Formula and Molecular Weight

C6H12O6·H2O 198.17 (for monohydrate)

Structural Formula

Anhydrous material shown.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 93 


REVIEW OF LITERATURE

Functional Category

Tablet and capsule diluent; therapeutic agent; tonicity agent; sweetening

agent.

Applications in Pharmaceutical Formulation or Technology

Dextrose is widely used in solutions to adjust tonicity and as a

sweetening agent. Dextrose is also used as a wet granulation diluent and binder,

and as a direct-compression tablet diluent and binder, primarily in chewable

tablets. Although dextrose is comparable as a tablet diluent to lactose, tablets

produced with dextrose monohydrate require more lubrication, are less friable,

and have a tendency to harden. The mildly reducing properties of dextrose may

be used when tableting to improve the stability of active materials that are

sensitive to oxidation.

Description

Dextrose occurs as odorless, sweet-tasting, colorless crystals or as a white

crystalline or granular powder. The JP 2001 describes dextrose as dextrose

anhydrous; the PhEur 2005 specifies dextrose as either dextrose anhydrous or

dextrose monohydrate; and the USP 28 specifies dextrose as dextrose

monohydrate.

Typical Properties

Acidity/alkalinity: pH = 3.5–5.5 (20% w/v aqueous solution)

Density (bulk):0.826 g/cm3

Density (tapped):1.020 g/cm3

Density (true):1.54 g/cm3

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 94 


REVIEW OF LITERATURE

Solubility:

Soluble in water; sparingly soluble in ethanol (95%) and ether.

Stability and Storage Conditions

Dextrose has good stability under dry storage conditions. Aqueous

solutions may be sterilized by autoclaving. However, excessive heating can

cause a reduction in pH and caramelization of solutions.

The bulk material should be stored in a well-closed container in a cool,

dry place.

Incompatibilities

Dextrose solutions are incompatible with a number of drugs such as

cyanocobalamin, kanamycin sulfate, novobiocin sodium, and warfarin sodium.

Erythromycin gluceptate is unstable in dextrose solutions at a pH less than 5.05.

Decomposition of B-complex vitamins may occur if they are warmed with

dextrose.

In the aldehyde form, dextrose can react with amines, amides, amino

acids, peptides, and proteins. Brown coloration and decomposition occur with

strong alkalis.

3.3.11 MAGNESIUM STEARATE 96

Synonyms

Magnesium octadecanoate; octadecanoic acid, magnesium salt; stearic acid,

magnesium salt.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 95 


REVIEW OF LITERATURE

Chemical Name and CAS Registry Number

Octadecanoic acid magnesium salt [557-04-0]

Empirical Formula and Molecular Weight

C36H70MgO4 ,591.34

The USPNF 23 describes magnesium stearate as a compound of magnesium

with a mixture of solid organic acids that consists chiefly of variable proportions of

magnesium stearate and magnesium palmitate (C32H62MgO4). The PhEur 2005

describes magnesium stearate as a mixture of magnesium salts of different fatty acids

consisting mainly of stearic acid and palmitic acid and in minor proportions other

fatty acids.

Structural Formula

[CH3(CH2)16COO]2Mg

Functional Category

Tablet and capsule lubricant.

Applications in Pharmaceutical Formulation or Technology

Magnesium stearate is widely used in cosmetics, foods, and pharmaceutical

formulations. It is primarily used as a lubricant in capsule and tablet manufacture at

concentrations between 0.25% and 5.0% w/w. It is also used in barrier creams.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 96 


REVIEW OF LITERATURE

Description

Magnesium stearate is a very fine, light white, precipitated or milled,

impalpable powder of low bulk density, having a faint odor of stearic acid and a

characteristic taste. The powder is greasy to the touch and readily adheres to the skin.

Typical Properties

Crystalline forms: high-purity magnesium stearate has been isolated as a

trihydrate, a dihydrate, and an anhydrate.

Density (bulk): 0.159 g/cm3

Density (tapped): 0.286 g/cm3

Density (true): 1.092 g/cm3

Stability and Storage Conditions

Magnesium stearate is stable and should be stored in a well-closed container in

a cool, dry place.

Incompatibilities

Incompatible with strong acids, alkalis, and iron salts. Avoid mixing with

strong oxidizing materials. Magnesium stearate cannot be used in products containing

aspirin, some vitamins, and most alkaloidal salts.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 97 


REVIEW OF LITERATURE

3.3.12. TALC 97

Synonyms

Altalc; E553b; hydrous magnesium calcium silicate; hydrous magnesium

silicate; Luzenac Pharma; magnesium hydrogen metasilicate; Magsil Osmanthus;

Magsil Star; powdered talc; purified French chalk; Purtalc; soapstone; steatite;

Superiore.

Chemical Name and CAS Registry Number

Talc [14807-96-6]

Empirical Formula and Molecular Weight

Talc is a purified, hydrated, magnesium silicate, approximating to the formula

Mg6(Si2O5)4(OH)4. It may contain small, variable amounts of aluminum silicate and

iron.

Structural Formula

Mg6 (Si2O5)4(OH)4.

Functional Category

Anticaking agent; glidant; tablet and capsule diluents; tablet and capsule

lubricant.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 98 


REVIEW OF LITERATURE

Applications in Pharmaceutical Formulation or Technology

¾ Talc was once widely used in oral solid dosage formulations as a lubricant and

diluent, although today it is less commonly used. However, it is widely used as

a dissolution retardant in the development of controlled-release products. Talc

is also used as a lubricant in tablet formulations; in a novel powder coating for

extended-release pellets; and as an adsorbent.

¾ In topical preparations, talc is used as a dusting powder, although it should not

be used to dust surgical gloves;. Talc is a natural material; it may therefore

frequently contain microorganisms and should be sterilized when used as a

dusting powder;

¾ Talc is additionally used to clarify liquids and is also used in cosmetics and

food products, mainly for its lubricant properties.

Description

Talc is a very fine, white to grayish-white, odorless, impalpable, unctuous,

crystalline powder. It adheres readily to the skin and is soft to the touch and free from

grittiness.

Typical Properties

Acidity/alkalinity: pH = 7–10 for a 20% w/v aqueous dispersion.

Hardness (Mohs): 1.0–1.5

Solubility:

Practically insoluble in dilute acids and alkalis, organic solvents, and water.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 99 


REVIEW OF LITERATURE

Stability and Storage Conditions

Talc is a stable material and may be sterilized by heating at 160°C for not less

than 1 hour. It may also be sterilized by exposure to ethylene oxide or gamma

irradiation.

Talc should be stored in a well-closed container in a cool, dry place.

Incompatibilities

Incompatible with quaternary ammonium compounds.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 100 


METHODOLOGY

CHAPTER-4.

METHODOLOGY

4.1 MATERIALS

Table 7. List of Materials

S. No. Name Manufacturer


1 Ketoprofen Concept Pharmaceuticals, Aurangabad.

2 Polyvinylpyrollidone K-30 M/s Healer’s Lab.Pvt.Ltd. Baddi.


3 Polyvinylglycol-6000 S.D.Fine Pvt.Ltd., Mumbai
4 Ac-Di-Sol M/s Healer’s Lab.Pvt.Ltd. Baddi
5 Sodium Starch Glycolate M/s Healer’s Lab.Pvt.Ltd. Baddi
6 Crospovidone M/s Healer’s Lab.Pvt.Ltd. Baddi
7 Sodium Bicarbonate S.D.Fine Pvt.Ltd., Mumbai
8 Citric Acid S.D.Fine Pvt.Ltd., Mumbai
9 Avicel PH 102 M/s Healer’s Lab.Pvt.Ltd. Baddi
10 Lactose M/s Healer’s Lab.Pvt.Ltd. Baddi
11 Dextrose M/s Healer’s Lab.Pvt.Ltd. Baddi
12 Magnesium Stearate S.D.Fine Pvt.Ltd., Mumbai
13 Talc S.D.Fine Pvt.Ltd., Mumbai
14 Cardamom flavor Chemwell Pvt.Ltd.,Bangalore
15 Sodium Phosphate S.D.Fine Pvt.Ltd., Mumbai
16 Sodium Hydroxide S.D.Fine Pvt.Ltd., Mumbai
17 Methanol LR S.D. Fine Chem. Ltd., Mumbai
18 Acetone LR S.D. Fine Chem. Ltd., Mumbai
19 Aluminium Foil Hindalco Industries Ltd., Silvasa

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 101 


METHODOLOGY

4.2. EQUIPMENT:

Table 8: List of Equipment Used

SR.
EQUIPMENTS COMPANY NAME
NO.
UV/VIS Double beam
1 Shimadzu 1700, Japan
Spectrophotometer
FTIR
2 Jasco FTIR 6100 type-A, Japan
Magnetic Stirrer
3 Remi Instrument Pvt Ltd., Mumbai
USP Tablet dissolution apparatus
4 Lab India Disso 2000
type II
10 Station rotary punch tableting
5 Riemek Minipress, Ahmedabad
machine
Hot air oven
6 EIE Instrument Pvt. Ltd., Ahmedabad
pH meter
7 Eutech, Singapur
Digital Weighing balance
8 Adair Dutt Instrument Pvt. Ltd.
Hardness tester
9 Pfizer hardness tester, Mumbai
Friability tester
10 Veego friability tester, Mumbai
Tapped density tester
11 Electro lab
Stability apparatus
12 Paramount

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 102 


METHODOLOGY

4.3 DRUG IDENTIFICATION TESTS: 98

I. Physical Appearance:

Physical appearance of drug was examined by various organoleptic properties.

II. Melting Point:

Melting point of the Ketoprofen was determined by capillary fusion method;

one sided closed capillary filled with drug and put into the Melting Point Apparatus.

Temperature was noted at which solid drug changed into liquid.

III. Infrared Spectral Assignment:

The pellet of approximately 01 mm diameter of the drug was prepared

grinding 3-5 mg of sample with 100-150 mg of Potassium Bromide using hydrostatic

press. The sample pellet was mounted in IR compartment and scanned at wavelength

4000 cm-1 – 500 cm-1. On analysis of the IR spectra of the reference spectra (A) given

in Indian Pharmacopoeia (1996) and pure drug (B), no major differences were

observed in the characteristic absorption peak (1696, 1655) pattern. The results were

shown in Figure 10 (A) and 11 (B).

IV. Differential Scanning Calorimetry (DSC):

DSC analysis was performed on 5 mg sample. Samples were heated in an open

aluminum pans at a rate of 100 per min–1 in a 30 to 3000C temperature range under a

nitrogen flow of 40 mL/min. It shows endothermic peak at about 94.120C Figure 12.

V. Ultraviolet Absorption Maxima:

Ultraviolet absorption in the rage 200 to 400 nm of a 5μg/ml solution in 5%

(v/v) methanolic Sorenson’s Buffer (pH 6.8) was measured. The absorption maxima

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 103 


METHODOLOGY

(λmax) of Ketoprofen (5 µg/ml) in this solution was found to be 260 nm which is

concordant with the Indian Pharmacopoeia (1996) shown in Figure 13.

4.4. PREFORMULATION STUDIES: 99

4.4.1 Solubility:

The solubility of Ketoprofen was determined in different solvent systems and

buffers. An excess quantity of the drug was mixed with 10 ml of each solvent in

screw capped glass tubes and shaken on constant water bath shaker for 24 hours at

25o. The solutions were examined physically for the absence or presence of drug

particles and also by spectrophometrically for quantitative determination of drug in

buffers. The results were shown in Table 22.

4.4.2 Preparation of Calibration Curve:

(a) Preparation of Sorenson’s Buffer (pH 6.8):

24.5 ml of 0.2 M dibasic sodium phosphate and 25.5 ml of 0.2 M monobasic

sodium phosphate was placed in 100 ml volumetric flask and make up the volume 100

ml with water.

(b) Calibration Curve:

50 mg of Ketoprofen was weighed accurately and dissolved in 5 ml of

methanol in a 100 ml of volumetric flask and volume was made up to 100 ml with the

Sorenson’s buffer (pH 6.8). 10 ml of this solution was diluted with 100 ml Sorenson’s

buffer (pH 6.8) to obtain a stock solution of 50µg/ml. From this stock solution,

aliquots of 1 ml, 2 ml, 3 ml, 4 ml and 5 ml were taken transferred to 10 ml volumetric

flask and volume was made up to 10 ml with Sorenson’s buffer (pH 6.8). The

absorbance of these solutions was measured at 260 nm against a blank Sorenson’s

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 104 


METHODOLOGY

buffer (pH 6.8). The calibration curve was plotted between concentration and

absorbance.

4.4.3 Drug Polymer Interaction Studies:

The infrared absorption spectra of pure polymer and physical mixture of

polymer and drug were performed for polymer drug interaction studies. Fourier

Transform Infrared spectra were recorded on samples prepared in potassium bromide

(KBr) disks. Samples were prepared in KBr disks by means of a hydrostatic press.

The scanning range was 400 to 4000 cm–1 and the resolution was 4 cm–1. The IR

spectra of physical mixture of polymers and drug were shown in Figure 15-20.

4.5. PREPARATION OF PHYSICAL MIXTURES OF KETOPROFEN:

Physical mixtures of Ketoprofen were prepared using Polyvinylpyrollidone k-

30 and Polyvinylglycol-6000 as a carrier in a weight ratio. First drug and carrier were

passed through a 40 mesh screen and then weighed and mixed by using motor and

pestle (Table 9 & 10).

Table-9: Composition of Ketoprofen-PVP K-30 Physical Mixture.

Sr.No. Formulation Drug : Carrier


Number Weight Ratio
1 KP1 1:1
2 KP2 1:2
3 KP3 1:3

Table-10: Composition of Ketoprofen-PEG-6000 Physical Mixture.

Sr.No. Formulation Drug : Carrier


Number Weight Ratio
1 KPG1 1:1
2 KPG2 1:2
3 KPG3 1:3

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 105 


METHODOLOGY

4.6. PREPARATION OF SOLID DISPERSIONS OF KETOPROFEN:100

Ketoprofen solid dispersions were prepared by solvent evaporation method

using carriers (i.e. PVP K-30, PEG-6000) in proportions, viz. 1:1, 1:2, 1:3 (Drug:

Carrier). Methanol is selected as common solvent for solid dispersion. The respective

amount of carrier was dissolved in methanol 20 ml and ketoprofen was added in parts

with continuous stirring. The solvent was then removed by evaporation. The prepared

solid dispersion were pulverized and shifted through sieve no. 100 and stored over a

fused calcium chloride in a desiccator for further use.

Table-11: Composition of Ketoprofen-PVP K-30 Solid Dispersions.

Sr.No. Formulation Drug : Carrier


Number Weight Ratio
1 KPVP1 1:1
2 KPVP2 1:2
3 KPVP3 1:3

Table-12: Composition of Ketoprofen-PEG-6000 Solid Dispersions.

Sr.No. Formulation Drug : Carrier


Number Weight Ratio
1 KPEG1 1:1
2 KPEG2 1:2
3 KPEG3 1:3

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 106 


METHODOLOGY

4.7. EVALUATION OF PHYSICAL MIXTURES AND SOLID DISPERSIONS:


101

The prepared physical mixtures and solid dispersions were evaluated for

solubility studies, percent drug content, dissolution efficiency, in-vitro drug release

and Fourier transform infrared (FTIR), Differential scanning calorimetry (DSC), X-

ray diffraction (XRD), scanning electron microscopy (SEM),

4.7.1. . Determination of Solubility of Solid Dispersions:

Ketoprofen, physical mixtures or solid dispersions equivalent to 10 mg of

Ketoprofen were added to 10 ml of Sorenson’s buffer pH 6.8 in a 10 ml volumetric

flask. The volumetric flasks were capped properly and shaken at 250 and 370 C in a

temperature controlled water bath (Shaking water bath) for 48 h. Resultant samples

containing undissolved solid dispersions suspended in the volumetric flask were

filtered through 0.45μm filters, suitably diluted with Sorenson’s buffer pH 6.8 and

analyzed by UV spectrophotometer at 260 nm.

4.7.2 Determination of Drug Content:

Drug content was calculated by dissolving solid dispersions equivalent to 100

mg Ketoprofen in 10 ml of methanol, filtered using 0.45μm Whatman filter paper,

suitably diluted with Sorenson’s buffer (pH 6.8) and analyzed by using UV

spectrophotometer against Sorenson’s buffer as blank.

4.7.3 In-vitro Drug Release:

Accurately weighed preparations equivalent to 100 mg of Ketoprofen were

added to 900 ml of dissolution medium in USP II Paddle type apparatus and stirred at

speed of 50 rpm at 37 ± 0.50 C. 5 ml aliquots were withdrawn at 5, 10, 15, 30, 45, 60

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 107 


METHODOLOGY

minutes and replaced by 5 ml of fresh dissolution media. The collected samples were

analyzed after filtration and dilution at 260 nm using UV-visible spectrophotometer

against the blank. Drug release studies were carried out in triplicate. The dissolution

of pure Ketoprofen was done similarly. The release profile data was analyzed for

cumulative percent dissolved at different time intervals and for dissolution efficiency

at 15 and 30 minutes.

4.7.4. Fourier Transform Infrared Spectroscopy:

Fourier Transform Infrared spectra were recorded on samples prepared in

potassium bromide (KBr) disks. Samples were prepared in KBr disks by means of a

hydrostatic press. The scanning range was 400 to 4000 cm–1 and the resolution was 4

cm–1.

4.7.5. Differential Scanning Calorimetry Analysis:

Differential Scanning Calorimetry analysis was performed on 5 mg samples.

Samples were heated in an open aluminum pans at a rate of 100 per min–1 in a 30 to

300°C temperature range under a nitrogen flow of 40 mL/min.

4.7.6. X- Ray Diffraction (XRD):

XRD patterns were recorded using Philips PW 1729 X-ray generator. Powder

X-ray diffraction patterns were traced for Ketoprofen, various carriers and solid

dispersions.

4.7.7. Scanning Electron Microscopy (SEM):

The external morphology of solid dispersions was analyzed by Scanning

Electron Microscope (SEM). The samples were examined under a scanning electron

microscope JSM 6100 JEOL JAPAN.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 108 


METHODOLOGY

4.8. FORMULATION OF FAST DISSOLVING TABLETS:

Fast dissolving tablets containing selected solid dispersion were prepared by

direct compression method using single punch tablet machine to produce convex

faced tablets weighing 500 mg each with a diameter of 11 mm. A minimum of 100

tablets were prepared for each batch. The formulations were developed by using

different techniques.

4.8.1 By Addition of Super Disintegrants 102

The superdisintegrants (Croscarmallose sodium, Sodium starch glycolate and

Crospovidone) in varying concentration (1-5%) were used to develop the tablets. All

the ingredients were shown in Table 13-15 were passed through sieve no. 60 and were

co- grounded in a glass pestle motor. These blends were evaluated for mass-volume

relationship (Bulk Density, Tapped Density, Hausners Ratio, Compressibility Index)

and flow properties (Angle of Repose). The mixed blend of excipients was

compressed using a single punch tablet machine (Cadmach, Ahmedabad) to produce

convex faced tablets weighing 500 mg each with a diameter of 11 mm.

Table-13: Formulation of Fast Dissolving Tablet Using Croscarmallose sodium.

INGRADIENT F1 F2 F3 F4 F5
KPVP3 200 200 200 200 200
Crosscarmalose 5 10 15 20 25
sodium
Lactose 70 70 70 70 70
Dextrose 70 70 70 70 70
Avicel PH 102 135 130 125 120 115
Talc 10 10 10 10 10
Mg. Stearate 10 10 10 10 10
Cardamom QS QS QS QS QS
flavor

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 109 


METHODOLOGY

Table-14: Formulation of Fast Dissolving Tablet Using Sodium Starch Glycolate.

INGRADIENT F6 F7 F8 F9 F10
KPVP3 200 200 200 200 200
Sodium starch 5 10 15 20 25
glycolate
Lactose 70 70 70 70 70
Dextrose 70 70 70 70 70
Avicel PH 102 135 130 125 120 115
Talc 10 10 10 10 10
Mg. Stearate 10 10 10 10 10
Cardamom QS QS QS QS QS
flavor

Table-15: Formulation of Fast Dissolving Tablet Using Crospovidone.

INGRADIENT F11 F12 F13 F14 F15


KPVP3 200 200 200 200 200
Crospovidone 5 10 15 20 25
Lactose 70 70 70 70 70
Dextrose 70 70 70 70 70
Avicel PH 102 135 130 125 120 115
Talc 10 10 10 10 10
Mg. Stearate 10 10 10 10 10
Cardamom QS QS QS QS QS
flavor

4.8.2 By Effervescence Technology

Fast dissolving tablets were prepared by using Citric acid and Sodium

bicarbonate in combination in (2:3 ratio) with other excipients shown in Table 16 was

co-grounded in glass pestle and mortar. These tablets contain (1-5%) effervescent

agent in various proportion. These blends were evaluated for mass-volume

relationship (Bulk Density, Tapped Density, Hausners Ratio, Compressibility Index)

and flow properties (Angle of Repose). The mixed blends of excipient were

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 110 


METHODOLOGY

compressed using a single punch machine (Cadmach, Ahmedabad) to produce convex

faced tablets weighing 500 mg each with a diameter of 11 mm.

Table-16: Formulation of Fast Dissolving Tablet Using Effervescent Agents

INGRADIENT F16 F17 F18 F19 F20


KPVP3 200 200 200 200 200
Citric acid 2 4 6 8 10
Sodium 3 6 9 12 15
bicarbonate
Lactose 70 70 70 70 70
Dextrose 70 70 70 70 70
Avicel PH 102 135 130 125 120 115
Talc 10 10 10 10 10
Mg. Stearate 10 10 10 10 10
Cardamom QS QS QS QS QS
flavor

4.9. CHARACTERIZATION OF BLENDS 103

The quality of tablet, once formulated by rule, is generally dictated by the

quality of physicochemical properties of blends. There are many formulations and

process variables involved in mixing step and all these can affect the characteristics of

blend produced. The characterization of mixed blend done for the flow property of

powder that are bulk density, tapped density, Hausners ratio, Compressibility index,

angle of repose. The various characteristics of blends tested are given below and

results were shown in Table 32.

4.9.1 Bulk Density

Apparent bulk density (ρb) was determined by pouring the blend into a

graduated cylinder. The bulk volume (Vb) and weight of powder (M) was determined.

The bulk density was calculated using the formula.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 111 


METHODOLOGY

M
ρb =
Vb

4.9.2 Tapped Density

The measuring cylinder containing a known mass of blend was tapped 100

times using density apparatus. The minimum volume (Vt) occupied in the cylinder

and the weight (M) of the blend was measured. The tapped density (ρt) was calculated

using the formula.

M
ρt =
Vt

4.9.3 Compressibility Index

The simplest way for measurement of flow of powder is its compressibility, an

indication of the ease with which a material can be induced to flow is given by

compressibility index (I) which is calculated as follows

ρt − ρb
I= × 100
ρt

Where, ρt = Tapped density

ρb = Bulk density

Table-17: Compressibility Index as an Indication of Powder Flow Properties

Carr's Index (%) Type of flow


>12 Excellent
12.0-16 Good
18-21 Fair to passable
23-35 Poor
33-38 Very poor
>40 Extremely poor

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 112 


METHODOLOGY

4.9.4 Hausner Ratio

Hausner ratio (HR) is an indirect index of ease of powder flow. It is calculated

by the following formula.

ρt
Hr =
ρb

Where, ρt is tapped density and ρb is bulk density.

Lower Hausner ratio (< 1.25) indicates better flow properties than higher ones (>

1.25).

4.9.5 Angle of Repose

Angle of Repose was determined using funnel method. The blend was poured

through a funnel that can be raised vertically until a specified cone height (h) was

obtained. Radius of the heap (r) was measured and angle of repose (θ) was calculated

using the formula.

h ⎛h⎞
tan θ = ; Therefore; θ = tan −1 ⎜ ⎟
r ⎝r⎠

Where, θ is angle of repose; h is height of cone; r is radius of cone

Table-18: Angle of Repose as an Indication of Powder Flow Properties.

Angle of repose(o) Type of flow


<25 Excellent
25-30 Good
30-40 Passable
>40 Very poor

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 113 


METHODOLOGY

4.10. CHARACTERIZATION OF FAST DISSOLVING TABLETS 104

After compression of powder, the tablets were evaluated for organoleptic

characteristics like color, odor, taste, diameter, thickness and physical characteristics

like hardness, friability, disintegration time, wetting time, dispersion time and

dissolution studies. The results were shown in Table 33-34.

4.10.1 General Appearance

Visual identification and over all ‘elegance’ were performed such as color,

presence or absence of an odour, taste, surface texture and physical flaws.

4.10.2 Tablet Thickness

Tablet thickness is an important characteristic in reproducing appearance and

also in counting by suing filling equipment. Some filling equipment utilizes the

uniform thickness of the tablets as a counting mechanism. Ten tablets were taken and

their thickness was recorded using micrometer (Mityato, Japan).

4.10.3 Uniformity of Weight

As per IP, twenty tablets were taken and weighted individually and

collectively using digital balance. The average weight of one tablet was calculated.

The weight variation test would be satisfactory method of determining the drug

content uniformity. Data are given in Table 19.

Table-19: Weight Variation Limits for Tablets as per IP.

Average of Tablets (mg) Maximum % difference allowed


130 or less 10
130-324 7.5
More than 324 5

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 114 


METHODOLOGY

4.10.4 Hardness

Hardness of the tablet of each formulation was determined using Pfizer

hardness tester.

4.10.5 Friability

Friability of the tablets was determined using Roche friabilator. This device

subjects the tablets to the combined effect of abrasions and shock in a plastic chamber

revolving at 25 rpm and dropping the tablets at a height of 6 inch in each revolution.

Preweighed sample of tablets was placed in the friabilator and were subjected to 100

revolutions. Tablets were dedusted using a soft muslin cloth and reweighed. The

friability (F %) is determined by the formula.

⎛ Wo ⎞
F % = ⎜1 − ⎟ X 100
⎝ W ⎠

Where, W0 is initial weight of the tablets before the test and W is the weight of

the tablets after test.

4.9.6 Disintegration Test

Disintegration of fast disintegrating tablets is achieved by saliva in the mouth,

however amount of saliva in the mouth is limited and no tablet disintegration test was

found in USP and IP to simulate in vivo conditions. A modified method was used to

determine disintegration time of the tablets. A cylindrical vessel was used in which

10-mesh screen was placed in such way that only 2 ml of disintegrating or dissolution

medium would be placed below the sieve (Figure 7). To determine disintegration

time, 6 ml of Sorenson’s buffer (pH 6.8), was placed inside the vessel in such way

that 4 ml of the media was below the sieve and 2 ml above the sieve. Tablet was

placed on the sieve and the whole assembly was then placed on a shaker. The time at

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 115 


METHODOLOGY

which all the particles pass through the sieve was taken as a disintegration time of the

tablet. Six tablets were chosen randomly from the composite samples and the average

value was determined.

Figure-7: Modified Device Used to Determine Disintegration Time.

4.10.7 Wetting Time

The method was followed to measure tablet wetting time. A piece of tissue

paper (12 cm X 10.75 cm) folded twice was placed in a small Petri dish (ID = 65 cm)

containing 6 ml of Sorenson’s buffer (pH 6.8), A tablet was put on the paper, and the

time for the complete wetting was measured. Three trials for each batch were

performed and the standard deviation was also determined.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 116 


METHODOLOGY

Figure-8: In-vitro Wetting Property.

4.10.8 In-vitro Dispersion Time:

In-vitro dispersion time was measured by dropping a tablet in a glass cylinder

containing 6 ml of Sorenson’s buffer (pH 6.8). Three tablets from each formulation

were randomly selected and in-vitro dispersion time was performed.

Figure-9: In-vitro Disintegration Property.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 117 


METHODOLOGY

4.11. CONTENT UNIFORMITY:

Ten randomly selected tablets were weighed and powdered in a glass mortar

pestle. The weight equivalent to 10 mg Ketoprofen was weighed and dissolved in 5

ml of methanol in volumetric flask using magnetic stirrer, the volume was adjusted to

100 ml with Sorenson’s buffer (pH 6.8) and the solution was filtered. An aliquot of

1.0 ml of solution were diluted to 10 ml Sorenson’s buffer (pH 6.8) in separate

volumetric flask. The content in was determined spectrophotometrically at 260 nm.

The results were shown in Table 35.

4.12. IN-VITRO DISSOLUTION STUDIES:

In-vitro dissolution studies of formulation were carried out using USP paddle

method at 50 rpm in 900 ml of Sorenson’s buffer (pH 6.8) as dissolution media,

maintained at 37±0.50C. 5 ml of aliquot was withdrawn at the specified time intervals,

filtered through whatmann filter paper and analysed spectorphotometrically at 260

nm. An equal volume of fresh medium, which was prewarmed at same condition was

replaced into the dissolution media after each sampling to maintain the constant

volume throughout the test.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 118 


METHODOLOGY

4.13. STABILITY STUDIES: 105, 106.

Definition:

Stability is defined as “the capacity of the drug product to remain within

specifications established to ensure its identity, strength, quality and purity” (FDA

1987). In other words the stability of a drug is its ability to resist detoriation.

Need for stability studies: Objective and Purpose:

9 It is important that the point of view of the safety of patients, it is important

that the patient receive a uniform dose of a drug throughout the whole of shelf-

life.

9 Consideration must be taken to the relevant legal requirements concerned with

the identity, strength, purity, and quality of the drug.

9 Such a study is important to prevent economic repercussion of marketing an

unstable product.

9 Detoriation of drug may take several forms arising from changes in the

chemical, physical and microbiological properties .These changes may affect

therapeutic value of a dosage form or increases toxicity.

Prediction of shelf-life:

Shelf-life is a period during which a dosage form keeps it qualities. The prediction of

shelf-life is based on applying the Arrhenius equation, which gives the effect of

temperature on rate constant, K of a chemical reaction. The stability of any active

component in a dosage form can be evaluated by determining some properties of the

degradation (such as color disappearance, concentration etc.) Under accelerated

storage conditions as a function of time. If this function is lineared with any of

chemical kinetic orders, temperature dependency of the degradation can be obtained

with the help of Arrhenius equation:

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 119 


METHODOLOGY

Considering a preparation whose instability is measured in terms of color

disappearance when subjected to different elevated temperatures and a plot is drawn

with absorbance v/s time.

The reaction velocity constant K for the decomposition at each of the elevated

temperature can be calculated from the slope of line. The most satisfactory method for

expressing the influence of temperature on reaction velocity is the quantitative

relation proposed by Arrhenius :

K = A e ‫ ־‬E a / RT

Where :

K = Specific rate constant,

R= Gas constant(1.987cals/day/mol)

T= Absolute temperature,

Ea=Energy of activation.

The Arrhenius equation is then employed to determine the value for decomposition at

room temperature. This is obtained from linear plot of the logarithm of values against

reciprocal of absolute temperature, which is then extrapolated, to room temperature

(250 C). The value of K at 250 C may then substituted in the appropriate rate equation

and an estimate of the time during which the product will maintain the required quantity

or potency (shelf-life).

Most recently a guideline issued by the International Conference on Harmonization

(ICH, 1993) indicates that the purpose of stability testing is to provide evidence on how

the quality of a drug substance or the drug product varies with time under the influence of

vvariety of environmental factors, such as temperature, humidity and light, and enables

recommended storage conditions, retest periods, and shelf life to be established.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 120 


METHODOLOGY

The purpose of stability study is not only to characterize the degradation of a drug

product but also to establish an expiration-dating period or shelf life applicable to all

future batches of drug product.

Types of stability studies:

1. The results provide an estimate of the kinetic parameters for the rate of

reactions.

2. The results can be used to characterize the relationship between degradation

and storage condition.

3. The results supply critical information in the design and analysis of long-term

stability studies under ambient conditions at the planning stage.

Long–term studies, which include both pre-approval and post-approval stability

studies, are usually conducted under ambient condition.

A pre-approval stability study is also known as NDA stability study, the purpose

of it is to determine (estimate) a drug expiration dating applicable to all future

batches.

A post approval stability study is usually referred to as a marketing stability study;

the purpose of it is to make sure that the drug product currently on market can meet

the USP/NF specifications up to the end of expiration dating period.

Table-20: Storage cconditions according to ICH guidelines

Study Storage condition Minimum time

Temperature Relative humidity (%)

250C±20C 60% ±5% RH 12 Months


Long term

Intermediate 300± 20C 65%± 5% RH 6 Months

Accelerated 400± 20C 75%± 5% RH 3 Months

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 121 


METHODOLOGY

Note: The analyst can select any one of the three study conditions. Stability study

was carried out at 400C / 75% RH for the optimized formulations.

The procedure was divided into two parts,

Part I:

Achieving of 60% RH:

26.66 gm. of sodium hydroxide was weighed and dissolved in 100 ml

of distilled water to get 26.66% sodium hydroxide solution. The solution was placed

in the desiccator over which a wire mesh was placed, over which the dosage form was

placed and the desiccator was sealed. The desiccator was placed in the oven

maintained at 250C to create the Relative Humidity OF 60%.

Achieving of 75% RH:

Saturated solution of sodium chloride was prepared and placed in the

desiccators over which a wire mesh was placed, over which the dosage form was

placed and the desiccator was sealed. The desiccator was kept in oven maintained at

400C to create the relative humidity of 75%.

Part II

The sealed formulation were placed in amber colored bottles, tightly plugged with

cotton and capped. They were then stored at 250C /60% RH and 400C / 75% RH for

two months and evaluated for their physical appearance and drug content.

Results are shown in table 45 and 53.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 122 


RESULTS

CHAPTER-5.

RESULTS

5.1. Identification Tests:

Physical Appearance:

Physical appearance of drug was examined by various organoleptic properties.

Color : White or almost white;

Odor : Odorless;

Taste : Tasteless;

State : Fine to granular powder.

Melting Point:

Melting point of the Ketoprofen was determined by capillary fusion method;

one sided closed capillary filled with drug and put into the Melting Point Apparatus.

Temperature was noted at which solid drug changed into liquid. It was found to be

950C.

Table-21: Melting point of Ketoprofen.

OBSERVED VALUE
STANDARD
CAPILLARY FUSION DIFFERENTIAL
VALUE
METHOD SCANNING
CALORIMETRY

94-970 C 950 C 94.540 C

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 123 


RESULTS

Infrared Spectral Assignment:

The pellet of approximately 01 mm diameter of the drug was prepared

grinding 3-5 mg of sample with 100-150 mg of Potassium Bromide using hydrostatic

press. The sample pellet was mounted in IR compartment and scanned at wavelength

4000 cm-1 – 500 cm-1. On analysis of the IR spectra of the reference spectra (A) given

in Indian Pharmacopoeia (1996) and pure drug (B), no major differences were

observed in the characteristic absorption peak (1696, 1655) pattern. The results were

shown in Figure 10 (A) and 11 (B).

Figure-10: IR Spectra (A) Ketoprofen IP Spectra

1 1 2 .5

% T

105

9 7 .5

90

8 2 .5

75

6 7 .5

60

5 2 .5

45

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
b a c k g ro u n d 1 /c m

Figure-11: IR Spectra (B) Sample of Ketoprofen

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 124 


RESULTS

Differential Scanning Calorimetry (DSC):

DSC analysis was performed on 5 mg sample. Samples were heated in an open

aluminum pans at a rate of 100 per min–1 in a 30 to 3000C temperature range under a

nitrogen flow of 40 mL/min. It shows endothermic peak at about 94.120C Figure 12.

Figure-12: Differential Scanning Calorimetry of Ketoprofen

Ultraviolet Absorption Maxima:

Ultraviolet absorption in the rage 200 to 400 nm of a 5μg/ml solution in 5% (v/v)

methanolic Sorenson’s Buffer (pH 6.8) was measured. The absorption maxima (λmax)

of Ketoprofen (5 µg/ml) in this solution was found to be 260 nm which is concordant

with the Indian Pharmacopoeia (1996) shown in Figure 13.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 125 


RESULTS

1.4

1.2

1
A b so rb an ce
0.8

0.6

0.4

0.2

0
180 230 280 330 380
Wavelength (nm)

Figure-13: Scan Graph of Ketoprofen

5.2. PREFORMULATION STUDIES

5.2.1. Solubility

The solubility of Ketoprofen was determined in different solvent systems and buffers.

An excess quantity of the drug was mixed with 10 ml of each solvent in screw capped

glass tubes and shaken on constant water bath shaker for 24 hours at 25o. The

solutions were examined physically for the absence or presence of drug particles and

also by spectrophometrically for quantitative determination of drug in buffers. The

results were shown in Table 22.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 126 


RESULTS

Table-22: Solubility of Ketoprofen in Different Solvents

S.No. Solvents Solubility

1. Distilled water -

2. Sorenson's buffer pH 6.8 -

3. 0.1 HCl +

4. 0.1 NaOH +

5. Ethanol ++

6. Methanol ++
Practically insoluble (-) slightly soluble (+) soluble (++)

Table-23: Solubility of Ketoprofen in Various Buffer Solutions

S.No Buffers(pH) Solubility(mg/ml)

1. 6.8 (Sorenson's buffer) 0.014±0.002

2. 7.0 (Water) 0.016±0.003

3. 7.4 (Buffer) 0.016±0.002


Data are expressed as mean ± S.D. (n = 3)

5.2.2. Result of calibration curve data

The calibration curve of Ketoprofen was prepared in Sorenson’s buffer (pH

6.8). The plot of different concentrations of Ketoprofen versus absorbance was found

to be linear in the concentration range of 5-25 µg/ml at 260 nm. The absorbances at

different concentrations were shown in Table 24. The data of standard curve were

linearly regressed. The slope and correlation coefficient values were found to be

0.0198 and 0.9979 respectively. The intercept on Y-axis found to be 0.1361. The

calibration curve was shown in Figure 14.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 127 


RESULTS

Table-24: Calibration curve data of Ketoprofen

SR.NO. CONCENTRATION ABSORBANCE

1 0 0

2 5 0.211

3 10 0.422

4 15 0.633

5 20 0.837

6 25 1.013

Figure-14: Calibration curve of Ketoprofen

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 128 


RESULTS

5.2.3. Drug Polymer Interaction Studies:

100

% T

95

90

85

80

75

70
4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
c ro o s c a r 1 /c m

Figure-15: IR Spectra of Croscarmallose sodium

100

% T

90

80

70

60

50

40

30
4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
c ro o s c a r+ d ru g 1 /c m

Figure-16: IR Spectra of Mixture of Drug and Croscarmallose sodium

9 7 .5

% T

90

8 2 .5

75

6 7 .5

60

5 2 .5

45

3 7 .5

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
ss g 1 /c m

Figure-17: IR Spectra of Sodium Starch Glycolate

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 129 


RESULTS

9 7 .5

% T

90

8 2 .5

75

6 7 .5

60

5 2 .5

45

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
s s g + d ru g 1 /c m

Figure-18: IR Spectra of Mixture of Drug and Sodium Starch Glycolate

9 7 .5

% T

90

8 2 .5

75

6 7 .5

60

5 2 .5

45

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
coss car 1 /c m

Figure-19: IR Spectra of Crospovidone

100

% T

90

80

70

60

50

40

30

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
d 1/

Figure-20: IR Spectra of Mixture of Drug and Crospovidone

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 130 


RESULTS

Table-25: Interpretation of drug polymer interaction study.

Characteristics absorption
Code Functional group
peak cm-2
Ketoprofen OH str 3000.50
CH str 2900.01
CHdf 1290.23
C=O str 1650
Ar-H 710
Crospovidone OH str 3200.3
CH str 2980.01
CHdf 1240.23
C=O str 1680.01
Crospovidone+Ketoprofen CH str 3000.01
CHdf 1280.23
C=O str 1700
Ar-H 710
Sodium starch glycolate OH str 3580.01
CH str 2980.01
CHdf 1320.23
C=O str 1600
Sodium starch glycolate+ CH str 3000.01
Ketoprofen CHdf 1280.23
C=O str 1700
Ar-H 710
Crosscarmalose sodium O-H str 3564.85
C-H str 2888.47
C=O str 1616.29
C-O str 997.71
Crosscarmalose sodium + CH str 3000.01
Ketoprofen CHdf 1280.23
C=O str 1700
Ar-H 710

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 131 


RESULTS

5.3. EVALUATION OF SOLID DISPERSION;

Prepared polymer drug conjugates were evaluated by

1) Determination of solubility of solid dispersion

2) Estimation of drug content

3) In- vitro dissolution studies

4) Dissolution efficiency of solid dispersion

5) FT-IR

6) X-ray diffraction (XRD)

7) Differential scanning calorimeter (DSC)

8) Scanning electron microscopy (SEM)

1. DETERMINATION OF SOLUBILITY OF SOLID DISPERSION:

Table-26: Solubility data of Ketoprofen, physical mixture and solid dispersion in


Sorenson’s buffer pH 6.8 at 25oC and 370C.

FORMULATION
CODE Ketoprofen solubility Ketoprofen solubility
0
(mg/ml) at 25 C (mg/ml) at 370 C
Pure Drug 0.014333 ± 0.003086 0.018417 ± 0.001809
KP1 0.28825 ± 0.002883 0.316167 ± 0.00527
KP2 0.383333 ± 0.002765 0.438917 ± 0.001665
KP3 0.46675 ± 0.003 0.564917 ± 0.002082
KPG1 0.2275 ± 0.002537 0.251833 ± 0.001507
KPG2 0.331 ± 0.002634 0.388333 ± 0.001127
KPG3 0.444917 ± 0.004856 0.501083 ± 0.001127
KPVP1 0.533083 ± 0.002126 0.603833 ± 0.001258
KPVP2 0.6775 ± 0.002537 0.751 ± 0.001146
KPVP3 0.803833 ± 0.003263 0.894417 ± 0.000878
KPEG1 0.481333 ± 0.00366 0.575333 ± 0.001127
KPEG2 0.620167 ± 0.006385 0.715417 ± 0.008098
KPEG3 0.71125 ± 0.005074 0.80475 ± 0.00522
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 132 


RESULTS

2. ESTIMATION OF DRUG CONTENT:

Table-27: Percent Drug Content of Ketoprofen-PVP K-30 & Ketoprofen-PEG-


6000 Physical Mixtures and Solid Dispersions

SR.NO. Formulation Number %Drug Content

1 KP1 99.08333±0.242813
2 KP2 98.66667±0.438986
3 KP3 98.25±0.450694
4 KPG1 99.025±0.15
5 KPG2 98.10833±0.448144
6 KPG3 98.08333±0.401819
7 KPVP1 99.36667±0.500208
8 KPVP2 98.61667±0.58648
9 KPVP3 98.51667±0.57027
10 KPEG1 98.91667±0.496446
11 KPEG2 97.94167±0.312583
12 KPEG3 98.08333±1.127035

Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 133 


RESULTS

3) IN-VITRO DISSOLUTION STUDIES:

Table-28: Dissolution Release Profile of Ketoprofen from Physical Mixture.

Time Cumulative Mean Percent Drug Release ± Standard Deviation


(min) Pure KP1 KP2 KP3 KPG1 KPG2 KPG3
drug
0 0 0 0 0 0 0 0
5 1.89 5.265 10.2 14.1075 4.38 8.8725 12.03
±0.2025 ±0.2475 ±0.225375 ±0.2475 ±0.213849 ±0.202916 ±0.213849
10 3.7071 12.73335 19.52633 26.19818 12.27487 17.46986 25.08587
±0.259056 ±0.225649 ±0.214098 ±0.214124 ±0.236576 ±0.225225 ±0.236576
15 6.028717 20.18749 26.68052 34.46977 20.531 29.07676 33.82123
±0.236852 ±0.225898 ±0.225862 ±0.237562 ±0.24834 ±0.225475 ±0.236839
30 13.92541 34.4374 42.73013 50.86552 37.80379 43.16403 50.43376
±0.237114 ±0.226148 ±0.226112 ±0.248275 ±0.237114 ±0.248564 ±0.225763
45 24.35837 44.66062 53.42753 62.36694 49.23825 54.54443 61.43971
±0.237376 ±0.237364 ±0.226358 ±0.214898 ±0.214885 ±0.237339 ±0.21486
60 34.2854 52.37515 61.87928 70.82108 54.94037 62.32241 67.83783
±0.226673 ±0.248788 ±0.226609 ±0.237626 ±0.215123 ±0.237601 ±0.20375
Data are expressed as mean ± S.D. (n = 3)

Table-29: Dissolution release profile of Ketoprofen from solid dispersion

Time Cumulative Mean Percent Drug Release ± Standard Deviation


(min) KPVP1 KPVP2 KPVP3 KPEG1 KPEG2 KPEG3
0 0 0 0 0 0 0
5 7.29 23.2725 36.465 9.93 16.59 25.02
±0.225 ±0.202916 ±0.213849 ±0.236339 ±0.236339 ±0.225
10 18.6156 38.98836 57.04802 23.62853 30.94093 45.3203
±0.22525 ±0.214074 ±0.248077 ±0.248103 ±0.236602 ±0.22525
15 28.70128 54.93165 74.11386 34.62728 43.74029 60.09313
±0.203415 ±0.203379 ±0.236852 ±0.236877 ±0.214373 ±0.203415
30 49.23813 75.73761 90.71859 54.96821 66.06383 78.79731
±0.248564 ±0.172529 ±0.248615 ±0.248641 ±0.214611 ±0.248564
45 61.69028 85.75162 99.81168 68.0492 75.34713 88.3347
±0.214848 ±0.130698 ±0.172872 ±0.237415 ±0.226 ±0.248839
60 70.81119 93.16666 - 74.41716 82.78815 94.59759
±0.226238 ±0.226 ±0.226338 ±0.22625 ±0.249115
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 134 


RESULTS

Figure-21: Cumulative Percent Release of Ketoprofen from Physical Mixtures of


Ketoprofen-PVP K-30 and Ketoprofen-PEG-6000 Systems

Figure-22: Cumulative Percent Release of Ketoprofen from Solid Dispersions of


Ketoprofen-PVP K-30 and Ketoprofen-PEG-6000 Systems

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 135 


RESULTS

4. DISSOLUTION EFFICIENCY:

Table-30: Dissolution Efficiency of Ketoprofen-PVP K30 &/ Ketoprofen-PEG


6000 Physical Mixtures and Solid Dispersions.

FORMULATION Dissolution Efficiency (%)


DE 15 DE 30
Pure drug 2.92 6.46
KP1 9.58 18.13
KP2 13.33 23.96
KP3 18.75 30.42
KPG1 8.33 18.95
KPG2 12.91 24.58
KPG3 17.92 29.17
KPVP1 12.91 25.83
KPVP2 30 47.92
KPVP3 44.16 63.33
KPEG1 17.50 31.67
KPEG2 23.33 39.17
KPEG3 34.17 52.08

5. FT-IR STUDY:

97.5

%T

90

82.5

75

67.5

60

52.5

45

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
PVP K30 1/cm

Figure-23: FT-IR Spectra of PVP K 30.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 136 


RESULTS

105
%T
97.5

90

82.5

75

67.5

60

52.5

45
4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
PVP K30 +KETOPROFEN 1/cm

Figure-24: FT-IR Spectra of Ketoprofen and PVP K 30.

100
%T
90

80

70

60

50

40

30

20

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
PEG-6000 1/cm

Figure-25: FT-IR Spectra of PEG-6000.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 137 


RESULTS

100
%T
90

80

70

60

50

40

30

20

10

4000 3500 3000 2500 2000 1750 1500 1250 1000 750 500
PEG-6000+KETOPROFEN 1/cm

Figure-26: FT-IR Spectra of Ketoprofen and PEG-6000.

Table-31: Interpretation of drug polymer interaction study

Code Functional group Charecteristics absorption


peak cm-2
PEG 6000 OH str 3200.50
CH str 2985.01
CHdf 1105.23
PEG 6000+Ketoprofen CH str 2800.01
CHdf 1110.23
C=O str 1700
Ar-H 890
PVP K30 OH str 3550.12
CH str 2980.01
C-N 800.00
C=N str 1650.00
PVP K30+ Ketoprofen CH str 3000.01
CHdf 1280.23
C=O str 1700
Ar-H 710

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 138 


RESULTS

5. DIFFERENTIAL SCANNING CALORIMETRY ANALYSIS:

Differential Scanning Calorimetry analysis was performed on 5 mg samples.


Samples were heated in an open aluminum pans at a rate of 100 per min–1 in a 30 to
300°C temperature range under a nitrogen flow of 40 mL/min.

Figure-27: DSC of PEG-6000

Figure-28: DSC of PVP K-30

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 139 


RESULTS

Figure-29: DSC of KPVP-3

Figure-30: DSC of KPEG-3

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 140 


RESULTS

6. X- Ray Diffraction (XRD)

XRD patterns were recorded using Philips PW 1729 X-ray generator. Powder
X-ray diffraction patterns were traced for Ketoprofen, various carriers and solid
dispersions.

Figure-31: X- Ray Diffraction Ketoprofen

Figure-32: X- Ray Diffraction PVP K-30

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 141 


RESULTS

Figure-33: X- Ray Diffraction PEG-6000

Figure-34: X- Ray Diffraction KPVP-3

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 142 


RESULTS

Figure-35: X- Ray Diffraction KPEG-3

7. SCANNING ELECTRON MICROSCOPY:

The external morphology of solid dispersions was analyzed by Scanning


Electron Microscope (SEM). The samples were examined under a scanning electron
microscope JSM 6100 JEOL JAPAN.

Figure-36: Scanning Electron Photomicrograph of Ketoprofen

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 143 


RESULTS

Figure-37: Scanning Electron Photomicrograph of PVP K-30

Figure-38: Scanning Electron Photomicrograph of Solid Dispersion (KPVP-3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 144 


RESULTS

5.4. Characterization of Blends for Fast Dissolving Tablets:

Table-32: Characterization of Blends.

Formulation Bulk Tapped Hausners Compressibilit Angle


Density density ratio y Index repose
F1 0.681092 0.783293 1.150054 13.04752 23.99959
±0.001094 ±0.001873 ±0.000933 ±0.070577 ±0.520574
F2 0.591251 0.673251 1.13869 12.17977 23.14928
±0.001069 ±0.001386 ±0.000285 ±0.022013 ±0.50656
F3 0.61501 0.704891 1.146146 12.75107 23.2435
±0.002005 ±0.002071 ±0.000411 ±0.031322 ±0.485106
F4 0.670249 0.75873 1.132016 11.66196 23.82844
±0.002695 ±0.002303 ±0.001117 ±0.087151 ±0.837309
F5 0.598567 0.680278 1.13651 12.01126 22.40116
±0.001802 ±0.002444 ±0.001053 ±0.081509 ±0.719261
F6 0.677508 0.755288 1.114806 10.29793 23.7341
±0.000918 ±0.001141 ±0.002766 ±0.222876 ±0.591321
F7 0.567537 0.641849 1.130937 11.57776 23.70906
±0.000644 ±0.000824 ±0.000168 ±0.013142 ±0.491507
F8 0.61535 0.697039 1.132748 11.71899 23.05747
±0.00239 ±0.003689 ±0.001647 ±0.128243 ±0.747942
F9 0.553101 0.630258 1.139499 12.24204 24.26282
±0.001621 ±0.00243 ±0.001186 ±0.091409 ±0.744986
F10 0.608027 0.68151 1.120855 10.78234 23.69731
±0.001129 ±0.00142 ±0.000871 ±0.069326 ±0.999613
F11 0.612746 0.700609 1.143392 12.54091 20.72383
±0.000751 ±0.001501 ±0.001105 ±0.084452 ±0.51672
F12 0.560748 0.656169 1.170168 14.5419 23.11178
±0.000363 ±0.000861 ±0.002224 ±0.16249 ±0.676691
F13 0.608027 0.699304 1.150119 13.05239 25.66395
±0.001129 ±0.001956 ±0.001191 ±0.089995 ±0.45653
F14 0.594061 0.691291 1.163659 14.0602 25.01529
±0.001079 ±0.006953 ±0.009671 2±0.71082 ±0.541887
F15 0.666076 0.75567 1.134509 11.85616 23.78599
±0.001356 ±0.001746 ±0.000311 ±0.024141 ±0.471261
F16 0.585941 0.66756 1.139296 12.22651 23.81718
±0.001727 ±0.001783 ±0.000485 ±0.037362 ±0.730814
F17 0.609016 0.694448 1.140278 12.30212 23.70906
±0.001484 ±0.001929 ±0.00039 ±0.029969 ±0.491507
F18 0.624222 0.722552 1.157521 13.60825 24.3578
±0.001559 ±0.003132 ±0.002128 ±0.158823 ±0.714602
F19 0.657607 0.751127 1.142213 12.45062 23.79845
±0.001 ±0.000652 ±0.000744 ±0.057075 ±0.661456
F20 0.634788 0.741109 1.167491 14.34621 26.10666
±0.001232 ±0.001679 ±0.000379 ±0.027839 ±0.462065
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 145 


RESULTS

5.5. CHARACTERIZATION OF FAST DISSOLVING TABLETS

Table-33: Characterization of Fast Dissolving Tablets.

Formulation Thickness (mm) Weight (mg) Friability (%) Hardness


(Kg/cm2)
F1 6.325±0.014 500.6667 0.478151 3.166667
±1.527525 ±0.000291 ±0.057735
F2 6.342±0.026 496.6667 0.440411 3.133333
±3.785939 ±0.000269 ±0.057735
F3 6.343±0.034 497.3333 0.639744 2.8
±0.57735 ±0.000256 ±0.1
F4 6.325±0.004 501.3333 0.781955 2.8
±2.081666 ±0.045364 ±0.1
F5 6.349±0.037 496.6667 0.876262 2.733333
±1.527525 ±0.000533 ±0.152753
F6 6.342±0.029 502.6667 0.719904 3
±1.527525 ±0.0006 ±0.173205
F7 6.348±0.043 500.3333 0.519308 3.6
±1.527525 ±0.000432 ±0.1
F8 6.349±0.021 500.3333 0.800108 3
±2.309401 ±0.00103 ±0.173205
F9 6.334±0.034 499.3333 0.638808 3.266667
±1.527525 ±0.00039 ±0.057735
F10 6.325±0.008 498.6667 0.519654 3.533333
±2.081666 ±0.000523 ±0.11547
F11 6.345±0.016 499.3333 0.519792 3.433333
±3.21455 ±0.000208 ±0.11547
F12 6.372±0.031 498.6667 0.719138 3.133333
±0.57735 ±0.000994 ±0.152753
F13 6.346±0.034 501.6667 0.76132 3.166667
±2.081666 ±0.000466 ±0.152753
F14 6.335±0.031 499.3333 0.519662 3.066667
±1.527525 ±0.002573 ±0.208167
F15 6.348±0.031 501.3333 0.478151 3.3
±1.527525 ±0.000396 ±0.2
F16 6.344±0.034 498.6667 0.760102 2.666667
±0.57735 ±0.000978 ±0.152753
F17 6.363±0.035 498.6667 0.67991 2.8
±1.527525 ±0.000415 ±0.173205
F18 6.343±0.016 499.3333 0.840785 2.566667
±2.081666 ±0.000514 ±0.208167
F19 6.366±0.041 498.6667 0.919755 2.5
±3.05505 ±0.000212 ±0.173205
F20 6.321±0.339 500.6667 0.958467 2.7
±1.527525 ±0.000383 ±0.264575
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 146 


RESULTS

Table-34: Characterization of Fast Dissolving Tablets.

Formulation Disintegration time Wetting time Dispersion time


(Seconds) (Seconds) (Seconds)
F1 95.53333 87.37 114.3467
±1.507459 ±1.770339 ±3.381262
F2 86.50333 80.13667 101.4433
±2.360452 ±3.928897 ±2.408513
F3 70.25667 65.34333 84.76
±3.769726 ±3.674647 ±4.26522
F4 60.65 55.04 71.81
±1.85696 ±3.125972 ±3.404453
F5 51.94667 49.22333 64.70667
±3.34403 ±3.511885 ±4.265024
F6 134.2233 125.6633 142.7133
±5.162183 ±5.760046 ±4.83864
F7 119.93 110.2733 124.9033
±4.994207 ±3.544014 ±4.639874
F8 59.48 80.09333 86.23333
±1.509073 ±4.400231 ±2.717174
F9 45.11333 62.69 69.72333
±2.155327 ±2.507349 ±1.804476
F10 35.60667 48.98333 55.07333
±1.471915 ±3.493799 ±3.127944
F11 47.48333 60.04333 67.46667
±1.878546 ±3.823354 ±2.541679
F12 36.01333 42.94 54.35
±1.651676 ±3.040049 ±2.626385
F13 29.02333 31.83667 42.83667
±1.708489 ±2.848233 ±1.180777
F14 27.71 29.67 33.69667
±1.141753 ±1.477329 ±2.080993
F15 25.68 27.44667 30.91
±1.411063 ±1.404754 ±1.681547
F16 68.31333 59.38333 69.56
±2.26809 ±1.8037 ±1.915385
F17 51.65667 49.47333 56.48
±2.110126 ±2.941451 ±3.137276
F18 39.41 43.66333 49.44667
±2.201704 ±2.033749 ±3.786322
F19 31.52667 36.40667 42.74333
±3.023183 ±1.420047 ±2.555902
F20 29.16 31.11667 34.19
±1.746396 ±2.146214 ±1.93
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 147 


RESULTS

Figure-39: Effect of Concentration of Superdisintegrant and Effervescent Agent


on Disintegration Time.

Figure-40: Effect of Concentration of Superdisintegrant and Effervescent Agent


on Dispersion Time

Figure-41: Effect of Concentration of Superdisintegrant and Effervescent Agent


on Wetting Time

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 148 


RESULTS

Table-35: Drug content in the Fast Dissolving Tablets of Ketoprofen

Formulation Drug content Drug content


(mg per tablets) %
F1 49.34167 98.68333
±0.339424 ±0.678847
F2 48.88333 97.76667
±0.440407 ±0.880814
F3 49.46667 98.93333
±0.398696 ±0.797392
F4 48.65833 97.31667
±0.146487 ±0.292973
F5 49.59167 99.18333
±0.177365 ±0.35473
F6 49.06667 98.13333
±0.330088 ±0.660177
F7 49.65 99.3
±0.139194 ±0.278388
F8 49.10833 98.21667
±0.473242 ±0.946485
F9 49.225 98.45
±0.229129 ±0.458258
F10 50.54167 101.0833
±0.052042 ±0.104083
F11 49.24167 98.48333
±0.376109 ±0.752219
F12 49.425 98.85
±0.388104 ±0.776209
F13 49.19167 98.38333
±0.357363 ±0.714726
F14 49.71667 99.43333
±0.177365 ±0.35473
F15 50.36667 100.7333
±0.028868 ±0.057735
F16 48.99167 97.98333
±0.189297 ±0.378594
F17 49.15 98.3
±0.417582 ±0.835165
F18 49.90833 99.81667
±0.23094 ±0.23094
F19 49.025 98.05
±0.238485 ±0.47697
F20 49.06667 98.13333
±0.300347 ±0.600694
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 149 


RESULTS

5.6. DETERMINATION OF IN-VITRO DRUG RELEASE:

Table-36: Zero Order Dissolution Release Profile of Ketoprofen from F1-F5.

Time Cumulative Mean Percent Drug Release ± Standard Deviation


(min) F1 F2 F3 F4 F5
0 0 0 0 0 0
4 38.655 44.565 50.34 54.78 59.655
±0.311769 ±0.340735 ±0.432406 ±0.578619 ±0.343693
8 48.08795 52.32452 60.61093 68.92587 73.49128
±0.289652 ±0.289688 ±0.332174 ±0.579262 ±0.37508
12 55.44633 59.6726 65.38822 73.14238 81.16287
±0.237475 ±0.290009 ±0.350381 ±0.579905 ±0.426119
16 61.08783 68.64878 78.09073 82.0885 86.86288
±0.23774 ±0.290331 ±0.325734 ±0.580548 ±0.358443
20 64.35053 73.31487 83.38228 86.30447 90.55913
±0.238004 ±0.290652 ±0.376293 ±0.327051 ±0.477898
Data are expressed as mean ± S.D. (n = 3)

Table-37: Zero Order Dissolution Release Profile of Ketoprofen from F6-F10.

Time Cumulative Mean Percent Drug Release ± Standard Deviation


(min) F6 F7 F8 F9 F10

0 0 0 0 0 0
4 27.855 32.085 41.805 54.195 61.035
±0.3245 ±0.38448 ±0.45 ±0.687386 ±0.542494
8 35.29595 41.72065 53.62645 70.93522 77.27282
±0.595313 ±0.306727 ±0.382633 ±0.663655 ±0.496284
12 46.58513 51.66697 67.11098 81.04897 86.1486
±0.375121 ±0.350297 ±0.450425 ±0.451475 ±1.039054
16 52.75682 56.43428 76.66543 84.99387 89.43915
±0.344469 ±0.325651 ±0.687676 ±0.342792 ±0.45225
20 56.4003 61.01183 82.94542 88.26805 91.29325
±0.307538 ±0.474162 ±0.676062 ±0.483679 ±0.45275
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 150 


RESULTS

Table-38: Zero Order Dissolution Release Profile of Ketoprofen from F11-F15.

Time Cumulative Mean Percent Drug Release ± Standard Deviation


(min) F11 F12 F13 F14 F15

0 0 0 0 0 0
4 45.9 50.04 54.345 59.91 63.54
±0.518965 ±0.45 ±0.93675 ±0.687386 ±0.54
8 55.026 59.5456 67.42538 72.12657 82.3456
±0.496258 ±0.474603 ±0.708264 ±0.68815 ±0.473278
12 67.92708 72.3467 76.54523 81.10163 91.69202
±0.680594 ±0.451025 ±0.901775 ±0.4515 ±0.708338
16 73.25243 78.24695 84.32513 87.10158 94.79372
±0.497553 ±0.527136 ±0.452775 ±0.689405 ±0.768666
20 77.60862 85.81867 91.13858 93.8581 99.54875
±0.430121 ±0.938291 ±0.633803 ±0.47544 ±0.45275
Data are expressed as mean ± S.D. (n = 3)

Table-39: Zero Order Dissolution Release Profile of Ketoprofen from F15-F20.

Time Cumulative Mean Percent Drug Release ± Standard Deviation


(min) F16 F17 F18 F19 F20

0 0 0 0 0 0
4 24.66 45.24 49.785 57.465 63.54
±0.45 ±0.800359 ±0.610962 ±0.518965 ±0.495
8 40.4374 55.28027 63.11532 72.37885 81.1606
±0.4505 ±0.344582 ±0.203281 ±0.590734 ±0.519514
12 57.5973 64.08663 73.71538 77.7692 85.7207
±0.473678 ±0.496918 ±0.473446 ±0.541225 ±0.688499
16 62.56622 68.20772 77.08215 85.28045 90.67577
±0.474203 ±0.497468 ±0.923888 ±0.197644 ±0.956807
20 67.13558 72.6933 81.96757 90.35495 94.75123
±0.519713 ±0.857738 ±0.665137 ±0.78961 ±0.948269
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 151 


RESULTS

Figure-42: Zero Order Dissolution Release Profile of Ketoprofen from F1-F5

Figure-43: Zero Order Dissolution Release Profile of Ketoprofen from F6-F10

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 152 


RESULTS

Figure-44: Zero Order Dissolution Release Profile of Ketoprofen from F11-F15

Figure-45: Zero Order Dissolution Release Profile of Ketoprofen from F15-F20.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 153 


RESULTS

Table-40: First Order Release Profile of Ketoprofen from F1-F5.

Time Cumulative Mean Percent Drug Retain ± Standard Deviation


(min) F1 F2 F3 F4 F5
0 2 2 2 2 2
4 1.787137 1.743189 1.695663 1.654145 1.605371
±0.001914 ±0.00213 ±0.002499 ±0.00437 ±0.002529
8 1.714759 1.67774 1.594699 1.490651 1.422364
±0.001901 ±0.00207 ±0.002774 ±0.00638 ±0.004533
12 1.649755 1.604942 1.538543 1.427029 1.273178
±0.0022 ±0.002451 ±0.003193 ±0.007398 ±0.007435
16 1.591083 1.495403 1.339686 1.250001 1.116111
±0.002522 ±0.003159 ±0.00462 ±0.011149 ±0.00914
20 1.553143 1.425265 1.218891 1.135015 0.970332
±0.002756 ±0.003718 ±0.007276 ±0.007452 ±0.017108
Data are expressed as mean ± S.D. (n = 3)

Table-41: First Order Release Profile of Ketoprofen from F6-F10.

Time Cumulative Mean Percent Drug Retain ± Standard Deviation


(min) F6 F7 F8 F9 F10
0 2 2 2 2 2
4 1.857931 1.831815 1.764873 1.660154 1.590298
±0.001392 ±0.001555 ±0.001943 ±0.004465 ±0.003815
8 1.804592 1.765118 1.664228 1.462246 1.356155
±0.00375 ±0.001707 ±0.003353 ±0.00674 ±0.005771
12 1.727155 1.683761 1.517007 1.277506 1.134667
±0.002247 ±0.002283 ±0.003449 ±0.006014 ±0.025065
16 1.673972 1.638683 1.373464 1.181646 1.02329
±0.002167 ±0.002316 ±0.008384 ±0.010476 ±0.01087
20 1.638938 1.590815 1.22344 1.07293 0.939256
±0.002293 ±0.003136 ±0.012335 ±0.014005 ±0.013235

Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 154 


RESULTS

Table-42: First Order Release Profile of Ketoprofen from F11-F15.

Time Cumulative Mean Percent Drug Retain ± Standard Deviation


(min) F11 F12 F13 F14 F15
0 2 2 2 2 2
4 1.732997 1.698605 1.657974 1.602161 1.561769
±0.002577 ±0.002264 ±0.006589 ±0.005106 ±0.003728
8 1.65279 1.606694 1.511876 1.443891 1.246512
±0.002906 ±0.003174 ±0.006355 ±0.007373 ±0.006938
12 1.506651 1.441689 1.369905 1.276296 0.921493
±0.004857 ±0.004107 ±0.009743 ±0.006031 ±0.018891
16 1.426966 1.331485 1.195016 1.107392 0.71432
±0.00491 ±0.009831 ±0.007304 ±0.016137 ±0.036197
20 1.350146 1.14619 0.941629 0.786447 #NUM!
±0.004714 ±0.021633 ±0.023239 ±0.020371 ±#NUM!
Data are expressed as mean ± S.D. (n = 3)

Table-43: First Order Release Profile of Ketoprofen from F16-F20.

Time Cumulative Mean Percent Drug Retain ± Standard Deviation


(min) F16 F17 F18 F19 F20
0 2 2 2 2 2
4 1.877018 1.739072 1.701126 1.628484 1.561777
±0.0015 ±0.003194 ±0.002799 ±0.00328 ±0.003416
8 1.774961 1.650852 1.56808 1.441851 1.274383
±0.0019 ±0.001673 ±0.001913 ±0.00483 ±0.007451
12 1.62729 1.555035 1.41951 1.346827 1.151927
±0.002878 ±0.003646 ±0.004647 ±0.006143 ±0.014526
16 1.573142 1.502065 1.359681 1.169857 0.960707
±0.003265 ±0.004125 ±0.010346 ±0.00314 ±0.033532
20 1.516572 1.435818 1.25414 0.983189 0.711549
±0.004071 ±0.00816 ±0.010936 ±0.02068 ±0.049413
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 155 


RESULTS

Figure-46: First Order Release Profile of Ketoprofen from F1-F5

Figure-47: First Order Release Profile of Ketoprofen from F6-F10

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 156 


RESULTS

Figure-48: First Order Release Profile of Ketoprofen from F11-F15

Figure-49: First Order Release Profile of Ketoprofen from F15-F20

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 157 


RESULTS

Table-44: Fit of various kinetics Models for fast dissolving Tablets of Ketoprofen

Formulatio Zero order First order


Code Intercept R2 Slope K Intercept R2 Slope K t1/2
-1
(mg/min) (min )
F1
16.29 0.797 2.831 6.519793 1.922 0.9 0.02 0.04606 15.04559
F2
17.88 0.808 3.187 7.339661 1.921 0.935 0.026 0.059878 11.57353
F3
20.23 0.806 3.606 8.304618 1.923 0.955 0.035 0.080605 8.597482
F4
23.89 0.757 3.697 8.514191 1.892 0.944 0.04 0.09212 7.522796
F5
26.56 0.734 3.872 8.917216 1.881 0.956 0.048 0.110544 6.268997
F6
10.19 0.891 2.628 6.052284 1.957 0.952 0.017 0.039151 17.7007
F7
12.76 0.856 2.771 6.381613 1.945 0.938 0.019 0.043757 15.83747
F8
15.63 0.883 3.805 8.762915 1.962 0.99 0.037 0.085211 8.132753
F9
24.39 0.76 3.884 8.944852 1.889 0.946 0.044 0.101332 6.838906
F10
28.2 0.711 3.932 9.055396 1.856 0.93 0.051 0.117453 5.900232
F11
18.78 0.812 3.45 7.94535 1.919 0.946 0.03 0.06909 10.0304
F12
20.05 0.821 3.76 8.65928 1.932 0.974 0.039 0.089817 7.715689
F13
22.67 0.8 3.962 9.124486 1.933 0.978 0.048 0.110544 6.268997
F14
25.69 0.761 3.998 9.207394 1.92 0.973 0.055 0.126665 5.471125
F15
29.06 0.761 4.291 9.882173 1.993 0.972 0.091 0.209573 3.306724
F16
8.74 0.924 3.332 7.673596 1.976 0.974 0.024 0.055272 12.53799
F17
19.4 0.774 3.151 7.256753 1.906 0.907 0.025 0.057575 12.03647
F18
21.73 0.776 3.588 8.263164 1.9 0.936 0.035 0.080605 8.597482
F19
25.26 0.752 3.861 8.891883 1.896 0.961 0.046 0.105938 6.541562
F20
29.32 0.704 3.998 9.207394 1.874 0.962 0.059 0.135877 5.100201

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 158 


RESULTS

5.7. STABILITY STUDY:


Table-45: Effect of Storage Condition (40°C/75%RH) on Weight of the

Ketoprofen Tablets.

FORMULATION PERIOD Weight (mg/tablet)


Initial 501.3333±2.081666
st
F4 1 month 501±1
2nd month 500.6667±1.154701
3rd month 499.6667±0.57735
Initial 498.6667±2.081666
F10 1st month 497.6667±0.57735
2nd month 497±1
rd
3 month 496.6667±0.57735
Initial 501.3333±1.527525
F15 1st month 500.6667±0.57735
nd
2 month 500.3333±0.57735
3rd month 499.3333±1.154701
Initial 499.3333±2.081666
F18 1st month 499.3333±0.57735
nd
2 month 499±1
rd
3 month 498.3333±1.527525
Data are expressed as mean ± S.D. (n = 3)

Table-46: Effect of Storage Condition (40°C/75%RH) on Hardness of the

Ketoprofen Tablets

FORMULATION PERIOD HARDNESS (Kg/cm2)


Initial 2.8±0.1
st
F4 1 month 2.766667±0.057735
2nd month 2.733333±0.057735
rd
3 month 2.633333±0.057735
Initial 3.533333±0.11547
st
F10 1 month 3.5±0.1
nd
2 month 3.466667±0.057735
3rd month 3.4±0.1
Initial 3.3±0.2
F15 1st month 3.233333±0.057735
2nd month 3.133333±0.11547
rd
3 month 3.1±0.1
Initial 2.566667±0.208167
F18 1st month 2.533333±0.057735
2nd month 2.5±0.1
rd
3 month 2.466667±0.057735
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 159 


RESULTS

Table-47: Effect of Storage Condition (40°C/75%RH) on Friability of the

Ketoprofen Tablets

FORMULATION PERIOD FRIABILITY (%)


Initial 0.781955±0.045364
F4 1st month 0.770353±0.022919
nd
2 month 0.770454±0.02283
3rd month 0.770353±0.022919
Initial 0.519654±0.000523
st
F10 1 month 0.505719±0.022938
2nd month 0.492471±0.022763
rd
3 month 0.479425±0.000383
Initial 0.478151±0.000396
F15 1st month 0.478723±0.00022
2nd month 0.465539±0.022727
3rd month 0.452306±0.02299
Initial 0.840785±0.000514
F18 1st month 0.840448±0.000194
nd
2 month 0.827441±0.02321
3rd month 0.814198±0.046149
Data are expressed as mean ± S.D. (n = 3)

Table-48: Effect of Storage Condition (40°C/75%RH) on Disintegration Time of

the Ketoprofen Tablets

FORMULATION PERIOD Disintegration time (Sec.)


Initial 60.65±1.85696
st
F4 1 month 59.96667±1.028462
2nd month 57.41333±0.276827
rd
3 month 56.25±1.166576
Initial 35.60667±4.400231
F10 1st month 35.07±0.343948
2nd month 33.91±0.765441
rd
3 month 31.84±1.557337
Initial 25.68±1.404754
F15 1st month 24.96667±0.341516
2nd month 23.19±0.629603
3rd month 21.91667±0.658584
Initial 39.41±2.033749
F18 1st month 38.52±0.52915
nd
2 month 36.51667±0.835244
rd
3 month 34.16667±0.501631
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 160 


RESULTS

Table-49: Effect of Storage Condition (40°C/75%RH) on Drug Content of the

Ketoprofen Tablets

FORMULATION PERIOD DRUG CONTENT (%)


Initial 97.31667±0.146487
F4 1st month 96.8±0.163936
nd
2 month 96.35±0.222205
3rd month 95.68333±0.076376
Initial 101.0833±0.052042
st
F10 1 month 100.2333±0.112731
2nd month 99.6±0.163936
rd
3 month 99.06667±0.052042
Initial 100.7333±0.028868
F15 1st month 100.3167±0.152753
2nd month 99.86667±0.062915
rd
3 month 99.08333±0.112731
Initial 99.81667±0.11547
F18 1st month 99.43333±0.146487
2nd month 98.71667±0.101036
3rd month 97.81667±0.062915
Data are expressed as mean ± S.D. (n = 3)

Table-50: In-vitro release profile of F4 during Stability studies at (40°C/75%RH)

Time Cumulative % drug release (X± S.D)*


(min) Initial 1month 2 month 3 month
0 0 0 0 0
4 54.78 54.225 53.28 48.57
±0.578619 ±0.16225 ±0.862076 ±0.706275
8 68.92587 68.41525 67.1842 60.98397
±0.579262 ±0.09005 ±1.024416 ±0.40575
12 73.14238 72.7512 72.1487 68.23667
±0.579905 ±0.315977 8±0.31787 ±0.563844
16 82.0885 81.45688 80.7338 76.99735
±0.580548 ±0.203206 ±0.348173 ±0.434243
20 86.30447 85.79215 84.60327 80.66768
±0.327051 ±0.248372 ±0.213942 ±0.40811
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 161 


RESULTS

Table-51: In-vitro release profile of F10 during Stability studies at


(40°C/75%RH)

Time Cumulative % drug release (X± S.D)*


(min) Initial 1month 2 month 3 month
0 0 0 0 0
4 61.035 60.285 56.715 53.595
±0.542494 ±0.226495 ±0.274955 ±0.18
8 77.27282 76.43198 74.73302 72.17955
±0.496284 ±0.324749 ±0.248143 ±0.34389
12 86.1486 85.08183 83.90598 80.47968
±1.039054 ±0.231533 ±0.338327 ±0.300171
16 89.43915 88.7912 87.92905 84.57395
±0.45225 ±0.182528 ±0.33263 ±0.338649
20 91.29325 90.52458 89.42148 87.38267
±0.45275 ±0.274748 ±0.22781 ±0.473953
Data are expressed as mean ± S.D. (n = 3)

Table-52: In-vitro release profile of F15 during Stability studies at


(40°C/75%RH)

Time Cumulative % drug release (X± S.D)*


(min) Initial 1month 2 month 3 month
0 0
0 0 0
4 63.54 63.36 62.505 60.315
±0.54 ±0.623538 ±0.433964 ±0.226495
8 82.3456 82.1654 81.26445 79.71702
±0.473278 ±0.574036 ±0.468855 ±0.206467
12 91.69202 91.34662 90.23467 88.07052
±0.708338 ±0.848396 ±0.522542 ±0.406302
16 94.79372 94.35793 92.83975 90.4032
±0.768666 ±1.04017 ±0.644826 ±0.341573
20 99.54875 99.20248 98.83762 96.93837
±0.45275 ±0.751317 ±0.314367 ±0.543773
Data are expressed as mean ± S.D. (n = 3)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 162 


RESULTS

Table-53: In-vitro release profile of F18 during Stability studies at


(40°C/75%RH)

Time Cumulative % drug release (X± S.D)*


(min) Initial 1month 2 month 3 month
0 0
0 0 0
4 49.785 49.47 47.955 47.685
±0.610962 ±0.774645 ±0.655725 ±0.230922
8 63.11532 62.82997 62.03328 61.50798
±0.203281 ±0.3252 ±0.350264 ±0.2263
12 73.71538 73.54972 72.81215 72.37627
±0.473446 ±0.560165 ±0.230413 ±0.612642
16 77.08215 76.6313 75.08292 74.27155
±0.923888 ±1.217887 ±0.43459 ±0.868621
20 81.96757 81.38122 80.53612 79.46885
±0.665137 ±0.317976 ±0.48254 ±0.852313
Data are expressed as mean ± S.D. (n = 3)

Figure-50: In-vitro release profile of F4 during Stability studies at

(40°C/75%RH)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 163 


RESULTS

Figure-51: In-vitro release profile of F10 during Stability studies at

(40°C/75%RH)

Figure-52: In-vitro release profile of F15 during Stability studies at

(40°C/75%RH)

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 164 


RESULTS

Figure-53: In-vitro release profile of F18 during Stability studies at

(40°C/75% RH)

Stability studies were conducted for the formulations F4, F10, F15 and F18.

The reasons for selection is, these four formulation have shown best results in term of

weight, hardness, friability, in-vitro disintegration, drug content, in-vitro drug release

studies. Stability studies of the prepared fast dissolving tablets were performed at

different temperatures (400C/75RH). The tablets were analyzed for weight, hardness,

friability, in-vitro disintegration time, and for drug content in each formulation at a

time interval of one month for the period of three months.

All the formulations showed no significant variation in all the parameters

under the test period conditions.

DEPT. OF PHARMACEUTICS, Dr.H .L. T. COLLEGE OF PHARMACY  Page 165 


DISCUSSION

CHAPTER-6.

DISCUSSION

In the present project FDT of Ketoprofen were prepared and evaluated for

achievement of fast action of active moiety. The tablets were prepared by direct

compression method by using solid dispersion technology. Fast disintegration of

tablets was achieved by using superdisintegrants and effervescent agents. The

Ketoprofen is water insoluble drug so this is necessary to increase the water solubility

of the drug for that purpose firstly the solid dispersion of Ketoprofen were prepared

with PVP K-30 and PEG-6000 and evaluated. The optimized solid dispersion was

incorporated in FDTs. These prepared tablets were evaluated for there quality control

parameter.

The gift sample of Ketoprofen was analyzed by various organoleptic,

physicochemical and spectrophotometric methods. The sample of Ketoprofen

possesses similar color, odor, taste and texture as given in officials. The melting point

of procured sample was analyzed by capillary fusion method and found 950 C. The

FT-IR spectrum of drug sample was concordant with reference spectra as given in IP

1996. The IR spectra of reference and sample are shown in Figure 10 (A) and 11 (B)

respectively. The FT-IR spectra verified the authenticity of the procured sample,

characteristics peak of Ketoprofen are present at 1700 cm-1 and 1650 cm-1 in sample

spectra. The absorption maxima of Ketoprofen was observed at 260 nm in 5%

methanolic Sorenson’s buffer, which is concordant with the value given in IP 1996.

The UV spectra of Ketoprofen were shown in Figure 13. The DSC of drug sample

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 166 


DISCUSSION

shows a sharp endothermic peak at 94.540C, which further support the authenticity of

the procure sample.

The qualitative solubility of Ketoprofen was determined in various solvent

systems. The maximum solubility was found in methanol and ethanol and least in

distilled water. The solubility of Ketoprofen was shown in Table 22. Solubility of

Ketoprofen was also determined quantitatively in different buffers (pH), results were

shown in Table 23. The calibration curve of Ketoprofen was prepared in Sorenson’s

buffer (pH 6.8). The plot of different concentrations of Ketoprofen versus absorbance

was found linear in the concentration range of 0-25 μg/ml at 260 nm. The absorbances

at different concentrations were shown in Table 24. The data of standard curve was

linearly regressed. The slope and correlation coefficient values were found 0.0409 and

0.9991 respectively. The intercept on Y-axis found 0.0083. The calibration curve was

shown in Figure 14.

Drug-polymer interaction study was carried out and evaluated for physical

changes, change in absorption maxima and by FT-IR studies. Results were shown in

Figure 15-20. There was not any sign of physical change and change in absorption

maxima at the end of study. The FT-IR spectra of the various physical mixtures retain

all the peaks of the pure drug. So there was no significant shift in the peaks

corresponding to the drug were observed on storage. Both the drug and polymers were

compatible with each other. Hence the drug and polymers can be successfully

incorporated in the design of solid dispersion as well as fast dissolving tablets.

Physical mixtures and solid dispersions of Ketoprofen with PVP K-30& solid

dispersions of Ketoprofen with PEG-6000 (1:1 to 1:3) were prepared by solvent

evaporation technique, the prepared PM and SD were evaluated for drug content,

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 167 


DISCUSSION

solubility, FT-IR, XRD, DSC and SEM. The composition of various formulations of

physical mixtures and solid dispersion were shown in Table 8-12.

The drug content of physical mixtures (KP1-KP3 & KPG1-KPG3) and solid

dispersions (KPVP1-KPVP3 & KPEG1-KPEG3) was found to be from 97.94 to

99.37, which is found to be within the range of ±1 % of the theoretical claim (Table

27), which shows the uniformity and reproducibility of the obtained method. The

saturation solubility of pure drug, PM and SD was found to be 0.014mg/ml,

0.228mg/ml to 0.711mg/ml and 0.252 mg/ml to 0.894 mg/ml. The results were shown

in Table 26.

It was observed that the saturation solubility of drug was increased by 20 to 50

folds by converting the drug into solid dispersion, due to change in physical state of

Ketoprofen from crystalline to amorphous state which was confirmed by the IR,

XRD, DSC and SEM studies.

The comparison of FT-IR of the physical mixture of PVP K30 and drug &/

PEG-6000 and drug with the FT-IR of its individual components was done to observe

any interactions between drug and polymer. The FT-IR spectra of Ketoprofen, PVP

K30, PEG-6000, and the corresponding physical mixture are shown in Figure 11 (B)

and 23-26. The FT-IR of various mixtures revel all the peaks of the drug. As it can be

seen, a strong absorption peaks at 1696 and 1655 cm−1 occurs in the IR spectrum of

Ketoprofen. So there is no significant shift in the peak corresponding to the drug or

the polymer was observed in the physical mixtures and solid dispersions. The results

clears that there is no chemical interaction in with the chemical entity of Ketoprofen,

only the physical entrapment is observed which is further evaluated by DSC and SEM

studies.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 168 


DISCUSSION

The DSC runs for Ketoprofen, PVP K30, PEG-600 and solid dispersions

(KPVP3, KPEG3) are shown in Figure 27-30. The DSC curve for Ketoprofen showed

a sharp melting endotherm at 94.540C. The DSC curve for PEG-6000 showed a sharp

melting endotherm at 63.270C The DSC curve for PVP K30 showed glass transition

peaks at 178.140C. The Solid dispersions KPVP3 & KPEG3 showed no shift in the

melting endotherm for Ketoprofen indicating that there is no chemical interaction

between the Ketoprofen and PVP K30 &/ PEG-6000 in the solid dispersions. All the

solid dispersion exhibited no endothermic peak corresponding to the melting of

Ketoprofen indicating that the drug is dispersed amorphously in the PVP K30 and

PEG-6000 matrix.

The X-ray diffraction patterns for pure Ketoprofen, PVP K 30, PEG-6000 and

solid dispersions (KPVP3, KPEG3) are depicted in Figure 31-35. The pure drug

showed numerous sharp peaks demonstrating the crystalline nature of the drug

whereas, Polyvinylpyrolidone K 30 showed diffused peaks indicating the amorphous

nature of the polymer and PEG-6000 also showed diffused peak. Ketoprofen showed

characteristic intense peaks between the 2θ of 6.5o and 23o due to its crystalline

nature. Whereas, in case of solid dispersions, no intense peaks related to drug were

noticed between the 2θ of 6.5o and 23o. All the SD granules showed diffused peaks

indicating that the drug is dispersed at the molecular level in the polymer matrix and

hence, no crystals were found in the solid dispersion.

The SEM of pure Ketoprofen, pure PVP K30 are shown in Figure 36 and 37

respectively. It was seen that Ketoprofen is highly crystalline material. PVPK30 was

present in globular form. It was found that the crystals of Ketoprofen were dispersed

between the carriers in the KPVP3 formulations as shown in Figure 38. The change in

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 169 


DISCUSSION

crystalline nature of Ketoprofen in solid dispersion can be easily seen in SEM, which

revels that the drug is completely dispersed in polymeric matrix. The SEM also

supports data obtained from IR, XRD and DSC.

The in-vitro release profile of Ketoprofen of PVP K30 physical mixtures,

Ketoprofen of PEG-6000 physical mixture, and solid dispersions formulations KP1,

KP2, KP3, KPG1, KPG2, KPG3, KPVP1, KPVP2, KPVP3, KPEG1, KPEG2 and

KPEG3 are shown in Table 26-27 and the graph for the comparison of the cumulative

percent release is illustrated in Figure 21-22. In all the cases, cumulative percent

release was much greater than pure Ketoprofen. It is apparent from the Table and the

Figure that as the percent of carrier (PVP K30 & PEG-6000) is increased the

dissolution rate is increased. Pure Ketoprofen yield the low release due to its

hydrophobic property causing the powder to float on the surface of the dissolution

media and prevented its surface to make contact with medium for initial time

intervals.

The percentage drug dissolved from the solid dispersions of Ketoprofen-PVP

K30 & Ketoprofen-PEG-6000 was compared with that dissolved from an equal

amount of physical mixtures and pure crystalline Ketoprofen. In all cases, increasing

the proportion of carrier resulted in the enhancement of the dissolution rate. This

extent of enhancement was markedly greater for the solid dispersions in comparison

with the pure drug and physical mixtures.

Several mechanisms may be possible for the enhanced release of Ketoprofen

in the solid dispersion formulation with the water soluble polymer PVP K30 and PEG

6000. The reduction of crystallinity of drug resulting in improved release (supported

by X-ray diffraction and SEM studies).

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 170 


DISCUSSION

This suggests that a uniform distribution of drug in the polymer crust at

molecular level in a highly dispersed state. Thus, when such system comes in contact

with an aqueous dissolution medium, the hydrophilic carrier dissolves and results in

precipitation of the embedded drug into fine particles, which increases the dissolution

surface available. Moreover, other factors such as absence of aggregation and/or re-

agglomeration phenomenon during dissolution and particle size reduction may be

attributed to better dissolution profile.

The enhancement of dissolution of Ketoprofen from solid dispersions may be

due to the amorphous nature of the drug in solid dispersions.

Dissolution efficiency of pure Ketoprofen and all the physical mixtures and

solid dispersion formulations at 15 minutes and 30 minutes were calculated which is

shown in Tables 30. As the dissolution time was increased from 15 to 30 minutes, the

dissolution efficiency was increased in all the formulations. Among the formulations

KPVP3 has shown maximum dissolution efficiencies of 44.16% and 63.33% at fifteen

minutes (DE15) and thirty minutes (DE30) respectively. However, KP1, KP2, KP3,

KPG1, KPG2, KPG3, KPVP1, KPVP2, KPEG1, KPEG2 and KPEG3 also produce

comparable results on terms of dissolution efficiency.

From the above characterization of solid dispersions the KPVP3 (solid

dispersion of Ketoprofen with PVP K30 in ratio 1:3) found to be more approachable

for incorporation in fast dissolving tablets.

In the present study, total twenty drug formulations were formulated using

KPVP3 (1:3 ratio Solid dispersion). Ingredients for prepared tablets are shown in

Table 13-16. The formulated blends and prepared tablets were examined for their

physical properties.
DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 171 
DISCUSSION

The characterization of mixed blend was done for determination of mass

volume relationship parameters. The evaluated parameters are bulk density, tapped

density, Hausners ratio, Compressibility index, angle of repose.

The bulk density of mixed blend varied between 0.553 to 0.681 gm/cm3. The

results indicating good packaging capacity of tablets. The tapped density was found in

the range of 0.630 to 0.783 gm/cm3. By using these two density data Hausners Ratio

and compressibility index was calculated. If the bed particle is more compressible

then the powder will be less flowable and vice versa. Material having value less than

16% termed as free flow materials. The powder blends of all formulation had

Hausners ratio of 1.2 or less indicating the good flowablity. The compressibility index

was found between 10.782 to 14.542 %. The compressibility-flowability correlation

data indicating a good flowability of the powder blend.

The flowability of the powder was also evidenced by the angle of repose. The

angle of repose is below than 30o range good to excellent flow properties of powder.

Lower the friction occurring within the mass and the better flow rate. The angle of

repose was found to be 20.72-26.11o. The results for characterization of blend were

shown in Table 32. This indicates the good flow property of the formulated mixed

blend due to the addition of talc as lubricant and magnesium stearate as glidant in the

2% w/w and 2% w/w of the tablet weight respectively.

After compression of powder, the tablets were evaluated for their organoleptic

(Color, Odour, Taste), physical (Size, Shape and Texture) and quality control

parameters (Diameter, Thickness, Hardness, Friability, Disintegration Time and

Wetting Time). All the formulations were white in color, odorless, flat in shape with

smooth surface not having any defects.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 172 


DISCUSSION

The thickness of the tablet was found 6.321-6.372 mm. The average weight of

the prepared tablet was found 496.67-502.67 mg. So it was predicted that all the

tablets exhibited uniform weight with low standard deviation values within the

acceptable variation as per IP. The results are shown in Table 33. The friability of all

the formulations was found to be less than 1.0 %, which indicates the tablet’s ability

to withstand abrasion in handling, packaging and shipment. The hardness of tablet

was varied from 2.5-3.6 kg/cm2, which has satisfactory strength to withstand with the

applied mechanical shocks. Friability of F5, F19 and F20 are found very near to one

so these formulations are not forwarded for further studies.

A disintegrant was incorporated in all the formulations to facilitate a breakup

or disintegration of the tablet when it contacts with water or saliva in mouth.

Disintegrants drawing the water into the tablet causes swelling and burst apart. In the

formulation of fast dissolving tablet the three superdisintegrants (croscarmallose

sodium, Sodium Starch Glycolate and Crospovidone) and effervescent agent (Citric

acid and Sodium bicarbonate) were used in different concentrations. The tablets with

Crospovidone disintegrate faster then the tablets with the Citric acid, Sodium Starch

Glycolate and croscarmalose sodium. The tablets prepared with effervescent

technology elaborates the carbon di oxide gas when the tablet comes in contact with

little amount of saliva or water due to reaction between citric acid and sodium

bicarbonate which results in breakup of tablets. The disintegration time of all the

formulations were found between 25.68±1.41 sec to134.22±5.16 Sec. The results

were shown in Table 34. The disintegration process of the tablet was fully dependable

on nature and concentration of used superdisintegrant. The in-vitro wetting time was

also studied to know the time required for complete wetting of tablets when placed on

tongue. The in-vitro wetting time of all the formulations were varied between

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 173 


DISCUSSION

27.45±1.40 to 125.66±5.76. The results were shown in Table 34. The swelling

properties of the superdisintegrant were depend upon their concentration and the

results shows that as the concentration of the superdisintegrant increased the time

taken for swallowing was reduced. The swelling time was rapid in Crospovidone

followed by citric acid, croscarmalose sodium and Sodium starch glycolate. The same

sequence was observed in case of measurement of dispersion time of the tablet. The

results were tabulated in 34.

The drug content of all the tablet formulations was determined

spectrophotometrically at 260 nm. It varied from 48.65833 ±0.146487 to 50.54167

±0.052042 mg per tablet. The correlation of variation was found to be less than 0.5,

indicating uniformity of the drug content in the prepared tablets. The results of the

content uniformity and percent drug content were shown in Table 35.

In-vitro drug release experiments were performed at 37±1oC in eight basket

dissolution apparatus. The results of dissolution profile are shown in Table 36-39 and

Figure 42-45. The maximum drug release was found in formulation F15 (99.56%).

The order of drug release was found to be:

F15>F20>F14>F10>F13>F5>F19>F9>F4>F12>F3>F8>F18>F11>F2>F17>F16>

F7>F1>F6

Formulations F5, F10, F15 and F20 which contains 5% of Croscarmallose

sodium, Sodium starch glycolate, Crospovidone and citric acid + sodium bicarbonate

respectively. The release was estimated after twenty minutes was 90.559%, 91.293%,

99.548% and 94.751% respectively. The formulation with Crospovidone shows more

release than the tablets with Citric acid, Croscarmallose sodium and Sodium starch

glycolate.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 174 


DISCUSSION

The experiment proves that the disintegration is also a release rate-limiting

step for the drug release. The disintegrant Crospovidone shows the faster

disintegration than other. So release of drug and release rate was higher from these

tablets. From the observed data, it can be clear that less time in disintegration enhance

the release rate of Ketoprofen from Fast Dissolving Tablet

Next the release data obtained were subjected for the kinetic treatment to

know the type and order of drug release.

The obtained data from in-vitro Drug release study are tabulated and

represented graphically as:

(a) Cumulative percentage drug release v/s Time (Zero order release

kinetics)

(b) Log cumulative percentage drug retained v/s Time (First order release

kinetics)

The zero order kinetics models data are shown in Table 36-39, and graphically

as Figure in 42-45.

The first order kinetics model data are shown in Table 40-43, and graphically

as Figure 46-49.

For the in-vitro drug release profile it is evident that the kinetics of drug

release is first order for all the prepared fast dissolving tablets as the plot between Log

cumulative percent drug retained versus Time showed the good linearity of ‘R2’

obtained was near to one. The prepared final preparation gives better in terms of

release profile.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 175 


DISCUSSION

Formulations which show maximum drug release from each disintegrants and

also having friability less than 0.9 % were selected for the stability studies. These

include F4, F10, F15 and F18.

Stability studies of the prepared fast dissolving tablets were performed at

temperatures (400C/75RH). The tablets were analyzed for weight, hardness, friability,

in-vitro disintegration time, and for drug content in each formulation at a time interval

of one month for the period of three months.

All the formulations showed no significant variation in all the parameters

under the test period conditions.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 176 


CONCLUSION

CHAPTER-7.

CONCLUSION

The data obtained from the study of “Formulation and evaluation of

fast dissolving tablets of Ketoprofen” reveals following conclusion:

For improved the solubility of Ketoprofen solid dispersion was prepared by

using PVP K30 and PEG 6000 in different drug: polymer ratio (1:1 to 1:3) by

solvent evaporation method.

Solid dispersions were evaluated for solubility, percent drug content,

dissolution efficiency, in-vitro drug release studies and drug polymer

interaction studied by FT-IR, DSC, XRD, and surface morphology by SEM,.

Formulations containing Ketoprofen-PVP K30 solid dispersions in 1:3 ratio

(KPVP3) showed better dissolution rate, dissolution efficiency. And on the

basis of in-vitro release formulation (KPVP3) was chosen for FDT.

Fast dissolving tablets were prepared by adding different concentrations

(1-5%) of superdisintegrants and by effervescent technology.

The characterization of mixed blend was carried out for all formulation.

The bulk density of mixed blend varied between 0.553±0.001621 to 0.681

±0.001094 gm/cm3.

The tapped density was found in the range of 0.630 ±0.00243 to 0.783

±0.001873 gm/cm3.

The powder blends of all formulation had Hausners ratio less than 1.2

indicating the good flowablity.

The compressibility index was found between 10.298±0.222876 to 14.542

±0.16249%.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 177 


 
CONCLUSION

The angle of repose was found between 20.72±0.51672 to 26.11o±0.462065.

The tablets were prepared & characterized.

The weight of the tablets of all formulation was found to be 496.67±1.527525

to 502.6667±1.527525 mg.

The friability of all formulation was found to be 0.440411±0.000269 to

0.958467±0.000383%.

The hardness of all formulation was found to be 2.5±0.173205 to 3.6±0.1

kg/cm2.

The disintegration time of all the formulations were found between

25.68±1.41 to134.22±5.16 secs.

The in-vitro wetting time of all the formulations were varied between

27.45±1.40 to 125.66±5.76 secs.

The dispersion time of all formulation were varied between 30.91±1.681547 to

142.7133±4.83864 secs.

Drug content of all formulation varied from 48.65833 ±0.146487 to 50.54167

±0.052042 mg per tablet.

The order of drug release was found to be: F15>F20>F14>F10>F13>F5>

F19>F9>F4>F12>F3>F8>F18>F11>F2>F17>F16>F7>F1>F6

Formulations F5, F10, F15 and F20 which contains 5% of Croscarmallose

sodium, Sodium starch glycolate, Crospovidone and citric acid + sodium

bicarbonate respectively. The release was estimated after twenty minutes was

90.559%, 91.293%, 99.548% and 94.751% respectively. The formulation with

Crospovidone shows more release than the tablets with Citric acid + Sodium

bicarbonate, Croscarmallose sodium and Sodium starch glycolate.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 178 


 
CONCLUSION

Stability study for F4, F10, F15 and F18 were performed at temperatures

(400C/75%RH). All the formulations showed no significant variation in all the

parameters evaluated under the test period condition.

From the above studies it was concluded that the F15 is best formulation for

Fast Dissolving Tablets of Ketoprofen.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 179 


 
SUMMARY

CHAPTER-8.

SUMMARY

Compounds with poor aqueous solubility are extremely challenging to be

developed as new formulations. It is well known that drug dissolution rather than

permeation through the epithelia of the gastrointestinal tract is responsible for a low oral

absorption. One of the pharmaceutical strategies to improve the oral bioavailability that

is solid dispersions. Ketoprofen was selected a model drug for the research work because

it has a poor aqueous solubility and low dissolution rate, while high permeability.

The Ketoprofen was formulated as physical mixture and solid dispersions with

PVP K30 & PEG-6000 in different ratios in order to improve the drug dissolution.

Ketoprofen-PVP K30 solid dispersions & Ketoprofen- PEG-6000 solid dispersions were

prepared by solvent evaporation technique.

Solid dispersions were evaluated for solubility, percent drug content, dissolution

efficiency (DE15 and DE30), in-vitro drug release studies and characterized by FT-IR,

DSC, XRD, SEM.

Formulations containing Ketoprofen-PVP K30 solid dispersions in 1:3 ratio

(KPVP3) prepared by solvent evaporation technique showed better dissolution rate and

dissolution efficiency. And on the basis of in-vitro drug release study formulation

(KPVP3) was selected for preparation of FDT.

Fast dissolving tablets were prepared

• by adding different concentrations (1-5%) of superdisintegrants

• by effervescent technology

The disintegration properties of tablet were observed as Crospovidone > Citric

acid + Sodium bicarbonate >Sodium starch glycolate > Ac-Di-Sol. The rapid drug

DEPT. OF PHARMACEUTICS, Dr.. H. L. T. COLLEGE OF PHARMACY.  Page 180 


SUMMARY

dissolution might be due to the easy and fast breakdown of tablet and rapid absorption

of drug into the dissolution media.

The drug release was found as F15>F20>F14>F10>F13>F5>F19>F9>F4>

F12> F3>F8>F18>F11>F2>F17>F16>F7>F1>F6.

The selected tablet formulations which possess the best physical quality were

Croscarmallose sodium 4% w/w (F4), Sodium starch glycolate 5% w/w (F10),

Crospovidone 5% w/w (F15) and Sodium bicarbonate + citric acid (1:2) 5% w/w

(F18). All prepared tablets followed First order release.

Stability study for F4, F10, F15 and F18 were performed at temperatures

(400C/75%RH). All the formulations showed no significant variation in all the

parameters evaluated under the test period condition.

On experimental data it was concluded that Fast dissolving tablet of

Ketoprofen would be an effective alternative approach for management of various

inflammatory disorders and pain. Formulation (F15) containing 5% w/w

Crospovidone is the most promising formulation for rapid release of Ketoprofen.

DEPT. OF PHARMACEUTICS, Dr.. H. L. T. COLLEGE OF PHARMACY.  Page 181 


BIBLIOGRAPHY

CHAPTER-9.

BIBLIOGRAPHY

1. D.M.Brahmankar, Sunil B. Jaiswal, Biopharmaceutics and Pharmakokinetics A

Treatise, 1st ed, Vallabh Prakasan, Delhi, 2005; 27,5-6.

2. James Swarbrick., James C Boylan., Encyclopedia of Pharmaceutical

Technology, 2nd edition, Vol-1:8.

3. V.Venkateswarlu. Biopharmaceutics and Pharmakokinetics, 2nd edition,

PharmaMed Press, Hyderabad, 2010: 5-6.

4. Dhirendra K., Lewis S., Udupa N., Atin K. Solid dispersions: A review. Pak. J.

Pharm. Sci., 2009; 22(2): 234-246.

5. Christian Leuner, Jennifer Dressman. Improving drug solubility for oral delivery

using solid dispersions. European Journal of Pharmaceutics and

Biopharmaceutics, 2000; 50: 47-60.

6. Duncan Q.M. Craig. The mechanisms of drug release from solid dispersions in

water-soluble polymers. International Journal of Pharmaceutics, 2002; 231:

131–144.

7. Teo filo Vasconcelos, Bruno Sarmento, Paulo Costa. Solid dispersions as

strategy to improve oral bioavailability of poor water soluble drugs. Drug

Discovery Today, 2007; 12: 1068-1078.

8. Kuchekar BS, Bhise SB, Arungam V. Design of Fast Dissolving Tablets. Indian

J Pharm Educ, 2005; 35: 150.

9. Chein YW. Oral drug delivery and delivery systems, 2nd ed., New York: Marcel

Dekker; 1992.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 182 


BIBLIOGRAPHY

10. Habib W, Khankari R, Hontz J. Fast-Dissolve Drug Delivery System. Crit Rev

Ther Drug Carrier Systems, 2000; 17(1): 61-72.

11. Srikonda V. Sastry, Janakiram Nyshadham. Process development and scale-up

of oral fast dissolving tablets. Drug delivery to the oral cavity, CRC Press, Boca

Raton,2005;311-334.

12. Suresh Bhandari, Rajendar Kumar Mittapalli, Ramesh Gannu, Vamsani

Madhusudan Rao. Orodispersible tablets: an overview. Asian journal of

pharmaceutics, 2008; 2(1): 2-11.

13. Srikonda V. Sastry, Janakiram Nyshadham. Process development and scale-up

of oral fast dissolving tablets. Drug delivery to the oral cavity, CRC Press, Boca

Raton,2005; 313-315.

14. Panigrahi D., Baghel S., Mishra B. Mouth dissolving tablets: An overview of

preparation, evaluation and patented technologies. Journal of Pharmaceutical

Research, 2005; 4(3): 33-38.

15. A Gupta, A. K. Mishra, V. Gupta, P. Bansal, R. Singh, A. K. Singh. Recent

Trends of Fast Dissolving Tablet - An Overview of Formulation Technology,

International Journal of Pharmaceutical & Biological Archives, 2010; 1(1): 1-

10.

16. Susijit Sahoo, B. Mishra, P. K. Biswal, Omprakash Panda, Santosh Kumar

Mahapatra, Goutam Kumar Jana. Fast Dissolving Tablet: As A Potential Drug

Delivery System, Drug Invention Today, 2010, 2(2): 130-133.

17. Troy M. Harmon. Orally disintegrating tablets: Avaluable life cycle

management stratergy. packaging drug delivery, pharmaceutical commerce,

www.Pharmaceuticalcomerce.com

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 183 


BIBLIOGRAPHY

18. Debjit Bhowmik, Chiranjib B., Krishnakanth, Pankaj, R. Margret Chandira. Fast

dissolving tablet: An overview. Journal of Chemical and Pharmaceutical

Research, 2009; 1(1): 163-177.

19. Jaysukh J. Hirani, Dhaval A. Rathod, Kantilal R. Vadalia. Orally disintegrating

tablets: A review. Tropical journal of pharmaceutical research, 2009; 8(2): 161-

172.

20. Arya Arun, Chandra Amrish. Fast Drug Delivery Systems: A Review. Der

Pharmacia Lettre, 2010; 2(2): 350-361.

21. Rakesh Pahwa, Mona Piplani, Prabodh C. Sharma, Dhirender Kaushik and

Sanju Nanda. Orally Disintegrating Tablets - Friendly to Pediatrics and

Geriatrics. Archives of Applied Science Research, 2010; 2(2): 35-48.

22. Wayne Camarco, Dipan Ray, Ann Druffner. Formulation selecting

superdisintigrants for orally disintegrating tablet formulations. Supplement to

pharmaceutical Technology Exipient Performance, 2006; 1-6.

23. Md.Nehal Siddiqui, Garima Garg, Pramod Kumar Sharma. Fast dissolving

tablets: Preparation, characterization and evaluation: An overview. International

Journal of Pharmaceutical Sciences Review and Research, 2010; 4(2): 87-97.

24. Dali Shukla, Subhashis Chakraborty, Sanjay Singh, Brahmeshwar Mishra.

Mouth dissolving tablets-I: An overview of formulation technology. Sci pharm.,

2009; 76: 309-326.

25. Rajan K. Verma, Sanjay Garg. Current status of drug delivery technologies and

future directions. Pharmaceutical Technology on line, 2001; 25(2): 1-14.

26. Srikonda Venkateswara Sastry, Janaki Ram Nyshadham and Joseph A. Fix.

Recent technological advances in oral drug delivery – a review. PSTT, 2000;

3(4): 138-146.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 184 


BIBLIOGRAPHY

27. Suresh Bhandari, Rajendar Kumar Mittapalli, Ramesh Gannu, Vamsani

Madhusudan Rao. Orodispersible tablets: an overview. Asian journal of

pharmaceutics, 2008; 2(1): 2-11.

28. Kuldeepak Sharma, William R. Pfister, Tapash K. Ghosh. Quick-Dispersing oral

drug delivery systems. Drug delivery to the oral cavity, CRC Press, Boca

Raton,2005; 261-289.

29. S. Indiran Pather, Rajendra Khankri, John Siebert. Quick-Dissolving intraoral

tablets. Drug delivery to the oral cavity, CRC Press, Boca Raton,2005; 291-309.

30. Late S. G., Yi-Ying Y., Banga A. K. Effects of Disintegration-Promoting Agent,

Lubricants and Moisture Treatment on Optimized Fast Disintegrating Tablets.

Int J Pharm, 2009; 365: 4–11.

31. Ahmed I. S., Nafadi M. M., Fatahalla F. A. Formulation of Fast-Dissolving

Ketoprofen Tablet Using Freeze-Drying in Blisters Technique. Drug Dev Ind

Pharm, 2006; 32(4): 437- 442.

32. Maria Victoria Margarit, Antonio cerezo, physical charecteristics and

dissolution kinetics of solid dispersion of Ketoprofen and polyethylene glycol

6000. Int J Pharm, 1994; 108: 101-107.

33. Piera Di Martino, Esienne Joiris, Roberto Gobetto, Amir Masic, Giovanni F.

Ketoprofen-poly(vinylpyrrolidone) physical interaction. J. of crystal growth,

2004; 256: 302-308.

34. R. Jachowicz, E. Nurnberg, B. Pieszczek, B. Kluczykowska, A. Maciejewska.

Solid dispersion of Ketoprofen in pellets. Int J Pharm, 2000; 206: 13-21.

35. Vasanthkumar S., Vijaya Raghavan Chellan. Immediate release tablets of

Telmisartan using superdisintigrant- formulation, evaluation and stability

studies. Chem. Pharm. Bull, 2008; 56(4): 575-577.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 185 


BIBLIOGRAPHY

36. Malleswara Rao V. S. N., Shyam T., Appa Rao B., Srinivasa Rao Y.,

Formulation and characterization of Meloxicam solid dispersions. Indian

Pharmacist, 2008; April: 67-70.

37. P. G. Patil, V. R. Patil. Enhancement of water solubility of Felodipine by

preparing solid dispersion using poly-ethylene glycol 6000 and poly-vinyl

alcohol. Asian journal of pharmaceutics, 2009; July-Sept: 240-244.

38. Dario Leonardi, Maria Gabriela Barrera, Maria Celina Lamas and Claudio

Javier Salomon. Development of Prednisone: polyethylene glycol 6000 fast-

release tablets from solid dispersion: solid-state characterization, dissolution

behavior, and formulation parameters. AAPS PharmaSciTech, 2007; 8(4): E1-

E8.

39. M. M .Patel, D. M. Patel. Fast dissolving Valdecoxib tablets containing solid

dispersion of Valdecoxib, Indian journal of pharmaceutical science, 2006; 3:

222-226.

40. D. Nagendrakumar, S. A. Raju, S. B. Shirsand, M. S. Para and M. V. Rampure.

Fast dissolving tablets of Fexofenadine Hcl by effervescent method. Indian

Journal of Pharmaceutical Sciences, 2009; March-April: 116-119.

41. S. B. Shirsand, Sarasija Suresh, M. S. Para and P. V. Swamy. Design of fast

disintegrating tablets of Prochlorperazine Maleate by effervescence method.

Indian Journal of Pharmaceutical Sciences, 2008; July-August: 447-451.

42. Bhalerao A. V., Deshkar S.S., Shirolkar S.V., Gharge V.G., Deshpande A. D.,

Development and Evaluation of Clonzepam Fast Disintigrating Tablets Using

Superdisintigrates and Solid Dispersion Technique. Research J.Pharm. and

Tech., 2009; 2(2): 375-377.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 186 


BIBLIOGRAPHY

43. Jashanjit Singh and Rajmeet Singh. Optimization and Formulation of

Orodispersible Tablets of Meloxicam. Tropical Journal of Pharmaceutical

Research, 2009; 8(2): 153-159.

44. Radke R. S., Jadhav J. K., Chajeed M. R. Formulation and Evaluation of

Orodispersible Tablets of Baclofen. International Journal of ChemTech

Research, 2009; 1(3): 517-521.

45. Gangane P.S., Mahajan K. G., Sawarkar H. S., Thenge R. R., Adhao V. S.

Formulation, Characterization and Evaluation of Rapid Disintegrating Tablet of

Gatifloxacin Sesquihydrate by ion exchange resin technique. IJPRIF, 2009;

1(4): 1212-1218.

46. Venkatesh D. P., Geetha Rao C. G. Formulation of taste masked oral-dispersible

tablets of Ambroxol hydrochloride, AJPS, 2008; 2: 261-264.

47. Patel D. M., Patel M. M., Optimization of Fast Dissolving Etoricoxib Tablets

Prepared by Sublimation Technique. Indian Journal of Pharmaceutical

Sciences, 2008; 71-76.

48. Furtado S, Deveswaran R, Bharath S, Basavaraj B. V., Abraham S. and

Madhavan V. Development and characterization of orodispersible tablets of

Famotidine containing a subliming agent. Trop J Pharm Res, 2008; 7(4): 1185-

1189.

49. Takao Mizumoto, Tetsuya Tamura, Hitoshi Kawai, Atsushi Kajiyama, Shigeru

Itai. Formulation Design of an Oral, Fast- Disintegrating Dosage Form

Containing Taste- Masked Particles of Famotidine. Chem. Pharm. Bull, 2008;

56(7): 946-950.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 187 


BIBLIOGRAPHY

50. Sajal Kumar Jha, Vijayalakshmi P., Roopa Karki, Divakar Goli, Formulation

and evaluation of melt-in-mouth tablets of Haloperidol. Asian Journal of

Pharmaceutics, 2008; 255-260.

51. Prabhu Namita B., Rao Leena and Amin Purnima. D. Studies on taste masking

of Drotaverine Hydrochloride and its formulation. Indian Drugs, 2007; 44(11):

848-851.

52. Mukesh C. Gohel, Rajesh K. Parikh, Bansari K. Brahmbhatt, Aarohi R. Shah.

Preparation and assessment of novel coprocessed superdisintegrant consisting of

Crospovidone and Sodium Starch Glycolate. AAPS PharmSciTech, 2007; 8(1):

E1-E7.

53. Jacob S., Shirwaikar A. A., Joseph A., Srinivasan K. K., Novel Co-Processed

excipients of Mannitol and Microcrystalline Cellulose for preparing fast

dissolving tablets of Glipizide. IJPS, 2007; 633-639.

54. Dina Nath Mishra, Madhu Bindal, Shailendra Kumar Singh, Sengodan

Gurusamy, Spray Dried Excipient Base : A Novel Technique for the

Formulation of Orally Disintegrating Tablets. Chem. Pharm. Bull., 2006; 54(1):

99-102.

55. Jinichi Fukami, Etsuo Yonemochi, Yasuo Yoshihashi, Katsuhide Terada,

Evaluation of rapidly disintegrating tablets containing glycine and

carboxymethylcellulose, International Journal of Pharmaceutics, 2006; 310:

101-109.

56. Chaudhari P. D., Kolhe S. R., Chaudhari S. P., More D. M., Use of Cation

Exchange Resins to Optimize The Taste Masking of Bitter Drug Rizatriptan

Benzoate. Indian Drugs, 2006; 43(10): 795-799.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 188 


BIBLIOGRAPHY

57. Gohel M. C. A review of co-processed directly compressible excipients. J.

Pharma Pharmaceut Sci., 2005; 8(1): 76-93.

58. Na Zhao, Larry L. Augsburger. Functionality Comparison of 3 Classes of

Superdisintegrants in Promoting Aspirin Tablet Disintegration and Dissolution.

AAPS PharmSciTech, 2005; 6(4): E634-E640.

59. Abdelbary G., Prinderre P., Eouani C., Joachim J., Reynier J. P. The preparation

of orally disintegrating tablets using a Hydrophilic waxy binder. International

Journal of Pharmaceutics, 2004; 278: 423-433.

60. Damrongsak Faroongsarng, Garnet E. Peck, Thermal Porosity Analysis of

Croscarmellose Sodium and SodiumStarchGlycolate by Differential Scanning

Calorimetry, AAPS PharmSciTech, 2003; 4(4): 1- 8.

61. Toshihiro Shimizu, Masae Sugaya, Yoshinori Nakano, Daisuke Izutsu, Yoshio

Mizukami, Formulation Study for Lansoprazole Fast-Disintegrating Tablet.III.

Design of Rapidly Disintegrating Tablets. Chem. Pharm. Bull., 2003; 51(10):

1121-1127.

62. Toshihiro Shimzu, Yoshinori Nakano, Shuji Morimoto, Tetsuro Tabata, Naoru

Hamaguchi, Yasutaka Igari, Formulation Study for Lansoprazole Fast-

Disintegrating Tablet-I: Effect of Compression on Dissolution Behavior. Chem.

Pharm. Bull., 2003; 51(8): 942-947.

63. Yutaka Morita, Yuki Tsushima, Masanobu Yasui, Ryoji Termoz, Junko Ajioka.

Evaluation of the Disintegration Time of Rapidly Disintegrating Tablets via a

Novel Method Utilizing a CCD Camera. Chem.Pharm. Bull., 2002; 50(9): 1181-

1186.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 189 


BIBLIOGRAPHY

64. Hector Fausett, Charles Gayser Jr., Alekha K. Dash, Evaluation of Quick

Disintegrating Calcium Carbonate Tablets. AAPS PharmaSciTech, 2000; 1(3):

article 20.

65. Chaulang G., Patel P., Hardikar S., Kelkar M., Bhosale A., Bhise S. Formulation

and Evaluation of Solid Dispersions of Furosemide in Sodium Starch Glycolate.

Trop J Pharm Res, 2009; 8: 43-51

66. Madan J., Sharma A. K., Singh R. Fast Dissolving Tablets of Aloe Vera Gel.

Trop J Pharm Res, 2009; 8: 63-70

67. Mohapatra A., Parikh R. K., Gohel M. C. Formulation, Development and

Evaluation of Patient Friendly Dosage Forms of Metformin, Part-I: Orally

Disintegrating Tablets. Asi J Pharm, 2008; July-September: 167-171.

68. Mohammad B. J., Hadi V., Siavoush D., Mohammad R., Siahi S., Azim B. J.,

Khosro A., Mohammad G. Enhancing Dissolution Rate of Carbamazepine via

Cogrinding with Crospovidone and Hydroxypropylmethylcellulose. Iran J

Pharm Res, 2007; 6(3): 159-165.

69. Fars K. A. Evaluation of Spray and Freeze Dried Excipients Bases Containing

Disintegration Accelerator for the Formulation of Metoclopramide Orally

Disintegrating Tablets. Saudi Pharmaceutical Journal, 2007; 15: 105.

70. Malke S., Shidhaye S., Kadam V.J. Formulation and Evaluation of

Oxcarbazepine Fast Dissolving Tablets. Indian J Pharm Sci, 2007; 69(2): 211-

214.

71. Modi A., Tayade P. Enhancement of Dissolution Profile by Solid Dispersion

(Kneading) Technique. AAPS Pharm Sci Tech, 2006; 7(3): 68.

72. Shirwaikar A. A., Ramesh A. Fast disintegrating Tablets of Atenolol by Dry

Granulation Method. Indian J Pharm Sci, 2004; 66(4): 422-426.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 190 


BIBLIOGRAPHY

73. Ravi Kumar, M. B. Patil, Sachin R. Patil. Development and characterization of

orodispersible tablets of Aceclofenac by sublimation technique, International

journal of pharmaTech Research, 2009; 4: 210-214.

74. D. M. Patil, N. M. Patil, R. R. Shah, P. D. Jogani, Studies in formulation of

orodispersible tablets of Rofecoxib, Indian journal pharmaceutical sciences,

2004; 7: 621-625.

75. Manivannam Rangasamy, Balasubramaniam Ayyasamy, Senthilkumar Raju,

Design and evaluation of the fast dissolving tablets of Terbutaline Sulphate,

Asian journal of pharmaceutics, 2009; 6: 215-217.

76. Sarasija Suresh, V. Pandit, H. P. Joshi. Preparation and evaluation of mouth

dissolving tablets of Salbutamol Sulphate, Indian journal of pharmaceutical

science, 2007; 5: 467-469.

77. H. S. Mahajan, B. S. Kuchkar and A. C. Badhan. Mouth dissolving tablets of

Sumatriptan succinate, Indian journal of pharmaceutical science, 2004; 3: 238–

240.

78. Shailesh Sharma, G. D. Gupta. Formulation and characterization of fast

dissolving tablets of Promethazine Theoclate, Asian journal of pharmaceutics,

2009; 1: 70-72.

79. A.S. Mundada, D. R. Meshram, H. B. Banbale, M. R. Bhalekar, J. G. Avari.

Formulation and evaluation of dispersible taste masked tablets of

Roxithromycin. Asian journal of pharmaceutics, 2008; April: 116-119.

80. Goodman, Gilman C. The pharmacological basis of therapeutic. 9th ed., The

McGraw-Hill Companies Inc; 1996.

81. www.wikipedia.com

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 191 


BIBLIOGRAPHY

82. Indian pharmacopoeia, Government of India, Ministry of Health and Family

welfare. The controller of publications, Delhi; Vol II: 424.

83. http://www.rxlist.com/cgi/generic/ketoprofen.htm29/07/09

84. https://www.medicinescomplete.com/ahfs/ketoprofen.htm29/07/09

85. Satoskar R. S., Bhandarkar S. D., Ainapure S. S. Pharmacology and

pharmacotherapeutics. 17th ed. Mumbai: Popular prakashan publishers; 2001:

209-218.

86. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 208-210

87. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 206-208

88. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 663-666

89. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 629-633.

90. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 181-183

91. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 517-522

92. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 581-585

93. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 129-133

94. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 364-369.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 192 


BIBLIOGRAPHY

95. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 222-225

96. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 404-407

97. Raymond C. Rowe. Paul J Sheskey, Marian E Quinn. Handbook of

pharmaceutical excipient, sixth edition. Pharmaceutical Press. 2009; 728-731.

98. Indian pharmacopoeia. Vol I, The controller of publication 2007; 606-611.

99. Dhingra Dipti, Bhandari Anil, Sharma R.B., Gupta Ranjana, Gupta Sachin

Enhancement of dissolution rate of slightly soluble drug Clomiphene Citrate by

Solid Dispersion. International Journal of PharmTech Research. 2010; 2(3):

1691-1697.

100. Gopal Venkatesh Shavi, Averineni Ranjith Kumar, Yogendra Nayak Usha,

Karthik Armugam, Om prakash Ranjan, Kishore Ginjupalli, Sureshwar Pandey,

Nayababhirama Udupa. Enhanced dissolution and bioavailability of Gliclazide

using solid dispersion techniques. International Journal of Drug Delivery, 2010;

2: 49-57.

101. S. Muralidhar, G. Devala Rao, V. Rajesh Babu, T. Karunakar Reddy and

Syeda Rana Nikhat. Fast dissolving Celecoxib tablets containing solid

dispersion of Celecoxib. IJPSR, 2010; 1(9): 34-40.

102. Margret Chandira R., Jaykar B., Chakrabarty B. L. Formulation and evaluation

of Orodispersible tablets of terbutaline sulphate. Drug Invention Today. 2010;

2(1): 31-33.

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 193 


BIBLIOGRAPHY

103. Suhas M. Kakade, Vinodh S. Mannur, Ketan B. Ramani, Ayaz A. Dhada,

Chirag V. Naval, Avinash Bhagwat. Formulation and evaluation of mouth

dissolving tablets of losartan potassium by direct compression techniques. Int. J.

Res. Pharm. Sci. 2010; 1(3): 290-295.

104. Debjit Bhowmik, B. Jayakar, K. Sampath Kumar. Design and Characterisation

of Fast Dissolving Tablet of Telmisartan. International Journal of Pharma

Recent Research. 2009; 1(1): 31-40.

105. C.V.S.Subrahmanyam. Textbook of Physical Pharmaceutics. Vallabha

Prakashan, Delhi. 2006; 13-71.

106. ICH Harmonized Tripartite Guidelines; 2003 stability testing of new drug

substances and product Q1A(R2).

DEPT. OF PHARMACEUTICS, Dr. H. L. T. COLLEGE OF PHARMACY.  Page 194 


ANNEXURES 
 

DEPT. OF PHARMACEUTIC, Dr.H.L.T COLLEGE OF PHARMACY.  Page 195 


 
ANNEXURES 
 
 

DEPT. OF PHARMACEUTIC, Dr.H.L.T COLLEGE OF PHARMACY.  Page 196 


 

You might also like