You are on page 1of 160

DEVELOPMENT AND EVALUATION OF CONTROLLED

RELEASE MATRIX TABLETS CONTAINING PERINDOPRIL IN


THE TREATMENT OF HYPERTENSION

By

N. SRIKANTH REDDY
B. Pharm.

DISSERTATION SUBMITTED TO
RAJIV GANDHI UNIVERSITY OF HEALTH SCIENCES,
BENGALURU, KARNATAKA.

IN PARTIAL FULFILLMENT
FOR THE AWARD OF THE DEGREE OF

MASTER OF PHARMACY
IN
PHARMACEUTICS

Under the Guidance of


Dr. SOMASHEKAR SHYALE
M.Pharm., Ph.D.

Professor
DEPARTMENT OF PHARMACEUTICS
V. L. COLLEGE OF PHARMACY
RAICHUR – 584103.

2011

i
‘I DEDICATE THIS BOOK TO MY DAD, MOM AND

DEAR SISTER’

‘TO GAYATHRI LAKSHMI.B WHO HAVE BEEN AN

IMMENSE SUPPORT AND BELIEVING IN ME’

ii
Rajiv Gandhi University of Health Sciences, Bengaluru, Karnataka.

DECLARATION BY THE CANDIDATE

I hereby declare that this dissertation entitled “Development and evaluation

of controlled release matrix tablets containing perindopril in the treatment of

hypertension” is a bonafide and genuine research work carried out by me under the

guidance of Dr. Somashekar Shyale, Professor, Department of Pharmaceutics, V.L.

College of Pharmacy, Raichur.

Date:

Place: Raichur (N.Srikanth Reddy)

iii
Rajiv Gandhi University of Health Sciences, Bengaluru, Karnataka.

CERTIFICATE BY THE GUIDE

This is to certify that the dissertation entitled “Development and evaluation

of controlled release matrix tablets containing perindopril in the treatment of

hypertension” is a bonafide research work done by Mr. N.Srikanth Reddy in partial

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

Pharmaceutics under my guidance at V.L. College of Pharmacy, Raichur.

Date: Dr. Somashekar Shyale


M.Pharm.,Ph.D.
Place: Raichur Professor, Dept. of Pharmaceutics,
V.L College of Pharmacy,
Raichur-584103.

iv
Rajiv Gandhi University of Health Sciences, Bengaluru, Karnataka.

ENDORSEMENT BY THE HOD, PRINCIPAL/HEAD OF THE INSTITUTION

This is to certify that the dissertation entitled “Development and evaluation

of controlled release matrix tablets containing perindopril in the treatment of

hypertension” is a bonafide research work done by Mr. N.Srikanth Reddy under

the guidance of Dr. Somashekar Shyale at V.L. College of Pharmacy, Raichur.

Dr. Y. Anand Kumar Dr. S.M Shanta Kumar


M.Pharm., Ph.D. M.Pharm., Ph.D.
Professor & Head, Principal,
Dept. of Pharmaceutics, V.L. College of Pharmacy,
V.L. College of Pharmacy, Raichur -584103.
Raichur -584103.

Date: Date:
Place: Raichur Place: Raichur

v
COPYRIGHT

Declaration by the Candidate

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

shall have the rights to preserve, use and disseminate this dissertation /thesis in print

or electronic format for academic /research purpose.

Date:

Place: Raichur (N.Srikanth Reddy)

© Rajiv Gandhi University of Health Science, Bengaluru, Karnataka.

vi
ACKNOWLEDGEMENTS

In the name of Lord Venkateshwara swamy, I bow myself to thank and acknowledge

the continued blessings showered on me to successfully complete my dissertation.

I deeply express my gratitude whole heartedly to my mother Smt. N. Aruna,

my Father Shri. N.Venkata Rami Reddy and my sister Miss N.padmaja for their

support and encouragement for the choices I have made, whom I owe in a substantial

measure and for which mere words cannot suffice. ‘You have given me a solid ground

for life’.

It gives me immense pleasure to acknowledge, the help rendered to me by a

host of people, to whom I owe gratitude for the successful completion of my M.Pharm

foremost amongst them are my uncle Mr. G.Venkata Rami Reddy, My aunty

Mrs.G.Tulasi for their constant encouragement, moral support and everlasting love

that have served as a source of inspiration, strength and determination at each and

every front of my life.

The research work embodied in this dissertation has been carried out at the

Department of Pharmaceutics V.L. College of Pharmacy, under the supervision of

esteemed and most respected Guide, Dr. Somashekar Shyale who gave me a chance

to work under his able guidance and for providing the necessary facilities and

pleasant working environment. I take this golden opportunity to thank and express my

sincere gratitude for his continuous encouragement, valuable suggestions, dynamic

guidance, moral support, keen interest to solve my queries, and blessings throughout

the period of this work.

vii
I hereby thank the most inspiring, charismatic and energetic Dr. S.M. Shanta

Kumar, Principal, V.L. College of Pharmacy Raichur, for his unconditional support

and encouragement.

I thank Dr. Y. Anand Kumar, Smt. K.M. Lokamatha Swamy, Dr. Ayesha

Shalam, Smt. Hafsa Mahamudi, Shri. P. Rajesh Kumar and Shri. Mallikaarjuna

Gowda, Department of Pharmaceutics, for their concern, co-operation and

encouragement throughout the duration of the course.

I thank respected Dr. K.M.K. Swamy, Dr. Srinivasalu, Shri. Zaheeruddin

and Shri. Manish Kumar Dept. of Pharmaceutical Chemistry, Dr. N. Venkat Rao

and Dr. Jeevan Mani Babu Dept. of Pharmacology, Dr. Hemant Kumar and Dr.

Pramod Kumar Dept. of Pharmacognosy, for their encouragement and moral

support.

I also acknowledge with heartfelt thanks the contributions of M/s Lupin

Pharmaceuticals Ltd. Pune and M/s Unichem Laboratories Ltd. Mumbai for

providing the polymers and excipients required for the present research.

I express my special thanks and regards to my roommates Mr. T.Rajesh, Mr.

A .Pavan kumar for filling competitive spirit in me through unending support and

encouragement.

I hereby acknowledge the cooperation and help rendered by my departmental

friends and well wishers, Mr. Syed Amezuddin Azhar, Mr. L. Prashanth Reddy,

Mr. Vivek Sood, Mr. Alluri Pavan Kumar, Mr. T. Rajesh, Mr. Suneel Kumar

viii
Batchu, Mr. Virupakshi Gouda, Mr. N. Abhilash, Mr. C S Arun Kumar, Miss.

Shashikala and Miss. Areefa .

I am also very grateful to my childhood friends and the closest friends from

the student days, especially Miss. Gayathri Lakshmi Bahumanyam,

Mr.P.V.BalaSubramanyam, Mr. Riaz basha, Mrs. Sujatha Reddy , Mr. Krishna

mohan , Miss.G.M.Sridevi, Mr. Ajay Babu, Mr. Alluri Pavan kumar. that they have

not forgotten me in spite of the long distance between us.

I am also thankful to my friends, Mr. Naga Venkat Dileep ,Mr. Shiv Patel,

Mr. Raghavendra Babu, Mr. Rajesh Pal, Mr. Sangram Deshmukh, Mr. Md. Bashir,

Mr. Rizwan Sheikh, Mr. Gajanan Dadge, Mr. Kapil Joshi, Mr. Mahesh Birajdar,

Mr. Manoj Kumar, Mr. Amol Phullari, , Mr. Fasih Ahmed, Mr. Mehboob, Mr.

Vinay, Mr. Uday, Miss Petricia, Miss Soumya, Miss Saritha, Miss Shwetha and

Miss Roopa for their co-operation and help.

I thank my seniors Miss. Anitha, Mrs. Bharthi, Mr. Srinivas, Mr. Sachin,

Mr. Srikanth, Mr. Hemanth, and others for their guidance and help.

I thank my juniors especially Miss Gayathri Lakshmi Bahumanyam Mr.

Bharath V , Mr. Viresh, Mr. Bharath D , Mr. Shyam , Mr. Uday, Mr. Kirtinidhi,

Mr. Amaregouda, Mr. Vinod, Mr. Aezaz, Miss. Bhanodaye Miss. Keerti Chandana,

Miss and for their cooperation and help.

I take this opportunity to thank all the teaching and non-teaching staff of V.L.

College of Pharmacy, Raichur, for their constant help and support. Also, I hereby

ix
express my gratitude to all the people involved directly or indirectly in the successful

completion of this dissertation work.

At last but not least I heartly thankful to my school Priyadarshini English

Medium High School, Banaganapalli ,Head mistress J.Shoba Rani and principal

J. kesarlingum sir school teachers Bhasa sir , Lakshmi teacher, Devi mam, where

motivate me to this position.

(Srikanth Reddy.N)

x
LIST OF ABBREVIATIONS USED

1. cm : Centimeter
2. conc. : Concentration
3. °C : Degree Celsius
4. DT : Disintegration Time
5. DC : Drug Content
6. ODT : Controlled Matrix Tablet
7. Fig : Figure
8. PE : Perindopril Erbumine
9. GIT : Gastro intestinal tract
10. gm : Grams
11. h : Hours
12. HCl : Hydrochloric Acid
13. ICH : International Conference on Harmonization
14. Kg : Kilogram
15. M : Molarity
16. mg : Milligram
17. min : Minutes
18. µg : Microgram
19. mL : Milliliter
20. mm : Millimeter
21. M.W : Molecular Weight
22. N : Normality
23. nm : Nanometer
24. pH : Negative logarithm of hydrogen ion concentration
25. r : Correlation Coefficient
26. RH : Relative Humidity
27. rpm : Revolutions per minute
28. SD : Standard Deviation
29. Sec : Seconds
30. U.V. : Ultra Violet
31. max : Wavelength Maxima

xi
32. WHO : World Health Organization
33. WT : Wetting Time
34. w/w : Weight by Weight

xii
ABSTRACT

Background and Objectives:

Perindopril Erbumine (PE) is an Ace inhibitor. It is effective in the treatment

of Hypertension and Blood pressure, has a half-life 0.8-1 h and oral bioavailability is

< 60 %. Perindopril erbumine shows promising pharmacokinetics and Physico-

chemical properties required for control release dosages. Therefore this drug was

selected as model drugs for this investigation.

The objectives of the present research work is to formulate and evaluate

controlled matrix tablets containing Perindopril Erbumine (PE) as a drug using

different ratios of polymers to avoid hepatic first pass metabolism and to increase

bioavailability of the drug.

Methods:

An UV Spectrophotometric method was developed to quantitate Perindopril

Erbumine either in bulk or in tablets and the method was validated for linearity,

accuracy and precision. Different matrix carriers like Xanthan gum, eudragit RL 100,

and almond gum were used to develop matrix tablets. Various rheological

characteristics of the powder bed like bulk density, tapped density, compressibility

index, and repose angle were evaluated and studied. Matrix tablets were compressed

on a 10 station pilot press using 6 mm flat faced punches (PPID, Chamunda, India)

and were all assessed for weight variation, hardness, thickness, friability, swelling

index, in vitro release of the drug by using USP XXIV dissolution testing apparatus.

The conditions were simulated and sink conditions were maintained. Further stability

xiii
studies of the Perindopril Erbumine formulations were conducted according to ICH

guidelines at 40°C/ 75 % RH for a period of 90 days.

Results:

The value of U.V. absorbance maxima (λ max) = 215.5 nm obtained during this

study corroborates with literature value 216 nm. Melting point was found to 126.5ºC

which corroborates with the literature value 1260C−1280C. Solubility of Perindopril

Erbumine was found to be 0.355 mg/mL in distilled water. The partition coefficient of

Perindopril Erbumine was found to be 0.464.

Bulk density was found to be between 0.293 gm/mL and 0.415 gm/mL for

all the formulations. Tapped density was found to be between 0.308 gm/mL and

0.498 gm/mL. The angle of repose ( ) was determined before adding the

lubricants/glidants and after adding lubricants/glidants. The repose angle before

incorporation of lubricants/glidants was found to be 21.800 to 27.120. After the

addition of lubricants/glidants the angle of repose was found to be 19.980 and

24.110. Carr’s Compressibility index was observed to be between 3.03 % and 12.6

% indicating that the powder bed is compressible.

Powder bed was compressed in 10 station rotary pilot press using 6 mm flat

faced punches. The weight of the matrix tablets was found to be fairly uniform

ranging between 100.0 ± 0.1 mg to 100.8 ± 0.2 mg for a 100 mg tablet (n=10). The

thickness of the tablets was determined using a micrometer (Mitutoyo, Japan). The

thickness of the tablets, were found to be between 3.008±0.0 mm to 3.010±0.002

(n=10). The hardness of the tablets were determined with a hardness tester (Pfizer,

xiv
India) and the hardness was found to be fairly consistent and uniform, ranging

between 3.26±0.115Kg/cm2 to 4.93±0.23Kg/cm2 (n=5).

Drug content of the all formulations were in the range of 3.806 mg to

3.94mg for matrix tablets. The friability of all the formulations were determined in a

friabilator operated for 5 min at 25 rpm, and the percent friability was found to be

0.16% to 1.1%.

The matrix tablets were found to swell gradually and the swollen tablet did

not lose its integrity for nearly 7 to 8 h. Increase in polymer content, increased

swelling index of the tablets.

In vitro release was studied by using USP XXIV dissolution apparatus, the

study was carried out in pH 7.4 and pH 6.8 buffer, at a constant temperature of 37 ±

0.5 0C stirred at 50 rpm for a period of 12 h. In vitro release data obtained was

statistically analyzed by ANOVA and a value of p<0.05 was considered to be

significant. The data was also subjected to regression analysis by least squares method

(r) to asses the linearity of the curve.

The release of Perindopril Erbumine from matrix tablets into the pH 6.8

buffer was found to be, F1 released 40.56%(r = 0.976 ), F2 released 42.53 % (r =

0.983), F3 released 45.11 % (r = 0.983 ), F4 released 44.39 % (r = 0.984), F5

released 45.18 % (r = 0.986), F6 released 44.78% (r = 0.982), F7 released 46.90 % (r

= 0.985), F8 released 47.58 % (r= 0.987), F9 48.08% (r = 0.986), respectively at the

end of 12h. Stability studies were conducted according to ICH guidelines for region

IV at 400C / 75 % RH. At regular intervals drug content was noted for 90 days and

also 90th day the matrix dosage form was subjected to dissolution.

xv
The flux from Perindopril Erbumine matrix tablets containing xanthun gum

(PE1, PE2, PE3) were found to be 0.119 mg /cm2 / h (r=0.976), 0.128 mg /cm2 / h

(r=0.983) and 0.136 mg /cm2 / h (r=0.983) respectively. The flux from Perindopril

Erbumine matrix tablets containing eudragit RL 100 (PE4, PE5, PE6) were found to

be 0.138 mg /cm2 / h (r=0.984), 0.14 mg /cm2 / h (r=0.986) and 0.137 mg /cm2 / h

(r=0.982) respectively. The flux from Perindopril Erbumine matrix tablets containing

xanthan gum (PE7, PE8, PE9) were found to be 0.147 mg /cm2 / h (r=0.985), 0.150

mg /cm2 / h (r=0.987) and 0.149 mg /cm2 / h (r=0.986) respectively.

Interpretation and conclusions:

The analytical method developed for Perindopril Erbumine was found to be

linear, accurate and precise. Melting point and λmax of the model drug determine in

this work, corroborates with previous reports, indicating the drug is pure. The

solubility of Perindopril Erbumine was found to decreasing with increase in pH. The

octanol: water partition coefficient of Perindopril Erbumine was found to be 0.464.

The drug content was found to be fairly uniform in the matrix tablets.

The thickness, diameter and weight of the tablets were found to be uniform

and consistent as indicated by their low SD values. The hardness of the compressed

tablets were determined using hardness tester (Pfizer, India) indicated that the tablets

are of adequate strength. The friability of the tablets was very low and therefore it was

understood that the tablets are of sufficient strength to withstand the stress of

transportation. Drug content of all tablet formulations was found to be uniform. All

the formulations containing matrix carriers swell considerably without losing the

shape or integrity of the tablet and also swelling increases with increase matrix

polymer content in the formulation.

xvi
In vitro release data showed that, matrix tablets containing xanthan gum

showed less release than the eudragit RL 100 and almond gum matrix tablets. To

ascertain the mechanism of release the data was plotted according to korsemeyer-

peppa equation. It was found that, the release from the tablet follows Super case II

transport owing polymer chain disentanglement and relaxation.

Stability studies data indicated no significant decrease in the drug content and

drug release . Periodic comparison of in vitro release profile during stability studies

indicated, there was a no significant difference between the profiles at p <0.05 for a

period of 90 days.

KEYWORDS: Controlled Matrix Tablets (CMTs); Perindopril Erbumine (PE);

Xanthan gum, Eudragit RL 100 and almond gum.

xvii
S.No Table of contents Page No.

1. Introduction 1 - 23
1.1. An overview of Controlled matrix drug delivery system 1

1.2. An overview of hypertension 1


1.2.1 Hypertension classification 2
1.2.2. Clinical classification of hypertension 2
1.2.3 Etiology and pathogenesis 3
1.2.4 Pathophysiology 6
1.2.5 Diagnosis 6
1.2.6. Treatment 6
1.2.7 Antihypertensive agents 7
1.2.8. Limitations of conventional dosage form 8
1.3. Rationale of controlled drug delivery system 9
1.3.1 Advantages of controlled drug delivery system 10
1.3.2 Disadvantages of controlled drug delivery system 10
1.4. Factors influencing the design of controlled release products 11
1.4.1. Physicochemical properties of drug 11
1.4.2. Route of administration 11
1.4.3. Acute/chronic therapy 12
1.4.4. Target sites 12
1.4.5. The patient 12

1.4.6. The diseased state 12


1.5. Oral controlled release system 13
1.5.1. Matrix Formulations 14

1.6. An overview on Perindopril Erbumine 19

1.7 Conventional methods 21

2. Aims and objectives of the Investigation 24-25

3. Review of literature 26-38

xviii
4. Methodology 39-74

4.1. Materials 39

4.2 Equipments 40

4.3 Profile of Perindopril Erbumine 41

4.4 Review of additives 46

4.4.1 Xanthan gum 46

4.4.2 Almond gum 49

4.4.3 Eudragit RL 100 52

4.4.4 Starch I.P 54

4.4.5 Magnesium Stearate 56

4.4.6 Talc 59

4.5 Analytical methods used for the estimation of Perindopril 62


Erbumine (PE) drug either in bulk, tablets or in dissolution
fluids

4.5.1 Method used to estimate Perindopril Erbumine 62

4.6 Methods used for characterization of the drug 65

4.6.1 Melting point determination 65

4.6.2 Solubility studies 66

4.6.3. Partition coefficient 66

4.6.4 Permeability coefficient 67

4.7. Preparation of Perindopril Erbumine Tablets 67

4.7.1. Method of preparation of granules 67

4.8 Evaluation of pre-compression characteristics of granule bed 68

4.9 Compression of tablets 69

4.10 Evaluation of post-compression characteristics of tablets 69

4.11 In vitro release study 73

xix
4.12 Stability studies 74

4.13 Statistics 74

5 Results 75-117

5.1 Characterization of Perindopril Erbumine 76

5.2 Development and evaluation of Controlled matrix tablets of 77


Perindopril Erbumine

5.3 In vitro studies 80

6. Discussion 118-122

7. Conclusion 123-126

8. Summary 127-128

9. Bibliography 129-137

xx
Table
List of tables Page No.
No.
1.1. Clinical classification of hypertension 3
1.2. In vivo performance of controlled release formulations 13
4.1. Materials used 39
4.2. Equipments used 40
4.3. Analysis of almond-tree gum 50
4.4. Spectrophotometric data for the estimation of Perindopril 62
Erbumine at 215.5 nm
4.5. Linearity studies of gemifloxacin 63
4.6. Accuracy and precision studies 64
4.7. List of the ingredients used in the formulae of different 74
Controlled Matrix tablets of Perindopril Erbumine
5.1. Various physico-chemical characteristics of Perindopril 90
Erbumine
5.2. Evaluation of rheological characteristics of CMT containing 91
Perindopril Erbumine
5.3. Evaluation of physical characteristics of CMT containing 92
Perindopril Erbumine
5.4. Swelling studies of controlled release matrix tablets of 93
Perindopril Erbumine
5.5. In vitro release of Perindopril Erbumine from controlled 94
matrix tablets containing 20%w/w Xanthan gum
5.6. In vitro release of Perindopril Erbumine from controlled 96
matrix tablets containing 25%w/w Xanthan gum
5.7. 98
In vitro release of Perindopril Erbumine from controlled
matrix tablets containing 30%w/w Xanthan gum

5.8. In vitro release of Perindopril Erbumine from controlled 100


matrix tablets containing 20%w/w Eudragit RL 100
5.9. In vitro release of Perindopril Erbumine from controlled 102
matrix tablets containing 25%w/w Eudragit RL 100
5.10. In vitro release of Perindopril Erbumine from controlled 104
matrix tablets containing 30%w/w Eudragit RL 100

xxi
5.11. In vitro release of Perindopril Erbumine from controlled 106
matrix tablets containing 20%w/w Almond gum
5.12. In vitro release of Perindopril Erbumine from controlled 108
matrix tablets containing 25%w/w Almond gum
5.13. In vitro release of Perindopril Erbumine from controlled 110
matrix tablets containing 30%w/w Almond gum

5.14. Drug content after 90 days of stability studies of controlled 112


release matrix tablets of Perindopril Erbumine (PE1, PE2,
PE3) at 400C/75% RH
5.15. Drug content after 90 days of stability studies of controlled 113
release matrix tablets of Perindopril Erbumine (PE4, PE5,
PE6) at 400C/75% RH
5.16. Drug content after 90 days of stability studies of controlled 114
release matrix tablets of Perindopril Erbumine (PE7, PE8,
PE9) at 400C/75% RH

5.17. In vitro release after 90 days of stability studies of controlled 115


release matrix tablets of Perindopril Erbumine (PE1, PE2,
PE3) at 400C/75% RH (n=3)
5.18. In vitro release after 90 days of stability studies of controlled 116
release matrix tablets of Perindopril Erbumine (PE4, PE5,
PE6) at 400C/75% RH (n=3)
5.19. In vitro release after 90 days of stability studies of controlled 117
release matrix tablets of Perindopril Erbumine (PE7, PE8,
PE9) at 400C/75% RH (n=3)

xxii
Fig. No. List of figures Page No.
1.1. Renin-angiotension activation 5
1.2. Schematically representation of a leaching-based release 16
mechanism.
4.1 Calibration curve of gemifloxacin 63
5.1. In vitro release of Perindopril Erbumine from controlled 95
matrix tablets containing 20%w/w Xanthan gum (PE 1)
5.2. In vitro release of Perindopril Erbumine from controlled 97
matrix tablets containing 25%w/w Xanthan gum(PE 2)
5.3. In vitro release of Perindopril Erbumine from controlled 99
matrix tablets containing 30%w/w Xanthan gum(PE 3)
5.4. In vitro release of Perindopril Erbumine from controlled 101
matrix tablets containing 20%w/w Eudragit RL 100 (PE 4)
5.5. In vitro release of Perindopril Erbumine from controlled 103
matrix tablets containing 25%w/w Eudragit RL 100 (PE 5)
5.6. In vitro release of Perindopril Erbumine from controlled 105
matrix tablets containing 30%w/w Eudragit RL 100 (PE 6)
5.7. In vitro release of Perindopril Erbumine from controlled 107
matrix tablets containing 20%w/w Almond gum (PE 7)
5.8. In vitro release of Perindopril Erbumine from controlled 109
matrix tablets containing 25%w/w Almond gum (PE 8)
5.9. In vitro release of Perindopril Erbumine from controlled 111
matrix tablets containing 30%w/w Almond gum (PE 9)

xxiii
1. INTRODUCTION

1.1. An overview of Controlled matrix drug delivery system:

Cardiovascular diseases are one of the life threatening diseases of mankind

and hypertension is the most common cardiovascular disease, which requires constant

monitoring. It is well known that hypertension is a major factor for congestive cardiac

failure and coronary artery disease. Control of hypertension reverses the risk of

congestive cardiac failure; where as the risk of coronary disease is not

reversed rapidly.1 It is very much essential to control the hypertension and maintain

sufficient blood circulation to heart to reduce the morbidity and make the patient to

lead a near normal life. The conventional treatment of chronic illnesses like

diabetes, hypertension ironically sometimes disturbs the normal rhythm of the life.

The ultimate aim of every therapy is to restore the normalcy of life without causing

adverse effects or the therapy must be with minimum adverse effects. Conventional

dosages cause fluctuation of drug levels in the plasma and hence, certain adverse

affects are seen. Therefore the success of a therapy depends on the continuous

administration of the drug to maintain constant drug plasma levels.2

1.2. An overview of hypertension:

Hypertension is defined as a systolic blood pressure (SBP) ≥ 140 mmHg and a

diastolic blood pressure (DBP) ≥ 90 mmHg for persons up to 60 years of age and for

subjects with diabetes mellitus or familiar hypercholesterolemia and as a SBP ≥ 160

mmHg and a DBP ≥ 90 mmHg for persons of 60 years and older without diabetes

mellitus or familiar hypercholesterolemia. Hypertension is a risk factor for myocardial

infarction, stroke, congestive heart failure, end-stage renal disease, and peripheral
Department of Pharmaceutics, V.L.C.P, Raichur. 1
vascular disease. The World Health Organization reported that suboptimal blood

pressure (SBP > 115 mmHg) is responsible for 62% of all cerebrovascular diseases

and 49% of all ischemic heart diseases. In addition, suboptimal blood pressure is the

number one cause of death throughout the Western world.

1.2.1. Hypertension classification: 4, 5

1) Primary or essential hypertension in which definite cause for the increase in

blood pressure is unknown.

2) Secondary hypertension in which increase in blood pressure is secondary to renal

endocrine or vascular lesions. Secondary hypertension comprises 5-10% cases

of hypertension.

Both essential and secondary hypertension may be benign or malignant.

Benign hypertension is moderate elevation of blood pressure and the rise is slow

over the years. About 90% patients of hypertension have benign hypertension.

Malignant hypertension is marked and rapid increase of blood pressure to

200/140mm Hg. Or more and patients have papilloedema, retinal haemorrhages and

hypertensive encephalopathy. Less than 5% of hypertensive patients

develop malignant hypertension and life expectancy after diagnosis in these patients

is less than 2 years if not treated effectively.

1.2.2. Clinical classification of hypertension:

Although hypertension is rise blood pressure above the normal clinical

values,which can be mild to malignant and therefore classified clinically as

summarized below.

Department of Pharmaceutics, V.L.C.P, Raichur. 2


Category Systolic (mm of Hg) Diastolic (mm of Hg)

Normal >130 <85

High Normal 130-139 85-89

Hypertension

Mild stage (stage 1) 140-159 90-99

Moderate (stage 2) 160-179 100-109

Severe (stage 3) 180-209 110-119

Very sever (stage 4) >210 >120

Malignant hypertension >200 >140

1.2.3. Etiology and pathogenesis:

Normal blood pressure is regulated by two haemodynamic forces and factors

which alter these factors would cause hypertension.

1) Cardiac output

2) Total peripheral vascular resistance

For essential hypertension mainly three following factors are responsible

1) Genetic factors

2) Racial factors

3) Risk factors modifying the course

1) Genetic factors: The role of familial aggregation, occurrence in twins has long

been suspected.

2) Racial and environmental factors: higher incidence of essential

hypertension is in blacks than in whites. A number of environmental factors like

Department of Pharmaceutics, V.L.C.P, Raichur. 3


salt intake, obesity, skilled occupation, higher living standards and patients

in high stress have been implicated in the development of hypertension.

3) Risk factors: The essential hypertension that begins in the middle life is

modified by a number of factors.

a) Age

b) Sex

c) Smoking

d) Obesity

e) Excess of alcoholic intake

f) Diabetes mellitus

For secondary hypertension mainly four factors are responsible:

1) Hypertension due to renal problems

2) Hypertension due to endocrine problems

3) Hypertension associated with coarctation of aorta

4) Neurogenic causes

Renal hypertension is produced by one of the following three inter-related

mechanisms:

a) Activation of renin angiotensin system

b) Sodium and water retention

c) Decreased release of vasodepressor

Department of Pharmaceutics, V.L.C.P, Raichur. 4


Activation of renin angiotensin system is shown as follows

Renin

Angiotensin-I

Angiotensin-II

Angiotensin-III

Sodium retention

Increase in volume

Blood pressure

Fig: 1.1. Renin-angiotension activation

Sodium and water retention regulates blood volume and cardiac output.

Sodium concentration in the blood is regulated by the release of aldosterone,

reduction in glomerular filtration rate and release of atriopeptin hormone from atria

of heart.

Decreased release of vasodepressor agents like prostaglandins

counters the vasopressor effect of Angiotensin – II.

Endocrinal hypertension is due to a number of endocrinal diseases

like adrenocortical hyperfunction, hyperparathyroidism.

Department of Pharmaceutics, V.L.C.P, Raichur. 5


Coarctation of aorta causes systolic hypertension due to constriction and

diastolic hypertension results from changes in the circulation.

Neurogenic hypertension is due to disease like psychogenic, increase in

intracranial pressure.

1.2.4. Pathophysiology:

The pathogenesis of essential hypertension is still unknown. Earlier it was

suggested that renal sodium retention, expanded vascular volume, increasing cardiac

output which led to increased vascular resistance. Later, it was suggested that

sympathetic nervous system plays primary role. Syndrome X relationship gives that

hypertension is related to obesity, insulin resistance, glucose intolerance and

hyperinsulinemia..

1.2.5. Diagnosis:

The diagnosis of hypertension is based on repeated, reproducible

measurements of elevated blood pressure. It serves primarily as prediction of

consequences for the patient and includes a statement about the cause of hypertension.

Since hypertension is usually asymptomatic until organ damage is imminent its

diagnosis depends mainly on measurements of blood pressure and not on symptoms

reported by patients

1.2.6. Treatment: 6

The first step in treating hypertension may be non-phamacologic which

includes sodium restriction, weight reduction in overweight patients. Pharmacologic

management includes a single drug for mild hypertension while for moderate to serve

hypertension a combination of two or more drugs.

Department of Pharmaceutics, V.L.C.P, Raichur. 6


1.2.7. Antihypertensive agents: 7

Antihypertensive agents are the drugs which lower the blood pressure in

hypertensive patients.Proteins, peptides and recombinant drugs:

a. Classification of antihypertensives:

1. Diuretics

eg. Chlorthalidone, Clopamide, Indapamide

2. β Adrenergic blockers

eg.Acebutolol, Atenolol, Metoprolol, Propranolol, Timolol

3. α Adrenergic blockers

eg. Terazosin, Prazosin, Doxazosin

4. α + β Adrenergic blockers

eg. Labetalol, Carvedilol

5. Ace inhibitors

eg. Perindopril, Captopril, Enalapril, Lisinopril, Fosinopril,

trandolapril, benazepril etc.

6. Calcium channel blockers

eg. Amlodipine, Felodipine Nifedipine, Nimodipine, Verapamil

7. Vasodilators

eg. Hydralazine, Minoxidil, Sodium nitroprusside

8. Angiotension-II receptor antagonists

eg. Candesartan, Losartan, Valsartan

9. Central sympatholytics

eg. Clonidine, Methyldopa

Department of Pharmaceutics, V.L.C.P, Raichur. 7


In the past three decades, the treatment of an acute disease or a chronic illness

has been mostly accomplished by delivery of drugs to patients using various

conventional dosage forms like tablets, capsules, ointments, liquids, and injectables,

as drug carriers. This type of drug delivery system is known to provide a prompt

release of drug. Therefore, to achieve as well as to maintain the drug concentration

within the therapeutically effective range needed for treatment, it is often necessary to

take the conventional type of drug delivery systems several times a day. This results

in a significant fluctuation of drug levels in the body.8

1.2.8. Limitations of conventional dosage form :

Poor patient compliance; increased chances of missing the dose of a drug with

short half-life for which frequent administration is necessary.

A typical peak-valley plasma concentration-time profile is obtained which

makes attainment of steady–state condition difficult.

The unavoidable fluctuations in the drug concentration may lead to under

medication or overmedication as the Css values fall or rise beyond the

therapeutic range.

The fluctuating drug levels may lead to precipitation of adverse effects like

nausea, vomiting, gastric irritation and toxicity especially of a drug with small
9
therapeutic index whenever overmedication occurs.

Continuous I.V. infusion has been recognized as a superior mode of

systemic drug delivery that can be tailored to maintain a constant and sustained

drug levels within therapeutic window for as long as required for effective

Department of Pharmaceutics, V.L.C.P, Raichur. 8


treatment. It also provides a means of direct entry into the systemic

circulation of drugs that are subjected to hepatic first-pass metabolism and/or

suspected of producing gastrointestinal incompatibility. Unfortunately, such a

mode of drug administration entails certain health hazards and therefore

necessitates continuous hospitalization during treatment and requires close medical

supervision.10

To duplicate the benefits of intravenous drug infusion without its potential

hazards, several technical advancements have been made.

There are two ways to overcome the situation:

• Development of new, better and safer drugs with long half- lives and large

therapeutic indices, and

• Effective and safer use of existing drugs through concepts and techniques of

controlled drug delivery systems.

1.3. Rationale of controlled drug delivery system:

An ideal controlled drug delivery system is the one that delivers the drug at a

pre-determined rate, locally or systemically, for a specified period of time. Controlled

release is differs from sustained release systems which simply prolong the drug

release and hence plasma drug level for a longer period of time. Thus the objective of

most products should be controlled delivery to reduce dosing frequency to an extent

that once daily dose is sufficient for therapeutic management through a

uniform plasma concentration at steady state.

Department of Pharmaceutics, V.L.C.P, Raichur. 9


1.3.1. Advantages of controlled drug deliverysystem:

Controlled release drug delivery systems have received much attention in past

two decades as they overcome the disadvantages of conventional therapy and offer

some benefits like:

• Controlled administration of a therapeutic dose at the desired delivery rate.

• Constant blood levels of the drug, reduction of side effects.

• Minimization of dosing frequency.

• Enhancement of patient compliance.

• To obtain better therapeutic efficacy and diminished toxicity.

• Increased safety margin of high potency drugs due to control of

plasma levels.

• Maximum utilization of drug enabling reduction in total amount of

dose administered.

• Reductions in health care costs through improved therapy, shorter treatment

period, less frequency of dosing and reduction in personnel time to dispense,

administers and monitor the patients.

1.3.2. Disadvantages of controlled drug delivery system :

Decreased systemic availability in comparison to immediate release

conventional dosage forms, this may be due to incomplete release, increased

first pass metabolism, increased instability, insufficient residence time for

complete release, site-specific absorption, pH dependent solubility.

Poor in vitro-in vivo correlation.

Possibility of dose dumping due to food.

Department of Pharmaceutics, V.L.C.P, Raichur. 10


Retrieval of drug is difficult in case of toxicity, poisoning or hypersensitivity

reactions.

Reduced potential for dosage adjustment of drugs normally administered in

varying strengths.
9
Higher cost of formulation.

1.4. Factors influencing the design of controlled release products:

The variables to be considered for design of controlled release products are,

1.4.1 Physicochemical properties of drug:

Physicochemical properties of drug such as solubility, partition

coefficient, protein binding, contribute in a major to the designing of controlled

release products.

1.4.2 Route of administration:

Some routes of administration exert a negative influence on drug

efficacy especially chronic administration and therefore route of application

should be considered. many physiological constraints imposed by the particular

route, that is GI motility, blood supply, first pass metabolism and sequestration

of small foreign particles by the liver and spleen influence the performance of

the controlled release systems.

1.4.3 Acute/chronic therapy:

Expected length of drug therapy to achieve cure or control of ailment

is important factor in design of controlled release product like development of one

year contraceptive implants represents a different case than does an antibiotic

Department of Pharmaceutics, V.L.C.P, Raichur. 11


treatment for acute microbial attack.

1.4.4 Target sites:

Side effects can be minimized by delivering the maximum fraction of

applied dose reaching the target site. this can be partially attained by localized

delivery or use of novel carriers.

1.4.5 The patient:

Condition of patient, ambulatory or bedridden, obese or gaunt, old or

young can effect the design of controlled design.

1.4.6 The diseased state:

Pathophysiological state of subject play important part in the design

of suitable controlled release delivery system, that is in hepatic failure oral

delivery of drug should be stopped.

The below table enlists the various drug related and biological

factors affecting the design and in vivo performance of controlled release

formulations.11

Department of Pharmaceutics, V.L.C.P, Raichur. 12


Drug related Aqueous Physiological

solubility Partition coefficient Prolonged drug absorption Variability in

Molecular size GI emptying and motility Gastrointestinal

Drug stability Protein blood flow

binding Pharmacokinetic

Biological Absorption Dose dumping first pass metabolism

Distribution Elimination variability of urinary pH effect on drug

Duration of action Margin of elimination enzyme induction/inhibition

upon multiple dosing pharmacological


safety Side effects Disease
changes in drug effect upon multiple
state
dosing sensitizing/tolerance

1.5 Oral controlled release system:

Oral controlled release dosage forms can be classified in different ways.

One way is to distinguish between single-unit dosage forms such as tablets and

capsules, and multiparticulate dosage forms such as pellets or beads. According to

Carmella et al oral sustained release technologies may be classified according to

different criteria including the type of release (e.g. continuous release, delayed

transit release, slow, prolonged, pulsed, repeat-action, etc.) the release

mechanism (e.g. diffusion, dissolution, etc.) or the type of technological

system. Combining the classification methodology used by Carmella et al and

controlled release products can be classified as follows:

Reservoir systems including enteric coated tablets, capsules, coated granules

and microcapsules.

Osmotic systems
Department of Pharmaceutics, V.L.C.P, Raichur. 13
Ion-exchange resins

Matrix systems

1.5.1 Matrix Formulations:

Matrix formulations are defined as a drug or other active ingredient

embedded in insoluble excipients in order to achieve release by a continuous

leaching of the drug from the inert matrix core.

Matrix systems can be divided into three types:

1. Monolithic matrix tablets

2. Gel forming hydrophilic matrix tablets

3. Erodable (hydrophobic) matrix tablets

Inert monolithic matrix tablets:

Probably the simplest method of obtaining controlled release of a drug from

an oral dosage form is incorporation of a drug in an inert matrix. In this inert

means noninteracting with the biological fluids. The main reason for its

popularity is that drug release from plastic matrix tablets is independent on the

state and condition of the digestive juices, which may show large inter- and intra

patient variability (pH, viscosity).

During its transit through the gastro-intestinal tract, the porous matrix

tablet does not disintegrate like conventional tablets, but remains intact and the

skeleton can be recovered in faeces. The materials used in the preparation of these

inert matrices are predominantly (insoluble) polymers and lipophilic compounds.

Department of Pharmaceutics, V.L.C.P, Raichur. 14


The first polymers to be used for the preparation of matrix tablets were (semi-)

synthetic polymers such as polyethylene, polyvinyl chloride, polymethyl

methacrylate, polystyrene, poly vinyl acetate, cellulose acetate and ethyl cellulose.

The fat compounds used included carnauba wax, hydrogenated castor oil, and

tristearin.

Major drawback of most of the inert polymeric matrix tablets were

their inherent first order drug release characteristics, their poor direct

compression characteristics and the problematic cleaning of agglomeration

equipment used for the preparation of agglomerates with the required compression

characteristics.

Mechanism of release of inert monolithic matrix tablets:

Release from inert matrix tablets occurs via leaching mechanism. Drug

particles dispersed in the polymer matrix dissolve in the penetrating gastro-

intestinal fluids and are released from the tablet by diffusion through the porous

network of already existing pores and pores that created by dissolution of the

drug particles. At drug loadings exceeding approximately 10-15 % volume, a

continuous structure connecting all drug particles exists (percolating drug

network). At considerably lower loadings, a particular fraction of the drug may

be completely surrounded by the polymer matrix (trapped fraction), which would

result in incomplete release.

Department of Pharmaceutics, V.L.C.P, Raichur. 15


Fig. 1.2. Schematically representation of a leaching-based release mechanism. 12

Solvent activated matrix tablets:

The use of solvent-activated matrix tablets as a method to obtain zero

order release i.e. constant release rates over an extended period was first

proposed by Hoffenberg. Solvent-activated drug delivery system is a collective

term comprising those systems in which the interaction between polymer and

water is responsible for achieving controlled release. The interaction with water

may include plasticization, swelling, dissolution, erosion or degradation of the

polymer. The two most important types of solvent activated matrix tablets are gel-

forming hydrophilic matrix tablets and erodable (hydrophobic) matrix tablets.

Gel-forming hydrophilic matrix tablets:

Gel-forming hydrophilic or swellable matrix systems are homogeneous

or heterogeneous systems in which the drug is dispersed in a swellable

hydrophilic polymer. These systems have been widely studied by researchers since

they offer the possibility to obtain a constant drug delivery over an extended

period of time. Drug release is a function of the polymer characteristics.

Department of Pharmaceutics, V.L.C.P, Raichur. 16


Upon swallowing gel-forming hydrophilic matrix tablets, the

hydrophilic polymer is plasticized by the aqueous gastro-intestinal due to

which undergoes macromolecular chain relaxation and volume expansion.

Consequently, upon penetration of the gastro-intestinal fluids into tablet, a sharp

front can be distinguished which separates a dry, glassy core from a hydrated and

rubbery gel layer. Release is governed by diffusion of the dissolved drug

through the swollen gel layer and generally shows a burst effect, caused by

dissolution and leaching of drug particles present at the surface prior to formation

of the release-controlling gel.

Other swellable polymers, which have been applied in swelling-

controlled oral drug delivery systems, which show solvent controlled release, are

guar gums, Xanthan gum, poly (ethylene oxide) (PEO), poly (vinyl alcohol),

ethylene-vinyl alcohol copolymers (EVA) and dextrans.

Erodable matrix tablets:

Erodable polymers such as polyanhydrides offer another interesting

material platform for z e r o -order drug r e l e a s e . Like several HPMC grades,

upon water penetration, polyanhydrides form a gel-layer, which erodes at a

specific rate. By choosing the right polymer composition the thickness of the gel-

layer may remain constant with time resulting in a constant release rate until
12
depletion of the drug.

In the last two decades, controlled-release dosage forms have made

significant progress in terms of clinical efficacy and patient compliance.

Preparation of drug- embedded matrix tablet that involves the direct

Department of Pharmaceutics, V.L.C.P, Raichur. 17


compression of a blend of drug, retardant material and additives is one of

the least complicated approaches for delivering drug in a temporal pattern into

the systemic circulation. The matrix system is commonly used for manufacturing

controlled rel ease dosage forms because it makes such manufacturing easy. A

wide array of polymers has been employed as drug retarding agents each of which

presents a different approach to the matrix concept. Polymers forming insoluble

or skeleton matrices constitute the first category of retarding materials, also

classed as plastic matrix systems.

The second class represents hydrophobic and water-insoluble materials,

which are potentially e r o d a b l e ; while t h e t h i r d g r o u p i n c l u d e s

p o l y m e r s t h o s e f o r m hydrophilic matrices.

Plastic matrix systems, due to their chemical inertness and drug

embedding ability, have been widely used for controlling the release of the

drug. Liquid penetration into the matrix is the rate-limiting step in such systems

unless channelling agents are us ed . The hydrophobic and waxy materials, on

the other hand, are potentially erodable and control the release of drug

through pore diffusion and erosion. Polymers belonging to hydrophilic matrix

systems, when exposed to an aqueous medium, does not disintegrate, but

immediately after hydration develops a highly viscous gelatinous surface barrier,

which controls the drug release from, and the liquid penetration into the centre of
13
the matrix system.

The use of hydrophilic polymers is actually the most used method in

controlling the release of drugs in the formulation of oral pharmaceutical

Department of Pharmaceutics, V.L.C.P, Raichur. 18


dosage forms. Hydroxypropyl methylcellulose has been extensively used since

the early 1960s as a rate-controlling polymer in oral extended-release dosage


14
forms.

Hydrophilic matrix systems are popular and versatile controlled release

system. Amongst polysaccharide derivatives used to produce such systems, these

are a range of cellulose ethers, e.g., Hydroxypropyl methylcellulose (HPMC) and

a diverse range of other materials, including sodium alginate, carrageenan,

chitosan, ands Xanthan gum

1.6. An overview on Perindopril Erbumine:

Numerous studies demonstrated the efficacy of perindopril in the therapy of

essential hypertension. Perindopril doses of 4 to 16 mg administered once daily are

more effective than placebo in the treatment of mild-to-moderate hypertension. Doses

greater than 8 mg offer no advantage in most patients; however, in some patients

doses of 12 or 16 mg daily provide greater therapeutic benefit. The antihypertensive

activity of perindopril is linear at doses up to 8 mg, with 2 mg doses having only

slight antihypertensive activity. Administration with hydrochlorothiazide,

indapamide, and nifedipine in patients demonstrating an inadequate response to

monotherapy has resulted in improved blood pressure control.

The long-term efficacy of perindopril was reported in the results of an open

trial evaluating 690 patients. Therapy was initiated with perindopril 4 mg once daily

and increased, if necessary, to 8 mg. If diastolic blood pressure remained greater than

90 mmHg on perindopril 8 mg, a diuretic was added, and then another

antihypertensive agent was added if necessary. After 1 year of therapy systolic and

Department of Pharmaceutics, V.L.C.P, Raichur. 19


diastolic blood pressure were reduced by 29 mmHg and 19 mmHg, respectively.

Perindopril monotherapy normalized blood pressure in 55% of the patients. Blood

pressure control was achieved in 78% of the patients. After 3 years, perindopril

monotherapy at 4 or 8 mg controlled blood pressure in 56% of the patients.

The efficacy of perindopril was demonstrated in several studies enrolling

elderly patients with essential hypertension. Perindopril doses of 2 to 8 mg daily

reduced systolic and diastolic blood pressure. In a double-blind study enrolling 34

patients with a mean age of 84 years, perindopril reduced systolic pressure 10% and

diastolic pressure 9%. Although this effect was greater than that observed in the

placebo group (5% and 4%, respectively), the difference was not significant. In a

follow-up open study enrolling 91 patients with mean age of 79.1 years, 6 months of

perindopril therapy reduced blood pressure in the 80 patients who completed the

study; systolic blood pressure was reduced by 36.7 mmHg and diastolic blood

pressure decreased by 18.2 mmHg. Blood pressure control (defined as blood pressure

< 160/95 mmHg) was achieved in 92.5% of patients (62.5% at the dosage of 2 mg per

day, 22.5% at 4 mg per day, 7.5% at 8 mg per day, and 5% at 8 mg per day plus 40

mg nifedipine). The efficacy of perindopril was evaluated in 2,927 elderly patients

(>70 years) in an open study. At initiation of therapy with perindopril 2 or 4 mg once

daily, diastolic blood pressure was between 94 and 115 mmHg. After 1 or 3 months

of therapy, the dose could be doubled or a diuretic added if the perindopril dose had

reached 8 mg in patients with diastolic blood pressure remaining above 90 mmHg.

Diastolic blood pressure was reduced to less than 90 mmHg in 69% of patients at 1

month, 86% of patients at 3 months (in patients on perindopril alone), and 94% at 6

months. At 6 months, diastolic blood pressure was lowered 28 mmHg and systolic

Department of Pharmaceutics, V.L.C.P, Raichur. 20


blood pressure 16.6 mmHg. At that time, 86% of the patients were on perindopril

alone and 14% were on perindopril plus a diuretic; perindopril doses were 2 mg in

6.7% of patients, 4 mg in 79.1%, and 8 mg in 14.2%.

The efficacy of perindopril was demonstrated in several groups of patients

with hypertension and concomitant disease including hyperlipidemia, Type II

diabetes, ischemic heart disease, cardiac arrhythmia, peripheral arterial occlusive

disease, nephropathy with proteinuria, and chronic obstructive pulmonary disease.

Efficacy was demonstrated in patients with hypertension receiving nonsteroidal anti-

inflammatory agents (Indomethacin or diclofenac). In all groups, perindopril 4 mg

once daily effectively reduced blood pressure without negatively affecting the

patient's concomitant disease state or therapy. Perindopril did not affect lipid or

apolipoprotein levels in patients with hyperlipidemia, did not affect glucose control in

diabetic patients, had an anti-ischemic and antianginal effect in those with ischemic

heart disease, and reduced urinary albumin excretion in patients with proteinuria.

Other studies have shown a lack of effect of perindopril on lipid or carbohydrate

metabolism. Several studies demonstrated the safety and efficacy of perindopril in the

treatment of hypertension in patients with diabetes or glucose intolerance. Perindopril

has not negatively affected glucose tolerance, insulin sensitivity, renal function, or

lipid profiles in diabetic patients treated long-term.

1.7. Conventional methods: 15

Conventional methods such as direct compression, dry granulation and wet

granulation methods can be adopted to produce Controlled matrix tablets. In

formulating CMTs, one of the important components is the Matrix carriers.

Department of Pharmaceutics, V.L.C.P, Raichur. 21


a) Direct Compression: 16

In this technique, tablets are compressed directly from the mixture of the drug

and excipients without modifying the physical nature of the materials. This method is

applicable for crystalline chemicals having good compressible characteristics and

flow properties such as potassium salts, chlorate, chloride, bromide etc.

The processing steps involved are:

Raw materials → weighing → screening → Mixing → Compression

b) Dry granulation technique: 16

Slugging may be used to form granules if the tablet ingredients are sensitive to

moisture and/or unable to withstand elevated temperature during drying. This method

is referred to as dry granulation. Compression granulation involves the compaction of

the components of a tablet formulation by means of a flat punch. These compact

masses, called slugs, are then milled and screened to produce granules. Specially

designed machines called roller compactors are used on a large scale.

The processing steps involved are:

Raw material → Weighing → Screening → Mixing → Slugging → Milling →

Screening → Mixing → Compression

c) Wet granulation method: 16

This is the most widely used method of tablet preparation. The active

ingredient, diluent and disintegrants are mixed or blended well. For large scale

production twin shell blenders, planetary mixers, sigma blade mixers, ribbon blade

mixers are used commonly used. Moist materials from wet milling step are placed on

Department of Pharmaceutics, V.L.C.P, Raichur. 22


large trays and placed in drying chambers with a circulating air current and

thermostable heat controller. After drying granulation, the lubricant or glidant is

added as fine powder to promote flow of granules. These granules are then

compressed to get tablets.

The processing steps involved are:

Raw material → Weighing → Screening → Wet massing → Sieving/Milling →

Drying → Screening → Mixing → Compression

Department of Pharmaceutics, V.L.C.P, Raichur. 23


2. AIMS AND OBJECTIVES OF THE INVESTIGATION

Hypertension confers an increased risk of heart attacks and strokes. Elevated

arterial pressure causes pathological changes in the vasculature and hypertrophy of

the left ventricle. As a consequence, hypertension is the principal cause of stroke,

leads to disease of the coronary arteries with myocardial infarction and sudden cardiac

death, and is a major contributor to cardiac failure, renal insuffiency, and dissecting

aneurysm of the aorta.1 Perindopril is a ACE inhibitor prodrug of Perindoprilet which

has been successfully used in the treatment of hypertension either alone or in

combination with other antihypertensive agents. When administered orally it is only

36 % available as a conventional medication.

However, some of the technologies used for the manufacture of CMTs have

disadvantages, pertaining to specialized equipment required , such as freeze-driers and

specially molded tableting machines, apart being a costly affair.

Since, Perindopril Erbumine CMT are not having Prominent significance in

the commercial market for use, an attempt was made to formulate and evaluate

Controlled matrix tablet(CMT) containing Perindopril Erbumine(PE), which can be

manufactured by commonly used production methods and equipment, with the

following objectives:

1. To carry out Preformulation studies on pure drug, pre-compressional

characterization of physical mixture of drug and excipients.

Department of Pharmaceutics, V.L.C.P, Raichur. 24


2. To evaluate tablets for various physico-chemical parameters such as hardness,

diameter, friability, weight variation, drug content, swelling index, in vitro

disintegration time, in vitro dissolution.

3. To study the influence of excipients on drug release from such tablets and to

improve the rate of release of drug from the CMT.

4. To carryout stability studies on selected fast dissolving tablets as per ICH

guidelines.

The work plan was outlined to fulfil the above objectives for different

formulation variables. Various methods were used for evaluation of the tablets to

qualify CMT requirements and the results obtained and the stability studies are all

discussed in the following chapters.

Department of Pharmaceutics, V.L.C.P, Raichur. 25


3. REVIEW OF LITERATURE

The literature from various text books, pharmacopoeias, journals, and World

Wide Web were thoroughly studied and understood. The references that were found

to be relevant to the current investigation have been cited appropriately, the following

abstracts of the previous reports would give an insight of the investigation that has

been undertaken:

Shirwaikar A.A.et.al.,17 studied Rosin, a natural resin, as an insoluble matrix

material for the release of diltiazem HCI, as a model drug. The granules prepared

were flowing with good compressibility. The tablets prepared were flat faced, which

retained their shape throughout. The method of preparation of matrix system and its

concentration were found to have a pronounced effect on the release of diltiazem HCI.

Various physical parameters of granules and the tablets were evaluated. The release

mechanisms and the release rate kinetics of the tablets were examined using different

models. The release was found to follow both the first order kinetics and Fickian

diffusion. Marked differences in the release rate of the drug from different

formulations were observed when % cumulative release was plotted against time. The

drug delivery was analyzed using the paddle method according to USP XXIII. All the

studies were done in distilled water.

Nevin Erk18 developed a new sensitive, simple, rapid and precise reversed-

phase high performance liquid chromatographic (HPLC) and two spectrophotometric

methods to resolve binary mixture of Perindopril and indapamide in the

pharmaceutical dosage forms. The first method is based on HPLC on a reversed-phase

column using a mobile phase of phosphate buffer pH 2.4 and acetonitrile (7:3 v/v)

Department of Pharmaceutics, V.L.C.P, Raichur. 26


was used. Linearity range for Perindopril and indapamide was 5.0–70.0 and 8.0–35.0

g ml−1. In the second method, the first derivative Spectrophotometry with a zero-

crossing technique of measurement is used for the simultaneous quantitative

determination of Perindopril and indapamide in binary mixtures without previous

separation step. Linear calibration graphs of first derivative values quantitative

determination of Perindopril and indapamide in binary mixtures at 225.7 and 255.4

nm for Perindopril and indapamide, respectively. The third method is based on ratio

derivative spectrophotometry, the amplitudes in the first derivative of the ratio spectra

at 226.5 and at 255.3 nm were selected to determine Perindopril and indapamide in

the binary mixture. All the proposed methods showed good linearity, precision and

reproducibility. The proposed methods were successfully applied to the

pharmaceutical dosage forms containing the above-mentioned drug combination

without any interference by the excipients.

Prameela Rani.A.etal.,19 developed A new high performance liquid

chromatographic (HPLC) method was developed and validated for the determination

of Perindopril Erbumine (PE) in pharmaceutical formulations. Optimum separation

was achieved in 10 min using C18 column (250 mm × 4.6 mm, i.d., particle size 5

mm), and elution was accomplished using a mobile phase (1ml/min). Detection was

carried out using a UV detector set at 215 nm. A linear relationship between mean

peak area and concentration of PE was observed in the range 4-20 μg/ml, with a

detection limit of 2 μg /ml and a quantization limit of 7.0 μg/ml. Intra-day and Inter-

day precision, and accuracy of the methods have been established according to the

current ICH guidelines. The developed method was successfully applied to the

determination of PE in pharmaceutical formulations. The results were statistically

Department of Pharmaceutics, V.L.C.P, Raichur. 27


compared with those of the reference method (UV method) by applying Student‟s t-

test and F-test. Accuracy, evaluated by means of the recovery method, was in the

range 99.00 - 100.5 %, with precision (RSD) 0.865%. No interference was observed

from the co formulated substances. The proposed method was successfully employed

for the determination of PE in various pharmaceutical preparations

The drug release behaviour of xanthan gum matrix tablets were studied by

Talukdar MM.et al., using three drugs having different properties, i.e., caffeine as a

soluble neutral drug, indomethacin as an insoluble acidic drug, and the sodium salt of

indomethacin as a soluble acidic drug. Swelling was ascertained by measuring the

axial and the radial expansion of matrix tablets following exposure to media of

physiological ionic strength. The mean drug dissolution time and swelling rate were

calculated from dissolution and swelling experiments, respectively, and were used as

responses for comparison under different experimental conditions. The dependence of

drug release on the swelling of the polymer matrix and on the type of the drugs added

was established. The former is mainly influenced by the ionic strength and buffer

concentrations. The latter is affected by the solubility of the drug. The mechanism of

matrix swelling follows Case I diffusion, whereas drug release from this polymer

matrix conforms to Case II diffusion.

Sarojini.S.etal.,20 Developed effect of natural almond gum as a binder in the

formulation of diclofenac sodium tablets. The study was undertaken to find out the

potential of gum from Almond gum to act as a binder and release retardant in tablet

formulations. The effect of almond gum and pvp on the release of diclofenac sodium

was studied. Seven formulations were prepared by wet granulation method containing

Microcrystalline cellulose as diluents, diclofenac sodium as model drug using


Department of Pharmaceutics, V.L.C.P, Raichur. 28
2%,4%,6%,8% and 10% w/v of almond gum solution and 2%,4% w/v of pvp gum

solution. This was carried out to find out the difference between synthetic and natural

gum and whether synthetic gum can be replaced by natural gums. The drug release

increased with almond gum when compared to synthetic gum concentration of 2%

and 4%. The values of release exponent were found to be less than 0.5. This implies

that the release mechanism is non-fickian diffusion. Tablet at 2% w/v binder

concentration showed optimum results as tablet binder. The Almond gum was found

to be useful for the preparation of uncoated tablet dosage form.

K S Lakshmi.etal.,21 A Validated HPTLC Method for Simultaneous

Determination of Losartan and Perindopril in Tablets. A Simple, fast and precise high

performance thin layer chromatographic method has been developed for the

simultaneous determination of Losartan and perindopril in tablet. Separation was

carried out on precoated TLC plates, coated with silica gel 60 F 254. The separation

was done using a mobile phase toluene: acetonitrile: formic acid (5:5:0.3 v/v/v). After

development, the chromatoplates were scanned at 215 nm. The Rf value of losartan

and perindopril was found to be 0.55 and 0.27 respectively. The results of the analysis

have been validated statistically and by recovery studies.

T. Raja sekharan.etal.,22 Formulation and Evaluation of Theophylline

Controlled Release Matrix Tablets using Xantham gum. Controlled release matrix

tablets of Theophylline were prepared with hydrophilic polymer xantham gum and

evaluated. Controlled release matrix tablets of Theophylline were prepared by wet

granulation technique by varying polymer ratios (1:1 and 1:2) and hardness (5, 6 and

7 kg/cm2). Tablets were prepared by wet granulation technique. The granules are

subjected for Preformulation studies. Compressed tablets were evaluated for hardness,
Department of Pharmaceutics, V.L.C.P, Raichur. 29
uniformity of weight, friability, drug content, thickness and diameter. All the

formulation showed compliance with pharmacopoeial standards. IR spectroscopy

revealed that there was no interaction between the drug and the polymer used in the

formulation. In vitro dissolution studies were performed using Disso 2000 (paddle

type). Among all formulations F6 showed controlled 73.18 + 1.55 % after 10 hr. The

kinetic treatment showed that the drug release followed higuchi model. F-6

formulation was subjected to stability studies at three different temperatures for

6months period. It was found to be stable. From this study it was proved that the

release of theophylline from matrix tablets was influenced by both polymer ratio and

hardness.

Bhanja Satyabrata.et al.,23 Design and in vitro Evaluation of Mucoadhesive

Buccal Tablets of Perindopril Prepared by Sintering Technique The aim of the present

study was to prepare and evaluate a mucoadhesive buccal tablet containing

antihypertensive drug i.e. Perindopril to avoid the first pass metabolism and to

improve its bioavailability with reduction in dose and also dose related side effects.

The half life of Perindopril is approximately 0.8 to 1 hrs. The tablets were prepared by

direct compression method containing polymer Polyethylene oxide and carnauba wax.

The prepared tablets were sintered at various temperatures like 600 C and 700C for 1.5

hr and 3 hr. The sintered tablets were tested for weight variation, hardness, surface

pH, drug Content Uniformity, swelling index, bio adhesive strength sand in-vitro drug

dissolution study. FTIR studies showed no evidence on interactions between drug,

polymers, and excipients. The Invitro release of Perindopril was performed under sink

conditions (Phosphate buffer PH 6.8, at 37±0.50C and 50 rpm) using USP-XXIV

dissolution apparatus. The sintering times and the sintering temperature markedly

Department of Pharmaceutics, V.L.C.P, Raichur. 30


affected the drug release properties of Perindopril buccal tablets. It is notable that the

release rate of Perindopril from buccal tablets was inversely related to the time of

sintering and the sintering temperature. This is may be due to increase in extent and

firmness of sintering which compacts the mass further, so that the drug release is

affected. The best in-vitro drug release profile was achieved with the formulation F4

A (sintered at 600c for 1.5 hr.) which contain the drug, polyethylene oxide and

carnauba wax in the ratio of 1:15:10.The surface pH, bio adhesive strength and

swelling index of formulation F4 A was found to be 6.27, 34.8 gm and 179.31 (after

12 hr). The tablets (formulation F4 A) containing 4 mg of Perindopril exhibited 8 hrs

sustained drug release (98 %) with desired therapeutic concentration. The drug release

followed diffusive mechanism with first order release kinetics. The stability studies

showed that optimized formulation was considered to be highly.

Ranjith Kumar Mamidala.et al,.24 Factors Influencing the Design and

Performance of Oral Sustained/Controlled Release Dosage Forms. Of all drug

delivery systems, oral drug delivery remains the most preferred option for

administration for various drugs. Availability of wide variety of polymers and

frequent dosing intervals helps the formulation scientist to develop

sustained/controlled release products. Oral Sustained release (S.R) / Controlled

release (C.R) products provide an advantage over conventional dosage forms by

optimizing bio-pharmaceutic, pharmacokinetic and pharmacodynamic properties of

drugs in such a way that it reduces dosing frequency to an extent that once daily dose

is sufficient for therapeutic management through uniform plasma concentration

providing maximum utility of drug with reduction in local and systemic side effects

and cure or control condition in shortest possible time by smallest quantity of drug to

Department of Pharmaceutics, V.L.C.P, Raichur. 31


assure greater patient compliance. This review describes the various factors

influencing the design and performance of sustained/controlled release products along

with suitable illustrations.

Thawatchai Phaechamud.et al,.25 Controlled Release of Propranolol HCl from

Chitosan-Lactose-Xanthan Gum Matrix Tablets. The application of low molecular

weight Materials chitosan as matrix component for controlled propranolol HCl

release prepared by direct compression was studied. The effect of the additives on

drug release from matrix tablets containing chitosan was investigated an

incorporation of xanthan gum into chitosan tablet could prolong the drug release

rather than that containing single polymer. The drug release could be modified by

addition of lactose. The drug release was gradually enhanced as the greater

amount of the lactose was added into the matrix. Most of drug dissolution profiles

could be well fitted with first order kinetic release.

Harry GB et.al.,26 characterized the two polymorphic modifications of

fosinopril sodium to their differences in melting behaviour, powder X-ray diffraction

patterns, fourier transform infrared spectra(FTIR),and solid state 31P- and 13C-NMR

specta. The polymorphs were found to be enantiotropically related based upon

melting point, heat of fusion, and solution mediated transformation data. Analysis of

solid state FTIR and 13C-NMR data indicated that the environment of the acetal side

chain of the fosinopril sodium differd in two polymorphs, and that there might be cis-

trans isomerization about the C6-N peptide bond. These conformational

differences are postulated as the origin of the observed polymorphism.

Department of Pharmaceutics, V.L.C.P, Raichur. 32


Basak SC et.al., 27 prepared propranolol hydrochloride matrix tablets with

hydroxypropyl methylcellulose polymer to control the release of drug with a view to

develop twice daily sustained release dosage for. The resulting matrix tablets prepared

with hydroxypropyl methylcellulose K4M fulfilled all the official requirements of

tablet dosage forms. The in vitro drug release was measured in aqueous solutions for a

total period of 12 h using 1.2 pH buffer for first 1 h and pH 7.5 buffer for the rest of

period. The drug release was within the limits of predetermined set vis-a-vis USP

requirements.

Sunil Jambhekar et.al.,28 made an effort to improve the bioavailability of the

insoluble drug indomethacin, three complexes were prepared with Indomethacin and

the soluble complexing agents β-hydroxyethyl and hydroxypropyl β-cyclodextrin. The

indomethacin content was similar among the complexes (P ≤ 0.05). To confirm

complex formation, each complex was characterized by ultraviolet, infrared, nuclear-

magnetic resonance, powder X-ray diffraction, and differential-scanning calorimetry

techniques. Powder diffraction studies show the β-cyclodextrin complex was

polycrystalline, and the hydroxyethyl and hydroxypropyl β-cyclodextrin complexes

were amorphous. Phase-solubility analysis confirmed the formation of complexes and

suggested the three complexes were bound similarly. Solubility studies show

complexation increased indomethacin solubility, and the hydroxyethyl and

hydroxypropyl- β-cyclodextrin complexes were more soluble than the β-cyclodextrin

complex in 0.1N hydrochloric acid and distilled water. Dosage forms were prepared

by encapsulating the complexes without the addition of excipients. Dissolution studies

show the encapsulated β- and hydroxyethyl- β-cyclodextrin complexes had superior

dissolution when compared to the hydroxypropyl- β-cyclodextrin and Indocin® (50

Department of Pharmaceutics, V.L.C.P, Raichur. 33


mg) capsules. Bioavailability studies were performed by administering the

indomethacin complex or Indocin capsules to male-albino, New Zealand rabbits.

Indomethacin plasma-time concentration data fit best to a compartment-independent

model for all capsule formulations. Bioavailability comparisons by ANOVA show no

significant difference (P ≤ 0.10) in the peak-plasma time and peak concentration

among the capsule formulations. The area-under-the-curve for the β-cyclodextrin

complex capsules was found to be significantly higher (P ≤ 0.10) than all other

capsule formulations. In conclusion, the bioavailabilty of indomethacinwas improved

by complexation with only β-cyclodextrin No correlations were found among the

bioavailability, solubility, and dissolution results.

Medenica.M et.al.,29 Evaluation of impurities level of perindopril tert-

butylamine in tablets Perindopril tert-butylamine is a new member of angiotensin-

converting enzyme inhibitors group used in the treatment of hypertension and heart

failure. In this paper, the evaluation of reversed-phase high-performance liquid

chromatographic method (RP-HPLC) for the determination of impurities level of

perindopril tert-butylamine in tablets was done. The chromatograms were recorded

using a Hewlett Packard 1100 chromatographic system with DAD detector.

Separations were performed on a YMC-Pack C8 column (250mm×4.6 mm; 5 _m

particle size) at 50◦C column temperature. Mobile phase was a mixture of

acetonitrile–potassium phosphate buffer (0.05 M) (37:63, v/v) (pH 2.5). pH of the

mobile phase was adjusted with orthophosphoric acid. Mixture of acetonitrile–water

(40:60, v/v) was used as a solvent. Injection volume was 50 _l, flow rate 1.7 ml

min−1 and UV-detection was performed at 215 nm. The developed method subjected

to method validation and parameters in terms of selectivity, linearity, precision,

Department of Pharmaceutics, V.L.C.P, Raichur. 34


accuracy, limit of detection, limit of quantitation and robustness were defined. The

validated method is suitable for the simultaneous determination of perindopril tert-

butylamine as well as its impurities in pharmaceuticals.

Hisham E. Abdellatef et.al.,30 Two sensitive, spectrophotometric and atomic

absorption spectrometric procedures are developed for the determinationof ramipril

and perindopril. Both methods are based on the formation of a ternary complex,

extractable withchloroform, between copper(II), eosin and the two cited drugs.

Spectrophotometrically under the optimum condition, the ternary complexes showed

an absorption maximum at 535 nm, with apparent molar absorptivities of 6.55

and4.00_103 mol_1cm_1 and Sandell‟s sensitivities of 5.80_10_2 and 1.04_10_1mg

cm_2 for perindopril andramipril, respectively. The solution of ternary complex

obeyed Beer‟s law in concentration ranges 10–60 and 20–100 mg ml_1 for

perindopril and ramipril, respectively. The proposed method was applied to the

determination of the two cited drugs in pharmaceutical tablets. The atomic absorption

spectrometric method, directly through the quantitative determination of copper

content of the organic extract of the complex, was also investigated for the purpose of

enhancing the sensitivity of the determination. The spectrophotometric and atomic

absorption spectrometric procedures hold their accuracy and precision well when

applied to the determination of ramipril and perindopril dosage forms.

Hisham E. Abdellatef et.al.,31 Simple, rapid, accurate and sensitive

spectrophotometric methods are described for the determination of perindopril. The

methods are based on the reaction of this drug as n-electron donor with 2,3-dichloro-

5,6-dicyano-p-benzoquinone(DDQ)-7,7,8,8-tetracyanoquinodimethane (TCNQ),

tetracyanoethylene (TCNE), chloranil (CL) and p-chloranilic acid (p-CA) as p-

Department of Pharmaceutics, V.L.C.P, Raichur. 35


acceptors to give highly coloured complex species. The coloured products are

measured spectrophotometrically at 588, 843, 419, 550 and 520 nm for DDQ, TCNQ,

TCNE, CL and p-CA, respectively, optimization of different experimental conditions

is described. Beer‟s law is obeyed in the range of 20–200 mg ml_1 and colours were

produced in non-aqueous media and were stable for at least 1 h. Application of the

suggested methods to perindopril tablets are presented.

Kenneth I. Ozoemenaa.et al.,32 Construction of enantioselective,

potentiometric membrane electrodes based on carbon-paste impregnated with and

cyclodextrin as chiral selectors for the assay of S-perindopril is described. Response

characteristics showed that the proposed electrodes could be reliably applied in the

assay of S-perindopril raw material and its pharmaceutical formulation. The best

enantioselectivity and time-stability were exhibited by and cyclodextrin based

electrodes.

33
Yeole PG et.al., made an attempt to increase therapeutic efficacy, reduce

frequency of administration, and improve patient compliance, by developing

sustained release matrix tablets of diclofenac sodium. Sustained release matrix tablets

of diclofenac sodium, were developed by using different drug: polymer ratios, such as

F1 (1:0.12), F2 (1:0.16), F3 (1:0.20), F4 (1:0.24) and F5 (1:0.28). Xanthan gum was

used as matrix former, and microcrystalline cellulose as diluent. All the lubricated

formulations were compressed using 8 mm flat faced punches. Compressed tablets

were evaluated for uniformity of weight, content of active ingredient, friability,

hardness, thickness, in vitro dissolution using basket method, and swelling index. All

the formulations showed compliance with pharmacopoeial standards. Among

different formulations, F1 showed sustained release of drug for 12 hours with 89.67%
Department of Pharmaceutics, V.L.C.P, Raichur. 36
release. The effect of other parameters like addition of release modifier (PEG 6000),

gum concentration, pH of dissolution medium, rotation speed and dissolution by

paddle method, were also studied. Selected formulation (F1) was subjected to stability

studies for three months at 0-4°, room temperature (28°), and 45° with RH 75±5%,

and showed stability with respect to release pattern. The kinetic treatment showed that

the release of drug follows zero order kinetic (R2 = 0.9758). Korsmeyer and Peppas

equation gave value of n = 0.9409 which was close to one, indicating that the drug

was released by zero order kinetic. Thus, Xanthan gum can be used as an effective

matrix former, to extend the release of diclofenac sodium.

34
Reynolds D, et al., investigated polymer erosion of matrices of similarly

substituted hydroxy propyl methylcellulose (HPMC) polymers was examined, and

drug release in terms of diffusion and erosion contributions was characterized,

focusing on matrices containing either polymer alone or a drug content of 25% level

with no added excipient A novel approach was utilized to separate diffusion and

erosional contributions to drug release. Diffusion drug release was determined by

fitting release data versus (time) 0.45, and the drug release due to erosion was

quantified by subtracting the percent predicted for diffusion drug release from the

total drug release at each specific time point. Drug release resulting from polymer

erosion was linear versus time and was found to be a function of the number average

Molecular weight of the polymer. In contrast, diffusion release rates were comparable

for all HPMC grades studied and, thus, were independent of number average

molecular weight of the polymers studied. Under stirring conditions of 10 - 100 rpm

as well as static condition, the detachment of individual polymer chains at the matrix

surface occurred at a faster rate relative to diffusion away from the matrix surface.

Department of Pharmaceutics, V.L.C.P, Raichur. 37


The erosion study indicated that polymer diffusion of the HPMC polymer chains

through the aqueous diffusion layer was the rate-limiting step for polymer erosion. In

general, polymer erosion was found to be inversely related to the polymer number

average molecular weight. A scaling law was used to relate polymer erosion rate with

the respective polymer number average molecular weight. Similar relationships were

obtained for matrices with and without drug at a stirring rate of 100 rpm.

35
Lutfi Genc et al., prepared controlled release dosage forms of

dimenhydrinate with different polymers as MC, HEC, Carbopol 934,Eudragit RLPM

and Eudragit NE 30 D at different concentrations 2.5–10%. Direct compression DC

and wet granulation WG techniques were used to prepare the tablets. Magnesium

stearate was the lubricant while starch gel was the binder. When the branched

polymer is in contact with synthetic gastric liquid, this polymer turns into a gel,

because of the liquid transfer into the polymer. On the other hand, a release of the

drug is observed, which does not follow a classical kinetic equation, as the kinetics is

partially controlled by diffusion. This study shows that the diffusion release

mechanism in a matrix system comprising an insoluble hydrophobic and a hydrophilic

gel-forming part depends greatly on the wettability of the added drug. Furthermore,

with wettable and water soluble drug, the matrix swells and release is mainly achieved

by diffusion due to dissolution of the gel formed

Department of Pharmaceutics, V.L.C.P, Raichur. 38


4. METHODOLOGY

4.1. Materials

The materials used for the formulation and evaluations are listed below:

S. No. Materials Source

Complimentary sample from Lupin drugs


1. Perindopril Erbumine
Ltd, Pune

2. Xanthan gum S.d. Fine chemicals limited, Mumbai

3. Almond gum INR Chemicals, Mumbai

4. Eudragit RL100 S.d. Fine chemicals limited, Mumbai

5. Starch I.P S.d. Fine chemicals limited, Mumbai

6. Talc S.d. Fine chemicals limited, Mumbai

Magnesium Stearate S.d. Fine chemicals limited, Mumbai


7.

Department of Pharmaceutics, V.L.C.P, Raichur. 39


4. 2. Equipments

S. No Name of the Equipment Supplier / Manufacturer

Digital balance
1. Afcoset electronic balance, Mumbai.

2. Hot air oven Spencers, Delhi.

10 station PP1D, Chamunda pharma,


3. Tablet compression machine
Ahmedabad.

Tablet Dissolution tester USP


4. DBK Instruments, Mumbai.
XXII

5. Disintegration apparatus Campbell electronics, Mumbai.

6. Friability test apparatus Konark instruments.Ambala,Haryana.

7. pH meter Systronics, Mumbai.

UV-visible
spectrophotometer
8. Shimadzu Corporation, Japan.
(UV-1700)

9. Hardness tester Pfizer hardness tester, India.

10. Blender Konark instruments.Ambala,Haryana.

11. Bulk density apparatus Konark instruments.Ambala,Haryana.

12. Rotary Shaker Konark instruments.Ambala,Haryana.

13. Vernier Calipers Mitutoyo, Japan

Department of Pharmaceutics, V.L.C.P, Raichur. 40


4.3. Profile of Perindopril Erbumine: 38-46

Perindopril Erbumine is a dipeptide monoacid monoester with a

perhydroindole group and no sulphydryl radical; chemical name, tert-butylammonium

(2S, 3aS, 7aS)-1-(N-[(S)-1-ethoxycarbonyl butyl]-L-alanyl) perhydroindole-2-

carboxylate and used for the treatment of patients with hypertension and symptomatic

heart failure.

Physical and chemical properties:

Empirical Formula : C19H32N2O5 , C4H11N

Molecular weight : 368.46 g/mol

Melting point : 126 -128 C

Category : Antihypertensive

Description : White crystalline powder, slightly hygroscopic

odourless, bitter in taste.

Solubility : The Perindopril Erbumine is soluble in water,

methanol, ethanol, acetic acid and ethyl acetate, very

slightly soluble in ether, chloroform and benzene.

Chemical Nature : Perindopril Erbumine dipeptide monoacid monoester

with a perhydroindole group and no sulphydryl

radical.

Department of Pharmaceutics, V.L.C.P, Raichur. 41


Structure:

Perindopril Erbumine

Chemical name: Tert-butylammonium (2S, 3aS, 7aS)-1-(N-[(S)-1-ethoxycarbonyl

butyl]-L-alanyl) perhydroindole-2-carboxylate

Pharmacokinetics:

Absorption : well absorbed after oral administration.

Route of administration : Oral

Protein Binding : 10 - 20%

Bioavailability : 65% - 75 %

Metabolism : Hepatic.

Half life : 1.2 h

Excretion : Renal.

Department of Pharmaceutics, V.L.C.P, Raichur. 42


Volume of Distribution : Approx. 140-160L/kg ;

Clearance : 219 – 362 mL/min.

Dissociation Constant (Pka): 2.6

Elimination Half life : 0.8-1 hr

Mechanism of Action:

Perindopril erbumine inhibits ACE in human subjects and animals, while the

principle mechanism of perindopril is blood pressure reduction is believed to be

through the renin-angiotension- aldoserone system.

There are two isoforms of ACE the somatic isoform, which exists as a

glycoprotein comprised of a single polypeptide chain of 1277 and the testicular

isoform, which has a lower molecular mass and is thought to play a role in sperm

maturation and binding of sperm to the oviduct epithelium. Somatic ACE has two

functionally active domains, N and C, which arise from tandem gene duplication.

Although the two domains have high sequence similarity, they play distinct

physiological roles. The C-domain is predominantly involved in blood pressure

regulation while the N-domain plays a role in haematopoietic stem cell differentiation

and proliferation. ACE inhibitors bind to and inhibit the activity of both domains, but

have much greater affinity for and inhibitory activity against the C-domain.

Perindoprilet, the active metabolite of perindopril, competes with ATI for binding to

ACE and inhibits and enzymatic proteolysis of ATI to ATII. Decreasing ATII levels

in the body decreases blood pressure by inhibiting the pressor effects of ATII as

described in the Pharmacology section above. Perindopril also causes an increase in

Department of Pharmaceutics, V.L.C.P, Raichur. 43


plasma renin activity likely due to a loss of feedback inhibition mediated by ATII on

the release of renin and/or stimulation of reflex mechanisms via baroreceptors.

Adverse effects:

Upper respiratory infection, asthenia, rhinitis, diarrhoea, oedema, pharyngitis,

urinary tract infection, abdominal pain, sleep disorder, chest disorder, chest pain,

tinnitus, flatulence, malaise, cold/hot sensation, angioedema, gastro enteritis,

bronchitis, rhino rhea, vaginitis, ventricular extra systole, myocardial infraction

vasodilation, gout, pruvitus, conjunctivitis, arthralgia.

Contraindications:

Diuretics reduce the bioavailability of perindopril erbumine, Pregnancy:

contraindicated.

Interactions:

Perindopril Erbumine is known to interact with other drugs like Fluorouracil,

Lithium, Phenobarbitone, Phenytoin (Na), Warfarin (Na), Digoxin, Azathioprine,

Aspirin. These interactions are sometimes beneficial and sometimes may pose threats

to life.

Indications:

The drug is effective in the treatment of Hypotension, Anaphylactic reactions

during desensitization, membrane exposure, Intestinal angioedema, Neutropenia.

Dosage:

For Adult dose; 4-8 mg as a single dose for 1 day.

Department of Pharmaceutics, V.L.C.P, Raichur. 44


Commercial dosage: Aceon 4mg (Solvey pharma.)

Conversyl 4mg ( Aurobindo Pharma)

Warnings / Precautions:

Alcoholic drinks and alcohol containing medicines should be avoided during

Perindopril Erbumine treatment. Do not administer to subjects with a history of blood

dyscrasia.

Specification : 4mg

Storage : Keep tightly closed and Store in a dry place,

Protect from direct light.

Package : tablets × 1 blister in PVC/aluminium blister packaging.

Shelf life : 24 months.

Department of Pharmaceutics, V.L.C.P, Raichur. 45


4.4. Review of additives:

4.4.1. Xanthan Gum:47-49

Chemical name : Xanthan gum

Empirical formula : (C35H49O29)n

Molecular weight : (933) n Daltons

Chemical Structure :

H
H OH H H OH H H
H
OH 4
O OH
O HO
HO 1 O HO OH
O H
H 1 O
HO 1 HO O 3
1 OH
OH H OH H H H HO
H H H n
H OH

2 H
H O
O H
O O O
H 6 OH H 1
OH
O O 6
O O
4
O 4 H O OH
1 O OH H H
CH 3 H
HO HO
H H O
H H CH 3

Xanthan is an anionic bacterial polysaccharide composed of a U-(1)-D-

Gle(1)-beta-D-GLC (cellulosic) backbone with a trisaccharide side chain linked to

C3 of every second glucose residue. The side chain is U-D-Man-(1)-U-D-Gle A-

1(/2)-6-O-acetyl-alpha-D-Man-(1/beta-D-Glca-((12)-alpha-D with

approximately 60% of the terminal mannose units being pyruvylated and 90% of the

proximal mannose units substituted at C6 with O-acetyl groups. It has side chains of 2

mannose and 1 gluconic glucoronic acid group.

Department of Pharmaceutics, V.L.C.P, Raichur. 46


Xanthan gum is an exocellular polysaccharide reduced by fermentation of the

bacteria Xanthomonas campestris.

Description:

Xanthan gum is off white granular powder. In cold water, it is dispersed to

form pseudo plastic mixtures. It is highly water soluble.

Melting point: 270 °C

Viscosity (dynamic): 1200–1600 mPas (1200-1600 cP) for 1% w/v aqueous solution

at 25°C.

Salt solutions: Compatible and stable in solutions with high salt concentrations.

Others:

It has unique enzyme resistance. Upon heating it may increase or maintain

viscosity. It is affected by shear but recovers hence, is thixotropic gelling agent. It

stabilizes solid, liquid, and gaseous dispersions, viscosity dispersions are highly

pseudo plastic, Xanthan exhibits pseudo plasticity on the basis of its helical structure.

Stability and Storage:

Xanthan gum is a stable material Aqueous solutions are stable over a wide pH

range (pH 3–12), although they demonstrate maximum stability at pH 4–10 and

temperatures of 10–60°C. Xanthan gum solutions of less than 1% w/v concentration

may be adversely affected by higher than ambient temperatures: for example,

viscosity is reduced. Solutions are also stable in the presence of enzymes, salts, acids,

and bases.

Department of Pharmaceutics, V.L.C.P, Raichur. 47


Applications in pharmaceutical formulation or technology:

Gum provides visibly clear solutions even at higher concentrations; it exhibits

less gummy mouth-feel than gums with more Newtonian characteristics. Xanthan

gum is commercially available in both clarified and non clarified forms. Clarified

Xanthan gives visibly clear solutions even at high concentrations, while unclarified

Xanthan gums are opaque. It acts as an emulsion stabilizer, holds water, enhances

freeze-thaw stability, inhibits starch retro gradation improves shelf-life and serves to

bring about stabilization dispersions, suspension, and emulsion, and thickeners.

Although primarily used as a suspending agent, Xanthan gum has also been used to

prepare sustained-release matrix tablets. Controlled-release tablets of diltiazem

hydrochloride prepared using Xanthan gum have been reported to sustain the drug

release in a predictable manner and the drug release profiles of these tablets were not

affected by pH and agitation rate.

Department of Pharmaceutics, V.L.C.P, Raichur. 48


4.4.2 Almond Gum :50-53

Synonym : Peach gum, Cedo gum. A kind of amylose natural

gum secreted from peach trees.

Structure:
H
O
H
O
O N
H
O O
O O O
H

H H
O O
O
H

Chemical name:

2-phenyl-2-[(2R,3R,4S,5S,6R)-3,4,5-trihydroxy-6-[[(2R,3R,4S,5S,6R)-3,4,5-

trihydoxy-6-(hydroxymethyl)oxan-2-yl]oxymethyl]oxan-2-yl]oxyacetonitrile.

Molecular Formula : C20H27NO11, 3H2O

Molecular Weight : 511.27 Daltons

pH : 6-7 (15 % w/v solution)

Department of Pharmaceutics, V.L.C.P, Raichur. 49


Chemistry:

Amygdalin, C20H27NO11, 3H2O, is the glucoside of d-phenylglycollic acid, and

is obtained from bitter almonds, or the seeds of other rosaceous plants. It is neutral in

reaction and laevorotatory ([α] D = -35.5°).By the action of emulsin, it is decomposed

into dextrose, benzaldehyde, and hydrocyanic acid this decomposition also occurs

under the influence of dilute acids.

Composition of Almond tree Gum :

The following table embodies the results obtained in the analysis of almond-

tree gum.

Table.4.3. Analysis of almond-tree gum

Almond - tree
Gum Form
(hard) (elastic)

Soluble in Water calculated 21.06 8.90

on Dry Substance %

Insoluble in water % 78.94 91.10

Moisture % 15.76 25

Ash % 2 .34 ----

Galactans as Galactose % 23.70 ----

Pentosans as Arabinose % 54.60 ----

Total Sugars 85 91

Department of Pharmaceutics, V.L.C.P, Raichur. 50


Melting point : It melts at about 200°C, after darkening in colour, and sets to

glassy mass, which melts at 125° C to 130°C.

Description : It occurs as a brownish, odorless, granular powder.

Solubility : Soluble in water or alcohol; insoluble in ether.

Applications:

Thickening agent and Stabilizer in foodstuff and cosmetics, auxiliary in ink

and pigment, Excipient, Gooey, etc.

Medicinal Uses:

As well as being a tasty addition to the diet, almonds are also beneficial to the

overall health of the body, being used especially in the treatment of kidney stones,

gallstones and constipation. Externally, the oil is applied to dry skins and is also often

used as carrier oil in aromatherapy. The seed is demulcent, emollient, laxative,

nutritive and pectoral. When used medicinally, the fixed oil from the seed is normally

employed. The seed contains 'laetrile', a substance that has also been called vitamin

B17.This has been claimed to have a positive effect in the treatment of cancer, but

there does not at present seem to be much evidence to support this. The pure

substance is almost harmless, but on hydrolysis it yields hydrocyanic acid, a very

rapidly acting poison - it should thus be treated with caution. In small amounts this

exceedingly poisonous compound stimulates respiration, improves digestion and

gives a sense of well-being. The leaves are used in the treatment of diabetes. The

plant contains the antitumor compound taxifolin.

Department of Pharmaceutics, V.L.C.P, Raichur. 51


4.4.3.Eudragit RL 10054-55

Chemical name: Polymethacrylate

Chemical Structure:

CH3 CH3 CH3 CH3


CH2 CH2 CH2 CH2
O O O O
O O O O
H CH3 H CH3
n

Polymethacrylates are synthetic cationic and anionic polymers of dimethylaminoethyl

methacrylates, methacrylic acid and methacrylic acid esters.

Molecular weight: 1, 50,000.

Form: Granular form.

Colour: colourless.

Odour: Faint like amine odour.

Density: Relative density 0.816 - 0.836 g /c3.

Functional category: Film former, tablet binder, tablet diluent.

Solubility: Eudragit RL 100 is soluble in ethanol, methanol, isopropyl alcohol,

dichloro methane, acetone and ethyl acetate.

Viscosity: Eudragit RL 100 is having viscosity range of 15 mPas.

Department of Pharmaceutics, V.L.C.P, Raichur. 52


Stability and storage: Dry powder polymer forms are stable at temperature less than

30 oC. Above this temperature, powders tend to form clumps, although this does not

affect the quality of the substance and the clumps can readily be broken up. Dry

powders are stable for atleast 3 years if stored in tightly closed containers at less than

30 oC.

Incompatibilities: Interaction between polymethacrylates and some drugs can occur,

although solid polymethacrylates and organic solutions are generally more compatible

than aqueous dispersions.

Department of Pharmaceutics, V.L.C.P, Raichur. 53


4.4.4. Starch I.P :56

Chemical name : Starch.

Empirical formula : (C6H10O5)n where n = 300–1000.

Structural formula : Starch consists of amylose and amylopectin, two

polysaccharides based on α-glucose.

Structure :

Starch is the mixture of two polymers, amylase,- a linear (1→4) α-D-Glucan

and amylopectin, a branched D-Glucan with mostly α-D (1→4) and approximately

4% α-D (1→6) linkages.

Department of Pharmaceutics, V.L.C.P, Raichur. 54


Solubility:

It is practically insoluble in cold ethanol (95%) and in cold water. Starch

swells instantaneously in water by about 5–10% at37°C. Polyvalent cations produce

more swelling than monovalent ions, but pH has little effect.

Description:

Starch is a white-colour, odourless fine powder, comprising very small

spherical or ovoid granules, whose size and shapes are characteristic for each

botanical variety.

Functional category: Binder, Diluent, Disintegrant.

Applications in pharmaceutical formulation or technology:

Starch is one of the most commonly used tablet disintegrant at concentrations

of 3–15 % w/w. It is utilized as a binder, diluent. Unmodified starch does not

compress well and tends to increase tablet friability and capping if used in high

concentrations. Starch is also used in topical preparation and is used as a protective

covering in ointment formulations applied to the skin. Starch mucilage has also been

applied to the skin as an emollient, has formed the base of some enemas, and has

been used in the treatment of iodine poisoning. Starch has been investigated as an

excipient in novel drug delivery systems for nasal, oral, periodontal, and other site-

specific delivery systems.

Department of Pharmaceutics, V.L.C.P, Raichur. 55


4.4.5. Magnesium Stearate: 57,58

Synonym: Magnesium octadecanoate, octadecanoic acid, stearic acid, magnesium

salt.

Chemical Name: Octadecanoic acid magnesium salt

Empirical Formula: C36H70MgO4

Structural Formula:

[CH3 (CH2)16COO] 2Mg

-
H3C O 2+
Mg
-
H3C O

Molecular Weight: 591.34

Melting Point: 117-150 °C (commercial samples); 126-130 °C (high purity)

Solubility: Practically insoluble in ethanol, ethanol (95%), ether and water; slightly

soluble in warm benzene and warm ethanol (95%).

Description: Magnesium stearate is a very fine, light, white, precipitated or milled,

impalpable powder of low bulk density, having a faint odour of stearic acid and a

characteristic taste. The powder is greasy to the touch and readily adheres to the skin.

Department of Pharmaceutics, V.L.C.P, Raichur. 56


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

Flow ability : poorly flowing, cohesive powder.

Melting range : 117–1500C (commercial samples)

Solubility : Practically insoluble in ethanol, ether and water;

slightly soluble in warm benzene and warm ethanol.

Specific surface area : 1.6–14.8 m2/g

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.

Applications in Pharmaceutical Formulation or Technology: Magnesium stearate

is widely used in cosmetics, foods, and Pharmaceutical formulations. It is primarily

Department of Pharmaceutics, V.L.C.P, Raichur. 57


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.

Functional Category: Tablet and capsule lubricant.

Stability and Storage Conditions: Magnesium stearate is stable and should be stored

in a well closed container in a cool, dry place.

Safety: Magnesium stearate is widely used as a pharmaceutical excipient and is

generally regarded as being non toxic following oral administration. However, oral

consumption of large quantities may produce a laxative effect or mucosal irritation.

Department of Pharmaceutics, V.L.C.P, Raichur. 58


4.4.8. Talc: 59

Synonyms: Hydrous magnesium calcium silicate, hydrous magnesium silicate,

magnesium hydrogen metasilicate, powdered talc, Purtalc, soap stone, steatite.

Chemical Name: Talc

Structural Formula: Mg6 (Si2O5)4(OH) 4

Empirical Formula: Talc is a purified, hydrated, magnesium silicate, approximating

to the formula Mg6 (Si2O5)4(OH)4. It may contain small, variable amounts of

aluminium silicate and iron.

Molecular Weight: 379.27 gm

Solubility: Talc is practically insoluble in dilute acids and alkalis, organic solvents,

and water.

Description: Talc is a very fine, white to greyish-white, odourless, impalpable,

unctuous, crystalline powder. It adheres readily to the skin and is soft to the touch and

free from grittiness.

Typical Properties:

Moisture content : Talc absorbs insignificant amounts of water at

2580C and relative humidity‟s up to about 90%.

Particle size distribution : varies with the source and grade of material.

Department of Pharmaceutics, V.L.C.P, Raichur. 59


Refractive index : nD20 = 1.54–1.59

Solubility : Practically insoluble in dilute acids and alkalis,

organic solvents, and water.

Specific gravity : 2.7–2.8

Specific surface area : 2.41–2.42m2/g

Storage Conditions : Talc should be stored in a well-closed container

in a cool, dry place.

Incompatibilities : Incompatible with quaternary ammonium

compounds.

Applications in Pharmaceutical Formulation or Technology: Talc is used in oral

solid dosage formulations as a glidant and lubricant (1-10%) and diluent (5-30%), as a

dissolution retardant in the development of controlled-release products, as an

adsorbent, as a dusting powder in topical preparations (90-99%).

Functional Category : It is used as an anti caking agent, glidant, tablet and capsule

diluent, tablet and capsule lubricant.

Storage Conditions: Talc should be stored in a well-closed container in a cool, dry

place.

Safety: Talc is not absorbed systemically following oral ingestion and is therefore

regarded as an essentially non toxic material. However, intranasal or intravenous

Department of Pharmaceutics, V.L.C.P, Raichur. 60


abuse of products containing talc can cause granulomas in body tissues, particularly

the lungs.

4.5. Analytical methods used for the estimation of Perindopril Erbumine (PE)

drug either in bulk, tablets or in dissolution fluids: 36,37

4.5.1. Method used to estimate Perindopril Erbumine:36

a) Preparation of Stock Solution:

100 mg of pure Perindopril erbumine was dissolved in 100 mL of distilled

water to obtain 1 mg /mL solution. This solution was further diluted with distilled

water till 10 μg/mL solution was obtained.

b) Analytical Methods:

The UV Spectrophotometric analytical method was developed for Perindopril

erbumine drug using a UV double beam Spectrophotometer (UV Pharmaspec 1700,

Shimadzu, Japan).

1) Determination of λ max:

10 μg/mL solution of Perindopril erbumine prepared as above was scanned for

absorbance in UV double beam spectrophotometer (UV Pharmaspec 1700, Shimadzu,

Japan) between range 200 to 400 nm, against distilled water as blank. It was found

that the solution showed absorbance maxima of 215.5 nm.

2) Estimation of Drug:

In present work, Spectrophotometric method was adopted using double beam

UV spectrophotometer (UV Pharmaspec 1700, Shimadzu, Japan).

Department of Pharmaceutics, V.L.C.P, Raichur. 61


3) Calibration curve of Perindopril erbumine:

Procedure:

Different aliquots were taken from stock solution in a series of 10 mL

volumetric flask and diluted with distilled water to prepare a series of concentration in

the range of 2-10 μg/mL. The solutions were scanned on spectrophotometer in the UV

range (200-400 nm) and the absorbance was measured at 215.5 nm using distilled

water as a blank.

The absorbance so obtained was tabulated in table 4.3. Calibration curve was

constructed and is shown in figure 4.1.

Later, the method was validated for linearity (table 4.4.), accuracy and

precision (table 4.5.). The results of validation study are presented in the following

tables.

Table 4.4. Spectrophotometric data for the estimation of Perindopril erbumine at


215.5 nm:

Concentration
Sl. No Absorbance*
(µg/mL)
1 0. 00 0.000

2 2. 00 0.032

3 4. 00 0.064

4 6. 00 0.099

5 8. 00 0.128

6 10. 00 0.160

*Average of three determinations

Department of Pharmaceutics, V.L.C.P, Raichur. 62


The linear regression analysis was done on absorbance data points. A straight-

line equation (Y = mx + c) was generated to facilitate the calculation of amount of

drug. Absorbance = slope x concentration + Intercept

y = 0.016 x - 0.00015

0.200
Absorbance (nm)

0.150

0.100

0.050

0.000
0 2 4 6 8 10 12
Concentration (µg)

Fig 4.1 Calibration Curve of Perindopril erbumine

4) Validation of analytical method developed in the study:

Table 4.5. Linearity studies of Perindopril erbumine:

S.No. Concentration ((µg/mL)) Absorbance (nm) Regression Data

1. 0.032
0. 00
2. 0.064
2. 00
c= -0.000124
m=0.0157

r= 0.9997

3. 0.098
4. 00
4. 0.131
6. 00
*
*

5. 0.160
8. 00
6. 0.032
10. 00

*m = slope; *c= intercept;* r = regression.

Department of Pharmaceutics, V.L.C.P, Raichur. 63


Table.4.6. Accuracy and precision studies:

Amount of Amount of
Drug Drug
Formulation Accuracy Precision
Drug added recovered
(mg/mL) (mg/mL)
PE1 4 3.80 95.51 0.001

PE2 4 3.85 96.30 0.003

PE3 4 3.84 96.14 0.001


Perindopril erbumine

PE4 4 3.92 98.02 0.001

PE5 4 3.94 98.54 0.001

PE6 4 3.93 98.38 0.002

PE7 4 3.93 98.43 0.001

PE8 4 3.91 97.91 0.002

PE9 4 3.90 97.70 0.0005

4.6. Methods used for characterization of the drug:

4.6.1. Melting point determination:

Melting point of the drug was determined by taking a small quantity of drug

in a capillary tube closed at one end which was placed in theil‟s melting point

apparatus. The temperature at which the drug melts was noted using liquid paraffin

as a solvent. Average of triplicate readings was recorded.

Department of Pharmaceutics, V.L.C.P, Raichur. 64


4.6.2. Solubility studies:

The solubility of the drug was determined in distilled water and different

solvents according to the method proposed by Diez et al. 63 Triplicate readings were

taken and average was calculated.

An excess amount of the drug was taken and dissolved in a measured

amount of distilled water in a volumetric flask to get a saturated solution. The

solution was shaken intermittently to assist the attainment of equilibrium with the un-

dissolved drug particles. Then measured quantity of the filtered drug solution was

withdrawn after 24 hours and successively diluted with distilled water suitably and

the concentration was measured in a UV spectrophotometer at their respective

absorbance maxima. Similarly the solubility of drug was determined in different

buffers, viz., pH 1.2, pH 2.2, pH 6.9, pH 7.0, pH 8.0, pH 9.0.

4.6.3. Partition coefficient:

The partition coefficient of the drug was determined by taking equal volumes

of n-octanol and aqueous phases in a separating funnel. Drug solution of 1mg/mL

was prepared, and 1mL of the prepared solution was added to n-octanol: water

(50:50) taken in a separating funnel and subsequently shaken for 10 minutes and

allowed to stand for 2 h. Both the phase was separated, centrifuged for 10 min at

2000 rpm. The aqueous and n-octanol phase was assayed before and after

partitioning using UV Spectrophotometer to get partition coefficient. Triplicate

readings (n=3) were taken and average was calculated.

Department of Pharmaceutics, V.L.C.P, Raichur. 65


4.6.4. Permeability coefficient:60

The permeability coefficient of drug was calculated by “Potts & Guy

equation”.

Log kp = − 2.7 + 0.71 × Log Ko/w − 0.0061 × Molecular weight

Where,

Log kp = Permeability coefficient

Ko/w = Partition coefficient

4.7. Preparation of Perindopril Erbumine Tablets

4.7.1 Method of preparation of granules:

Granules of Perindopril erbumine were prepared by wet granulation

technology. All the ingredients as per the formulae mentioned in table no. 4.6 were

weighed and grinded to fineness in a mortar and pestle. The powder blend was then

passed through sieve # 120. The powder was then kneaded with a clean and dry

pestle using starch paste. The wet mass so obtained was passed through mesh # 16.

The particles were then subjected to drying for 2-3 h in an oven at 40 °C. The dried

granules were then passed through mesh # 20. Fine particles to an extent of 20 %

were blended thoroughly with the particles of #16/20 and this powder blend was used

for further processing.

4.8. Evaluation of pre-compression characteristics of granule bed:

Department of Pharmaceutics, V.L.C.P, Raichur. 66


Granules prepared by wet granulation technology were evaluated for various

rheological properties like bulk density, tapped density, compressibility index, flow

properties (angle of repose) by using standard procedures. All studies were carried

out in triplicate (n=3) and average values were reported.

4.8.1. Bulk density (Db): 61

Bulk density was determined (bulk density apparatus, Konark instruments,

India) by taking accurately weighed quantity of the dried granules in a measuring

cylinder and recording the volume and weight of the total granules. Bulk density is

expressed in gm/mL and is given by,

Db = M / Vo

Db = Bulk density (gm/mL)

M is the weight of granules (gm)

Vo is the bulk volume of granules (mL)

4.8.2. Tapped Density (Dt): 61

Tapped density was determined (Tapped density apparatus, Konark

instruments, India) by taking accurately weighed quantity of the dried granules in a

measuring cylinder and recording the volume of granules after 100 tapping and

weight of the total granules.

Dt = M / V

Where, Dt = Tapped density (gm/mL)

M is the weight of granules (gm)


Department of Pharmaceutics, V.L.C.P, Raichur. 67
V is the tapped volume of granules (mL)

4.8.3. Compressibility index: 61

Compressibility index was determined by placing the dried granules in a

measuring cylinder and the volume (V0) was noted before tapping, after 100 tappings

again volume (V) was recorded.

Compressibility index = (1- V/ V0) X 100

V0 = volume of powder/granules before tapping.

V = volume of powder/granules after 100 tappings.

4.8.4. Angle of repose (θ): 61

It is defined as the maximum angle possible between the surface of pile of the

powder and the horizontal plane. Fixed funnel method was used. A funnel was fixed

with its tip at a given height (h), above a flat horizontal surface on which a graph

paper was placed. Powder was carefully poured through a funnel till the apex of the

conical pile just touches the tip of funnel. These studies were carried out before and

after incorporating lubricant/glidant. The angle of repose (θ) was then calculated.

θ = tan-1(h/r)

Where, θ = angle of repose

h = height of pile,

r = radius of the base of the pile.

Department of Pharmaceutics, V.L.C.P, Raichur. 68


4.9. Compression of tablets:

After adding lubricant (talc) and anti-adherent (magnesium stearate) to the dry

granule bed and subsequent blending, the granules were compressed into tablets on a

pilot press machine (PP1D, Chamunda Pharma, India) using 6 mm diameter, flat

faced punches at a pressure of approximately 5 kg /cm2.

4.10. Evaluation of post-compression characteristics of tablets:

Tablets were evaluated for their thickness, weight uniformity, hardness,

friability, disintegration time and dissolution profiles by using standard procedures.

4.10.1. Thickness and diameter: 62

Control of physical dimensions of the tablet such as thickness and diameter is

essential for consumer acceptance and tablet uniformity. The thickness and diameter

of the tablet was measured using Vernier callipers (Mitutoyo, Japan). The

measurements were in mm. Average of three readings were taken and the results were

tabulated (n = 3).

4.10.2. Hardness test: 62

Prepared tablets were evaluated for their hardness by using Pfizer hardness

tester. Scale was adjusted to zero; load was gradually increased until the tablet

Department of Pharmaceutics, V.L.C.P, Raichur. 69


fractured. The value of the load at that point gives a measure of hardness of the tablet.

Hardness was expressed in Kg/cm2. Triplicate readings were taken and average was

computed.

4.10.3. Weight uniformity: 62

Randomly selected ten tablets were weighed individually and together in a

single pan balance. The average weight was noted and standard deviation and

percent coefficient of variance was computed.

4.10.4. Friability test (F): 62

Tablet friability was tested using Roche Friabilator. Pre-weighed tablets were

allowed for 100 revolutions (4 min), taken out and dedusted. The percentage weight

loss was calculated by reweighing the tablets. The % friability was then calculated

by,

4.10.5. Disintegration time: 62

Disintegration test was performed for the prepared tablets in 900 mL, 0.1N

HCl at 37±2 oC by using USP disintegration apparatus. Time was noted with a digital

chronometer. Triplicate readings were taken and average was computed.

4.10.6. Swelling index: 64

Department of Pharmaceutics, V.L.C.P, Raichur. 70


The swelling properties of the tablets were evaluated by determination of

percent of swelling. Each tablet was weighed (W1) and immersed in a simulated

saliva fluid at pH6.8 for predetermined times. After immersing the formulation for

specified time, the tablets were wiped off to remove excess of surface water by using

filter paper and weighed (W2).

%Swelling = (W2) – (W1)/ (W1) X 100

Where,

W 1 is the initial weight of the tablet.

W2 is the weight of the tablet after the particular swelling time interval.

R = 100 (Wa - Wb) / Wb

4.10.7. Drug content uniformity: 62

From each batch three randomly selected tablets were weighed accurately and

powdered in a clean and dry glass mortar with pestle. Powder equivalent to 100 mg

of drug was transferred into 100 mL volumetric flask containing distilled water; the

remaining volume was made up to 100 mL with distilled water. Shaken

intermittently for 24 h and the solution was filtered, make up desired dilutions and

analysed for drug content at max 215.5.5 nm, using a distilled water as a blank.

Triplicate readings were taken and average was computed.

4.11 In vitro release study: 65

Department of Pharmaceutics, V.L.C.P, Raichur. 71


Apparatus : USP XXII (Type 2) Dissolution apparatus

Dissolution medium : 900 mL 0.1 N HCl (pH 1.2)

Temperature : 37± 0.5 °C

RPM : 50

Volume withdrawn and replaced : 1 mL

λ max : 215.5 nm

Blank solution : 0.1 N HCl

Procedure:

In-vitro drug release studies were carried out using USP XXII dissolution

apparatus type II (Campbell electronics, Mumbai) at 50 rpm. The dissolution medium

consisted of 900 mL of 0.1 N HCl, maintained at 37 + 0.5 °C. The drug release at

different time intervals was measured using a double beam UV spectrophotometer

(UV Pharmaspec 1700, Shimadzu, Japan) at 215.5 nm. The study was conducted in

triplicate.

4.12. Stability studies: 66

The stability experiments were conducted to investigate the influence of

temperature and relative humidity on the drug content and dissolution profile of

various fast dissolving tablets.

The formulations were exposed to a temperature of 40 °C and also a relative

humidity of 75 % RH. The sample was removed from the oven at the end of 24 hours

and analysed for drug content for 90 days. At the end of 90 days the tablets of each

formulation were subjected to dissolution as outlined in sec 4.11.8. Average of

Department of Pharmaceutics, V.L.C.P, Raichur. 72


triplicate readings was taken. The observations were tabulated. The dissolution

profiles were compared with dissolution profile performed on tablets kept at ambient

conditions.

4.13. Statistics: 67-68

The in vitro data was subjected to regression analysis by least squares method.

The standard deviation was calculated and reported. In vitro data was analysed by

ANOVA, a value of p< 0.05 was considered to be statistically significant.

Department of Pharmaceutics, V.L.C.P, Raichur. 73


Table.4.7. List of the ingredients used in formulae of different Fast dissolving tablets containing Perindopril erbumine.

S.No Ingredients (mg/tab) PE1 PE2 PE3 PE4 PE5 PE6 PE7 PE8 PE9

4 4 4 4 4 4 4 4 4
1. Perindopril erbumine

Starch (as 10% w/w) 15%w/w 15 15 15 15 15 15 15 15 15


2.
Xanthan gum
20 25 30 -- -- -- -- -- --
3.
(25%, 30% & 35% w/w)

Almond gum (25%,30% & 35%) -- -- -- 20 25 30 -- -- --


4.
Eudragit RL 100
-- -- -- -- -- -- 20 25 30
5.
(25%,30% & 35%)

Micro crystalline cellulose


59 54 49 59 54 49 59 54 49
6.
(diluent)

2 2 2 2 2 2 2 2 2
7. Talc and Mg stearate(2:1) 1.5%w/w

100 100 100 100 100 100 100 100 100


Total Tablet Weight (mg)

Department of Pharmaceutics, V.L.C.P, Raichur. 74


5. RESULTS

To maintain constant drug release, a drug delivery system should be able

to with stand a variety of physiological variables which may influence gastric

retention such as food, position of body and volume fluid intake; food related dose

dumping is also major concern.

Development of oral controlled release system has been a challenge to

a formulator because of their in ability to restrain and localize the system in the

targeted area of the GIT. Oral delivery of drugs is by far, the most referable

route of drug delivery due to the ease of administration and patient compliance.

There has been a tremendous progress in research and patent, from immediate

release tablet to controlled release tablets.

An ideal controlled drug delivery system is the one that delivers the drug

at a pre-determined rate, locally or systemically, for a specified period of time.

Controlled release is differs from sustained release systems which simply

prolong the drug release and hence plasma drug level for a longer period of time.

Thus the objective of most products should be controlled delivery to reduce

dosing frequency to an extent that once daily dose is sufficient for therapeutic

management through a uniform plasma concentration at steady state.

Hence drugs whose absorption window is in stomach or proximal

small intestine are incompletely absorbed and most of it is lost. It is also understood

that, as the solubility of the drug decreases the time available for drug dissolution

becomes less adequate and transit time becomes a significant factor affecting

Department of Pharmaceutics, V.L.C.P, Raichur. 75


the drug absorption, hence controlled drug delivery dosages are effective in

delivery of water soluble drugs like Perindopril erbumine. The objective of the study

was to formulate and evaluate controlled matrix tablets of Perindopril erbumine.

In this light of information the drug Perindopril erbumine was initially

characterized for its various physico-chemical properties. The results obtained from

various experiments, for the purpose of developing a suitable controlled matrix tablet,

are discussed below.

5.1. Characterization of Perindopril Erbumine:

The pure drug obtained was characterised for its melting point, solubility and

Partition coefficient. Melting point of the drug was found to be 126.5 C (n=3) which

corroborates with the previous report 126 to 128 C. Partition coefficient of the pure

drug was determined in n-Octanol: Water (50:50), and it was found to be 0.464 (n=3).

Later, an analytical method for the quantification of Perindopril erbumine was

developed using a double beam UV spectrophotometer. U.V. absorption maxima of

Perindopril Erbumine (PE) in water was found to be 215.5 nm (n=3) which is nearly

same as literature value 215 nm36. the analytical method was validated for linearity,

accuracy and precision. The results are given in tables 5.1. to 5.4.

Later, solubility of Perindopril Erbumine in distilled water, acidic and alkaline

pH buffers was studied. A saturated solution of the drug was prepared in respective

solvents and shaken, under ambient conditions, for 24 h. At the end of 24 h, 1mL of

the solution was pipetted out and the concentration (mg/mL) was determined using a

Department of Pharmaceutics, V.L.C.P, Raichur. 76


double beam UV spectrophotometer. The solubility was found to be 0.355 mg/mL in

distilled water. Similarly, the solubility of the drug in different buffers was also

determined. It was found that the solubility of Perindopril Erbumine is 0.555 mg/mL

in pH 1.2, 0.501 mg/mL in pH 2.2, 0.315 mg/ mL in pH 7.0, 0.098 mg/ mL in pH 8.0

and 0.066 mg/mL in pH 9.0. Average of triplicate readings was taken (n=3) and the

results obtained were tabulated and are summarized in table 5.1.

5.2. Development and evaluation of Controlled matrix tablets of Perindopril

Erbumine:

Perindopril Erbumine tablets were prepared by Wet granulation method using

different ratios of polymers like xanthun gum, eudragit RL 100, almond gum and

15% starch, microcrystalline cellulose. 15 % w/w of Starch paste (10 % w/w) was

used as binder in all formulations. All the powders were weighed and grounded to

fineness in a clean and dry mortar and pestle. The powder blend was than passed

through a sieve # 120. The powder was then kneaded with a clean and dry pestle

using starch paste prepared in distilled water as granulating fluid. The wet mass was

then passed through a mesh # 16. The particles were allowed to dry for 2-3 h in an

oven at 400C. The dried granules were then passed through a mesh # 18. Fine particles

to an extent of 20 % w/w were blended thoroughly with the particles of #16/18 and

than to this powder blend was added talc and magnesium stearate at 2:1 ratio and used

for further processing.

Department of Pharmaceutics, V.L.C.P, Raichur. 77


The dried granules of Perindopril Erbumine were characterized for rheological

properties. after mixing when granules are ready for punching then compressed into

tablets into 100 mg tablets on a pilot press machine (PP1D, Chamunda Pharma, India)

using 6 mm diameter, flat faced punches at a pressure of approximately 5-6 kgs /cm2.

Totally 9 formulations (PE1-PE9) were developed and studied.

The tablets were studied for compressional properties, Swelling index. In vitro

dissolution studies were carried out in USP XXIV dissolution apparatus and the

tablets were stored at 40°C / 75 % RH (ICH guidelines) to study the stability profile of

the drug.

5.2.1. Evaluation of pre-compression and compression characteristics of powder

bed of Perindopril Erbumine:

The compression powder bed were studied for their rheological behavior by

determining, angle of repose ( ), bulk density (gm/mL), tapped density (gm/mL),

and compressibility index (%), by adopting standard techniques described in Sec 4.8,

and the results are presented in the following paragraphs.

Carr‟s Compressibility index was found to be less than 15 % for all the 9

formulations. The index was observed to be between 3.03 % and 12.6 % indicating

that the powder bed is compressible.

Bulk density was found to be between 0.293 gm/mL and 0.415 gm/mL for

all the formulations. Tapped density was found to be between 0.308 gm/mL and

0.498 gm/mL. The angle of repose ( ) was determined before adding the

Department of Pharmaceutics, V.L.C.P, Raichur. 78


lubricants/glidants and after adding lubricants/glidants. The repose angle before

incorporation of lubricants/glidants was found to be 21.800 to 27.120. After the

addition of lubricants/glidants the angle of repose was found to be 19.980 and

24.110. These results indicate that, the powder beds of all the formulations are freely

flowable and easily compressible. The results, average of thee readings, was

computed and are given in table no 5.2.

Compressed tablets of Perindopril Erbumine were evaluated for several

compressional characteristics like thickness, diameter, weight uniformity, drug

content, swelling index, drug release profiles and stability studies . The weight of

the tablets was found to be fairly uniform ranging between, 100.8 ± 1.54 mg to

105.4 ± 6.32 mg for a 100 mg tablet (n=10). The thickness of the tablets was

determined using a micrometer (Mitutoyo, Japan). The thickness of the tablets, were

found to be between 3.008±0.0 mm to 3.010 ±0.002 (n=10). The hardness of the

tablets were determined with a hardness tester (Pfizer, India) and the hardness was

found to be fairly consistent and uniform, ranging between 3.26 ±0.115Kg/cm2 to

4.93 ±0.23Kg/cm2 (n=5). Drug content of the all formulations were in the range of

3.806 mg to 3.94mg for controlled matrix tablets. The friability of all the

formulations were determined in a friabilator operated for 5 min at 25 rpm, and the

percent friability was found to be 0.16% to 1.1%..

Further swelling behavior of the matrix tablets of Xanthan gum, eudragit

RL100 and almond gum were studied at pH6.8. The swelling index was computed as

discussed in section 4.10.6. The tablets of formulation from PE 1 to PE 7 were found

Department of Pharmaceutics, V.L.C.P, Raichur. 79


to loose their integrity and shape at the end of 8 h and similarly the tablets of PE 8 to

PE 9 lost their integrity at the end of 8 h. Hence the study was discontinued either

after 8 h or 6 h. The observations are presented in table.

It was also found that, increase in polymer content increases the swelling

index of the tablet formulations containing xanthun gum and the order of swelling

was found to be PE3 > PE2> PE1.

It was found that, increase in polymer content increases the swelling index of

the tablet formulations containing chitosan and the order of swelling was found to be

PE6> PE5> PE4. The study revealed that, amount of matrix carriers in the

formulation influences the swelling behavior rather than the amount of diluent. Also,

all the formulations containing matrix carriers swell considerably without losing the

shape or integrity of the tablet.

5.3. In vitro studies:

5.3.1. In vitro release studies of formulations PE1, PE2 and PE3:

The formulations PE1, PE2 and PE3 were prepared with 20% w/w, 25% w/w

and 30 % w/w of xanthun gum as polymer. Each of the tablet in all the formulations

contain 4 mg of Perindopril Erbumine, microcrystalline cellulose and 15% starch

paste as diluent and matrix binders .

The in vitro release data of formulation PE 1, containing 20 % w/w xanthun

gum as matrix carrier, is given in table 5.5 (n=3). The study was conducted in pH 7.4

Department of Pharmaceutics, V.L.C.P, Raichur. 80


for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h. „PE 1‟

formulation released nearly 40.56 %. The tablet was observed to be swelling and the

shape of the tablet was not distorted and it remained intact. When dissolution was

continued in buffer of pH 6.8 up to 24 h the formulation released nearly 83.49 % of

drug. Averages of triplicate readings were taken. The raw data obtained was subjected

to regression analysis by least squares method (r). The data was also analyzed

statistically by ANOVA, a value of p< 0.05 was considered to be significant. A high

regression coefficient (r= 0.978), indicating the percentage cumulative drug release vs

time curve is linear. There was no significant difference observed between the „p‟

values of any two readings of the same sampling interval. Plot percentage cumulative

drug release (%) vs time (h) is shown in figure 5.1 showed the curve obtained is linear

with an r = 0.978.

The swollen tablet left at the end of dissolution was carefully lifted and

collected on a blotting paper to drain any moisture present, The amount released in

dissolution study and the amount remaining in the tablet is found to be same as the

drug content of the tablet formulation. The tablet when further examined periodically

with naked eye showed no significant changes up to 8 h. After 8 h, small particles

were seen dissolution media (pH 6.8) for up to 24 h. After 24 h of dissolution, intact

gel mass remained in the basket but of reduced dimensions.

The results of controlled matrix tablets PE 2 containing 25 % w/w xanthun

gum as matrix carrier were tabulated as in table 5.6 (n=3). The study was conducted

in pH 7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h. PE

Department of Pharmaceutics, V.L.C.P, Raichur. 81


25 formulation released nearly 42.53 % of the drug in dissolution media. The tablet

was observed to be swelling and the shape of the tablet was not distorted and it

remained intact. When dissolution was continued in buffer of pH 6.8 upto 24 h the

formulation released 84.18 % (approx) of drug. Averages of triplicate readings were

taken. The raw data obtained was subjected to regression analysis by least squares

method (r). The data was also analyzed statistically by ANOVA, a value of p< 0.05

was considered to be significant. A high regression coefficient (r= 0.984), indicating

the percentage cumulative drug release vs time curve is linear.

There was no significant difference observed between the „p‟ values of any

two readings of the same sampling interval. Plot of percentage cumulative drug

release (%) vs time (h) is shown in figure 5.2 showed the curve obtained is linear with

an r = 0.984. The swollen tablet left at the end of dissolution was carefully lifted and

collected on a blotting paper to drain any moisture present, than remaining drug

content was quantified as described. The amount released in dissolution study and the

amount remaining in the tablet is found to be same as the drug content of the tablet

formulation.

The results of in vitro release data of formulation PE 3, containing 30% w/w

xanthun gum as matrix carrier, is given in table 5.7 (n=3). The study was conducted in

pH 7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h of study

and later the study was continued in pH 6.8 buffer up to 24 h. „PE 3‟ formulation

released nearly 45.11%. The tablet was observed to be swelling and the shape of the

tablet was not distorted and it remained intact. When dissolution was continued in

Department of Pharmaceutics, V.L.C.P, Raichur. 82


buffer of pH 6.8 up to 24 h the formulation released nearly 90.22 % of drug, Averages

of triplicate readings were taken. The raw data obtained was subjected to regression

analysis by least squares method (r). The data was also analyzed statistically by

ANOVA, a value of p< 0.05 was considered to be significant. A high regression

coefficient (r= 0.984), indicating the percentage cumulative amount drug release vs

time curve is linear.

There was no significant difference observed between the „p‟ values of any

two readings of the same sampling interval. Plot of the percentage cumulative amount

drug release (%) vs time (h) is shown in figure 5.3 showed the curve obtained is linear

with an r = 0.984. The swollen tablet left at the end of dissolution was carefully lifted

and collected on a blotting paper to drain any moisture present, than remaining drug

content. The amount released in dissolution study and the amount remaining in the

tablet is found to be same as the drug content of the tablet formulation.

5.3.2. In vitro release studies of formulations PE4, PE5 and PE6:

The formulations PE4, PE5 and PE6 were prepared with 20% w/w, 25% w/w

and 30 % w/w of Eudragit RL 100 as polymer. Each of the tablet in all the

formulations contain 4 mg of Perindopril Erbumine, microcrystalline cellulose and

15% starch paste as diluent and matrix binders .

The in vitro release data of formulation PE 4, containing 20 % w/w Eudragit

RL 100 as matrix carrier, is given in table 5.8 (n=3). The study was conducted in pH

7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h. „PE 4‟

Department of Pharmaceutics, V.L.C.P, Raichur. 83


formulation released nearly 44.39 %. The tablet was observed to be swelling and the

shape of the tablet was not distorted and it remained intact. When dissolution was

continued in buffer of pH 6.8 up to 24 h the formulation released nearly 88.79 % of

drug. Averages of triplicate readings were taken. The raw data obtained was subjected

to regression analysis by least squares method (r). The data was also analyzed

statistically by ANOVA, a value of p< 0.05 was considered to be significant. A high

regression coefficient (r= 0.986), indicating the percentage cumulative drug release vs

time curve is linear. There was no significant difference observed between the „p‟

values of any two readings of the same sampling interval. Plot percentage cumulative

drug release (%) vs time (h) is shown in figure 5.4 showed the curve obtained is linear

with an r = 0.986.

The swollen tablet left at the end of dissolution was carefully lifted and

collected on a blotting paper to drain any moisture present, The amount released in

dissolution study and the amount remaining in the tablet is found to be same as the

drug content of the tablet formulation. The tablet when further examined periodically

with naked eye showed no significant changes up to 8 h. After 8 h, small particles

were seen dissolution media (pH 6.8) for up to 24 h. After 24 h of dissolution, intact

gel mass remained in the basket but of reduced dimensions.

The results of controlled matrix tablets PE 5 containing 25 % w/w Eudragit

RL 100 as matrix carrier were tabulated as in table 5.9 (n=3). The study was

conducted in pH 7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to

12 h. PE 25 formulation released nearly 45.18 % of the drug in dissolution media. The

Department of Pharmaceutics, V.L.C.P, Raichur. 84


tablet was observed to be swelling and the shape of the tablet was not distorted and it

remained intact. When dissolution was continued in buffer of pH 6.8 upto 24 h the

formulation released 90.18 % (approx) of drug. Averages of triplicate readings were

taken. The raw data obtained was subjected to regression analysis by least squares

method (r =0.987). The data was also analyzed statistically by ANOVA, a value of p<

0.05 was considered to be significant. A high regression coefficient (r= 0.987),

indicating the percentage cumulative drug release vs time curve is linear.

There was no significant difference observed between the „p‟ values of any

two readings of the same sampling interval. Plot of percentage cumulative drug

release (%) vs time (h) is shown in figure 5.5 showed the curve obtained is linear with

an r = 0.987. The swollen tablet left at the end of dissolution was carefully lifted and

collected on a blotting paper to drain any moisture present, than remaining drug

content was quantified as described. The amount released in dissolution study and the

amount remaining in the tablet is found to be same as the drug content of the tablet

formulation.

The results of in vitro release data of formulation PE 6, containing 30% w/w

Eudragit RL 100 as matrix carrier, is given in table 5.10 (n=3). The study was

conducted in pH 7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to

12 h of study and later the study was continued in pH 6.8 buffer up to 24 h. „PE 3‟

formulation released nearly 44.76%. The tablet was observed to be swelling and the

shape of the tablet was not distorted and it remained intact. When dissolution was

continued in buffer of pH 6.8 up to 24 h the formulation released nearly 90.22 % of

Department of Pharmaceutics, V.L.C.P, Raichur. 85


drug, Averages of triplicate readings were taken. The raw data obtained was subjected

to regression analysis by least squares method (r). The data was also analyzed

statistically by ANOVA, a value of p< 0.05 was considered to be significant. A high

regression coefficient (r= 0.983), indicating the percentage cumulative amount drug

release vs time curve is linear.

There was no significant difference observed between the „p‟ values of any

two readings of the same sampling interval. Plot of the percentage cumulative amount

drug release (%) vs time (h) is shown in figure 5.6 showed the curve obtained is linear

with an r = 0.983. The swollen tablet left at the end of dissolution was carefully lifted

and collected on a blotting paper to drain any moisture present, than remaining drug

content. The amount released in dissolution study and the amount remaining in the

tablet is found to be same as the drug content of the tablet formulation.

5.3.3. In vitro release studies of formulations PE7, PE8 and PE9 :

The formulations PE7, PE8 and PE9 were prepared with 20% w/w, 25% w/w

and 30 % w/w of Almond gum as polymer. Each of the tablet in all the formulations

contain 4 mg of Perindopril Erbumine, microcrystalline cellulose and 15% starch

paste as diluent and matrix binders .

The in vitro release data of formulation PE 7, containing 20 % w/w Almond

gum as matrix carrier, is given in table 5.11 (n=3). The study was conducted in pH 7.4

for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h. „PE 7‟

formulation released nearly 46.90 %. The tablet was observed to be swelling and the

Department of Pharmaceutics, V.L.C.P, Raichur. 86


shape of the tablet was not distorted and it remained intact. When dissolution was

continued in buffer of pH 6.8 up to 24 h the formulation released nearly 93.02 % of

drug. Averages of triplicate readings were taken. The raw data obtained was subjected

to regression analysis by least squares method (r). The data was also analyzed

statistically by ANOVA, a value of p< 0.05 was considered to be significant. A high

regression coefficient (r= 0.986), indicating the percentage cumulative drug release vs

time curve is linear. There was no significant difference observed between the „p‟

values of any two readings of the same sampling interval. Plot percentage cumulative

drug release (%) vs time (h) is shown in figure 5.7 showed the curve obtained is linear

with an r = 0.986.

The swollen tablet left at the end of dissolution was carefully lifted and

collected on a blotting paper to drain any moisture present, The amount released in

dissolution study and the amount remaining in the tablet is found to be same as the

drug content of the tablet formulation. The tablet when further examined periodically

with naked eye showed no significant changes up to 8 h. After 8 h, small particles

were seen dissolution media (pH 6.8) for up to 24 h. After 24 h of dissolution, intact

gel mass remained in the basket but of reduced dimensions.

The results of controlled matrix tablets PE 8 containing 25 % w/w Almond

gum as matrix carrier were tabulated as in table 5.12 (n=3). The study was conducted

in pH 7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h. PE 8

formulation released nearly 47.58 % of the drug in dissolution media. The tablet was

observed to be swelling and the shape of the tablet was not distorted and it remained

Department of Pharmaceutics, V.L.C.P, Raichur. 87


intact. When dissolution was continued in buffer of pH 6.8 upto 24 h the formulation

released 92.18 % (approx) of drug. Averages of triplicate readings were taken. The

raw data obtained was subjected to regression analysis by least squares method (r

=0.987). The data was also analyzed statistically by ANOVA, a value of p< 0.05 was

considered to be significant. A high regression coefficient (r= 0.987), indicating the

percentage cumulative drug release vs time curve is linear.

There was no significant difference observed between the „p‟ values of any

two readings of the same sampling interval. Plot of percentage cumulative drug

release (%) vs time (h) is shown in figure 5.8 showed the curve obtained is linear with

an r = 0.987. The swollen tablet left at the end of dissolution was carefully lifted and

collected on a blotting paper to drain any moisture present, than remaining drug

content was quantified as described. The amount released in dissolution study and the

amount remaining in the tablet is found to be same as the drug content of the tablet

formulation.

The results of in vitro release data of formulation PE 9, containing 30% w/w

Almond gum as matrix carrier, is given in table 5.13 (n=3). The study was conducted

in pH 7.4 for first 6 h and the media was replaced with pH 6.8 buffer up to 12 h of

study and later the study was continued in pH 6.8 buffer up to 24 h. „PE 9‟

formulation released nearly 48.02%. The tablet was observed to be swelling and the

shape of the tablet was not distorted and it remained intact. When dissolution was

continued in buffer of pH 6.8 up to 24 h the formulation released nearly 96.22 % of

drug, Averages of triplicate readings were taken. The raw data obtained was subjected

Department of Pharmaceutics, V.L.C.P, Raichur. 88


to regression analysis by least squares method (r). The data was also analyzed

statistically by ANOVA, a value of p< 0.05 was considered to be significant. A high

regression coefficient (r= 0.986), indicating the percentage cumulative amount drug

release vs time curve is linear.

There was no significant difference observed between the „p‟ values of any

two readings of the same sampling interval. Plot of the percentage cumulative amount

drug release (%) vs time (h) is shown in figure 5.9 showed the curve obtained is linear

with an r = 0.986. The swollen tablet left at the end of dissolution was carefully lifted

and collected on a blotting paper to drain any moisture present, than remaining drug

content. The amount released in dissolution study and the amount remaining in the

tablet is found to be same as the drug content of the tablet formulation.

Stability studies were conducted according to ICH protocol at 400 C

/75% RH for a period of 90 days. The data of stability indicates that there was no

decrease in the drug content and In vitro drug release was observed over a period of

90 days and data is given in table 5.14 to5.19

Department of Pharmaceutics, V.L.C.P, Raichur. 89


Table: 5.1. Various physico-chemical characteristics of Perindopril Erbumine.

Solubility mg/ml Solubility mg/ml in Buffer pH


SL. Melting point Partition Log
Drug Name 0
No C coefficient KP
Water pH 1.2 pH 2.2 pH 7.0 pH 8 pH 9.0

Perindopril
126.50C 0.355 0.555 0.501 0.315 0.098 0.066 0.464 -0.3334
1 Erbumine

Department of Pharmaceutics, V.L.C.P, Raichur. 90


Table: 5.2 Evaluation of rheological characteristics of controlled release matrix

tablets of Perindopril Erbumine. (n=3).

Angle of repose ( º θ)
Bulk Tapped
Formulation Compressibility
density density Before After
Code index %
gm/ml gm/ml adding adding
glidants glidants

PE1 12.6 0.415 0.498 26.8 23.3

PE2 8.96 0.342 0.3757 27.07 20.85

PE3 12 0.398 0.452 28.9 24.11

PE4 6.06 0.308 0.32 22.56 22.30

PE5 3.03 0.298 0.308 21.80 19.98

PE6 6.06 0.293 0.312 22.33 22.06

PE7 7.06 0.299 0.318 26.5 23.19

PE8 12.1 0.297 0.338 26.21 22.05

PE9 8.96 0.342 0.375 27.12 20.85

Department of Pharmaceutics, V.L.C.P, Raichur. 91


Table: 5.3. Evaluation of Physical Characteristics of controlled release matrix tablets of Perindopril Erbumine (n=3).

Drug
Hardness content Disintegration
Formula Weight Thickness Diameter Friability
( kg/cm2 ( mg ± SD Time
code (mg ±SD) (mm ± SD) ( mg ± SD) %
±SD) (sec)

PE 1 100.4±1.24 4.93±0.23 3.009±0.002 6.008±0.000 3.806±0.001 0.8 9.667±0.115

PE 2 100.9±1.61 4.13±0.30 3.009±0.002 6.010±0.002 3.85±0.003 0.4 12.133±0.208

PE 3 100.4±1.56 3.73±0.11 3.008±0 6.009±0.002 3.84±0.001 0.16 14.267±0.152

PE 4 100.3±1.28 3.93±0.230 3.009±0.002 6.010±0.002 3.93±0.001 0.4 15.200±0.450

PE 5 100.8±1.16 4.13±0.115 3.008±0 6.009±0.002 3.91±0.002 0.6 17.567±0.115

PE 6 100.0±1.42 4.4±0.2 3.010±0.002 6.008±0.000 3.9±0.0005 0.6 21.500±0.500

PE 7 100.8±1.00 4.0±0.2 3.009±0.002 6.009±0.002 3.92±0.001 0.9 20.933±0.404

PE 8 100.8±1.28 3.53±0.115 3.009±0.002 6.010±0.002 3.94±0.001 1.8 23.000±0.500

PE 9 100.3±1.04 3.26±0.115 3.009±0.002 6.010±0.002 3.93±0.002 1.1 26.833±0.288

Department of Pharmaceutics, V.L.C.P, Raichur. 92


Table: 5.4. Swelling studies of controlled release matrix tablets of Perindopril Erbumine.

Swelling Index%

Time (h) 0.25 0.50 0.75 1 2 3 4 5 6 7 8

PE 1 31.26 39.07 43.67 48.33 52.91 54.46 55.89 61.39 64.10 66.61 70.86

PE 2 37.45 42.54 46.68 51.46 54.75 55.59 57.72 63.41 66.73 68.83 72.41

PE 3 41.15 44.81 48.53 52.33 56.61 57.38 59.62 64.59 68.81 71.98 74.97

PE 4 18.72 22.76 27.62 30.93 34.93 36.49 38.56 42.51 46.75 51.54 56.48

PE 5 27.83 33.58 36.48 39.59 42.92 44.42 50.57 53.36 56.22 56.58 59.47

PE 6 35.41 40.94 44.38 48.43 51.12 53.12 55.45 59.05 61.98 62.85 64.21

PE 7 30.58 32.50 37.92 43.45 45.38 47.41 50.13 55.42 58.17 60.68 61.88

PE 8 38.52 43.09 46.98 47.41 55.62 60.74 62.68 64.53 - - -

PE 9 39.47 45.62 48.21 51.28 59.45 63.56 65.31 66.54 - - -

Department of Pharmaceutics, V.L.C.P, Raichur. 93


Table: 5.5. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 20%w/w Xanthan gum , (PE1) (n=3).

Cumulative amount % Cumulative


S.No Time ( h )
(mg) Drug Release ± SD

1 0 0
0.000
2 0.16 0 0.000

3 0.5 0 0.000

4 0.75 0 0.000

5 1 0.004 0.105

6 2 0.039 1.037

7 3 0.115 3.038

8 4 0.220 5.780

9 5 0.340 8.959

10 6 0.478 12.578

11 7 0.631 16.607

12 8 0.795 20.911

13 9 0.968 25.462

14 10 1.148 30.204

15 11 1.344 35.357

16 12 1.542 40.566

Y=0.125X - 0.147

Department of Pharmaceutics, V.L.C.P, Raichur. 94


50

45

40
% Cumulative release

35

30

25

20

15
10

5
0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time (h)

Fig.5.1. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 20%w/w Xanthan gum (PE 1) .

Department of Pharmaceutics, V.L.C.P, Raichur. 95


Table: 5.6. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 25%w/w Xanthan gum (PE 2) (n=3).

Cumulative amount % Cumulative


S.No Time ( h )
(mg) Drug Release ± SD

1 0 0 0

2 0.16 0 0

3 0.5 0 0

4 0.75 0.004 0.109

5 1 0.016 0.407

6 2 0.076 1.980

7 3 0.171 4.449

8 4 0.283 7.378

9 5 0.413 10.742

10 6 0.552 14.377

11 7 0.705 18.365

12 8 0.869 22.624

13 9 1.043 27.154

14 10 1.232 32.091

15 11 1.430 37.245

16 12 1.633 42.535

Y=0.133 X - 0.136

Department of Pharmaceutics, V.L.C.P, Raichur. 96


50

45

40
% Cumulative release

35

30

25

20

15

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time(h)

Fig.5.2. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 25%w/w Xanthan gum (PE 2).

Department of Pharmaceutics, V.L.C.P, Raichur. 97


Table: 5.7. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 30%w/w Xanthan gum (PE 3) (n=3).

Cumulative amount % Cumulative


S .No Time ( h )
(mg) Drug Release ± SD

1 0 0 0

2 0.16 0 0

3 0.5 0 0

4 0.75 0.006 0.163

5 1 0.026 0.678

6 2 0.081 2.116

7 3 0.177 4.612

8 4 0.293 7.623

9 5 0.426 11.095

10 6 0.577 15.028

11 7 0.749 19.504

12 8 0.932 24.278

13 9 1.126 29.324

14 10 1.324 34.478

15 11 1.526 39.741

16 12 1.732 45.112

Y= 0.142X - 0.146

Department of Pharmaceutics, V.L.C.P, Raichur. 98


50

45

40

35
% Cumulative release

30

25

20

15

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time(h)

Fig.5.3. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 30%w/w Xanthan gum (PE 3) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 99


Table: 5.8. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 20%w/w Eudragit RL 100 (PE 4) (n=3).

Cumulative amount % Cumulative


S .No Time ( h )
(mg) Drug Release

1 0 0 0

2 0.16 0 0

3 0.5 0 0

4 0.75 0.003 0.080

5 1 0.022 0.557

6 2 0.075 1.908

7 3 0.170 4.320

8 4 0.297 7.554

9 5 0.446 11.344

10 6 0.614 15.612

11 7 0.789 20.065

12 8 0.968 24.624

13 9 1.153 29.342

14 10 1.343 34.166

15 11 1.543 39.255

16 12 1.745 44.397

Y = 0.145 - 0.986

Department of Pharmaceutics, V.L.C.P, Raichur. 100


50

45

40

35
% Cumulative release

30

25

20

15

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time (h)

Fig.5.4. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 20%w/w Eudragit RL 100 (PE 4) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 101


Table: 5.9. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 25%w/w Eudragit RL 100 (PE 5) (n=3).

Cumulative amount % Cumulative


S .No Time ( h )
(mg) Drug Release ± SD

1 0 0 0

2 0.16 0 0

3 0.5 0 0

4 0.75 0.007 0.186

5 1 0.029 0.746

6 2 0.090 2.291

7 3 0.184 4.715

8 4 0.313 7.992

9 5 0.461 11.802

10 6 0.626 16.011

11 7 0.799 20.434

12 8 0.980 25.069

13 9 1.168 29.865

14 10 1.359 34.767

15 11 1.560 39.908

16 12 1.767 45.183

Y = 0.146 X - 0.987

Department of Pharmaceutics, V.L.C.P, Raichur. 102


50

45

40

35
% Cumulative release

30

25

20

15

10

0
0 2 4 6 8 10 12
Time (h)

Fig.5.5. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 25%w/w Eudragit RL 100 (PE 5) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 103


Table: 5.10. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 30%w/w Eudragit RL 100 (PE 6) (n=3).

Cumulative amount % Cumulative


SI .No Time ( h )
(mg) Drug Release ± SD

1 0 0 0

2 0.16 0 0

3 0.5 0.000 0.000

4 0.75 0.005 0.128

5 1 0.014 0.369

6 2 0.073 1.864

7 3 0.171 4.375

8 4 0.289 7.420

9 5 0.426 10.919

10 6 0.580 14.872

11 7 0.751 19.252

12 8 0.935 23.980

13 9 1.129 28.948

14 10 1.331 34.129

15 11 1.536 39.391

16 12 1.747 44.786

Y = 0.143 X - 0.151

Department of Pharmaceutics, V.L.C.P, Raichur. 104


50

45

40
% Cumulative release

35

30

25

20

15

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time(h)

Fig.5.6. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 30%w/w Eudragit RL 100 (PE 6) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 105


Table: 5.11. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 20%w/w Almond gum (PE 7) (n=3).

Cumulative amount % Cumulative


SI .No Time ( h )
(mg) Drug Release ± SD

1 0 0 0

2 0.16 0 0

3 0.5 0 0

4 0.75 0.003 0.080

5 1 0.013 0.319

6 2 0.073 1.860

7 3 0.175 4.464

8 4 0.311 7.945

9 5 0.472 12.038

10 6 0.654 16.688

11 7 0.841 21.445

12 8 1.032 26.334

13 9 1.227 31.303

14 10 1.425 36.352

15 11 1.630 41.587

16 12 1.839 46.902

Y = 0.153 X - 0.158

Department of Pharmaceutics, V.L.C.P, Raichur. 106


50
45
40
% Cumulative release

35
30
25
20
15
10
5
0
0 1 2 3 4 5 6 7 8 9 10 11 12
Tim e(h)

Fig.5.7. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 20%w/w Almond gum (PE 7) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 107


Table: 5.12. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 25%w/w Almond gum (PE 8) (n=3).

Cumulative amount % Cumulative


SI .No Time ( h )
(mg) Drug Release

1 0 0 0

2 0.16 0 0

3 0.5 0 0

4 0.75 0.019 0.476

5 1 0.046 1.163

6 2 0.103 2.617

7 3 0.200 5.076

8 4 0.340 8.619

9 5 0.499 12.664

10 6 0.680 17.264

11 7 0.868 22.023

12 8 1.061 26.941

13 9 1.258 31.937

14 10 1.458 37.014

15 11 1.664 42.222

16 12 1.875 47.589

Y = 0.156 X - 0.158

Department of Pharmaceutics, V.L.C.P, Raichur. 108


50

45

40

35
% Cumulative release

30

25

20

15

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time(h)

Fig.5.9. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 25%w/w Almond gum (PE 8) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 109


Table: 5.13. In vitro release of Perindopril Erbumine from controlled

matrix tablets containing 30%w/w Almond gum (PE 9) (n=3).

Cumulative amount % Cumulative


SI .No Time ( h )
(mg) Drug Release ± SD

1 0
0.000 0.000
2 0.16
0.000 0.000
3 0.5 0.000 0.000

4 0.75 0.021 0.530

5 1 0.050 1.272

6 2 0.110 2.810

7 3 0.211 5.381

8 4 0.339 8.614

9 5 0.490 12.458

10 6 0.657 16.725

11 7 0.844 21.469

12 8 1.043 26.532

13 9 1.248 31.754

14 10 1.456 37.055

15 11 1.669 42.462

16 12 1.888 48.028

Y = 0.155 X - 0.146

Department of Pharmaceutics, V.L.C.P, Raichur. 110


50

45

40

35
% Cumulative release

30

25

20

15

10

0
0 1 2 3 4 5 6 7 8 9 10 11 12
Time(h)

Fig: 5.10. In vitro release of Perindopril Erbumine from controlled matrix tablets containing 30%w/w Almond gum (PE 9) (n=3).

Department of Pharmaceutics, V.L.C.P, Raichur. 111


Table 5.14: Drug content after 90 days of stability studies of controlled release
matrix tablets of Perindopril Erbumine (PE1, PE2, PE3) at
400C/75% RH (n=3).

S.No Time PE1 PE2 PE3


(days)
D.C (mg) D.C (mg) D.C (mg)

1 0 3.806 3.852 3.846

2 1 3.784 3.83 3.842

3 3 3.766 3.81 3.814

4 5 3.748 3.784 3.796

5 7 3.736 3.756 3.76

6 15 3.714 3.728 3.738

7 30 3.684 3.702 3.706

8 45 3.667 3.694 3.692

9 60 3.641 3.687 3.685

10 90 3.806 3.852 3.846

*Theoretical content = 4 mg

Department of Pharmaceutics, V.L.C.P, Raichur. 112


Table 5.15: Drug content after 90 days of stability studies of controlled release
matrix tablets of Perindopril Erbumine (PE4, PE5, PE6) at
400C/75% RH (n=3).

S.No Time PE4 PE5 PE6


(days)
D.C (mg) D.C (mg) D.C (mg)

1 0 3.938 3.917 3.908

2 1 3.876 3.842 3.89

3 3 3.852 3.8 3.814

4 5 3.796 3.778 3.746

5 7 3.749 3.752 3.732

6 15 3.678 3.73 3.711

7 30 3.644 3.714 3.687

8 45 3.632 3.691 3.674

9 60 3.624 3.679 3.663

10 90 3.938 3.917 3.908

*Theoretical content = 4 mg

Department of Pharmaceutics, V.L.C.P, Raichur. 113


Table 5.16: Drug content after 90 days of stability studies of controlled release
matrix tablets of Perindopril Erbumine (PE7, PE8, PE9) at
400C/75% RH (n=3).

PE7 PE8 PE9


Time
S.No
(days)
D.C (mg) D.C (mg) D.C (mg)

1 0 3.921 3.942 3.935

2 1 3.916 3.868 3.87

3 3 3.894 3.846 3.842

4 5 3.868 3.822 3.804

5 7 3.794 3.764 3.77

6 15 3.752 3.76 3.734

7 30 3.743 3.654 3.69

8 45 3.736 3.648 3.681

9 60 3.722 3.631 3.668

10 90 3.921 3.942 3.935

*Theoretical content = 4 mg

Department of Pharmaceutics, V.L.C.P, Raichur. 114


Table: 5.17. In vitro release after 90 days of stability studies of controlled release matrix tablets of Perindopril Erbumine
(PE1, PE2, PE3) at 400C/75% RH (n=3)

PE 1 PE 1 90 day PE 2 PE 2 90 day PE 3 PE 3 90 day

s.no time % cum amt % cum amt % cum amt % cum amt % cum amt % cum amt
1 0 0 0 0 0 0 0
2 0.16 0 0 0 0 0 0
3 0.5 0 0 0 0 0 0
4 0.75 0 0.006 0.004 0 0.006 0.003
5 1 0.004 0.025 0.016 0.006 0.026 0.013
6 2 0.039 0.063 0.076 0.041 0.081 0.072
7 3 0.115 0.119 0.171 0.116 0.177 0.169
8 4 0.220 0.194 0.283 0.216 0.293 0.281
9 5 0.340 0.294 0.413 0.338 0.426 0.409
10 6 0.478 0.422 0.552 0.475 0.577 0.547
11 7 0.631 0.556 0.705 0.628 0.749 0.700
12 8 0.795 0.703 0.869 0.797 0.932 0.859
13 9 0.968 0.875 1.043 0.972 1.126 1.028
14 10 1.148 1.050 1.232 1.153 1.324 1.213
15 11 1.344 1.234 1.430 1.347 1.526 1.406
16 12 1.542 1.425 1.633 1.544 1.732 1.609

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 115


Table: 5.18. In vitro release after 90 days of stability studies of controlled release matrix tablets of Perindopril Erbumine
(PE4, PE5, PE6) at 400C/75% RH (n=3).

PE 4 PE 4 90 day PE 5 PE 5 90 day PE 6 PE 6 90 day

s.no time % cum amt % cum amt % cum amt % cum amt % cum amt % cum amt
1 0 0 0 0 0 0 0
2 0.16 0 0 0 0 0 0
3 0.5 0 0 0 0 0 0
4 0.75 0 0 0 0 0 0
5 1 0.003 0 0.007 0 0.005 0
6 2 0.022 0.009 0.029 0.006 0.014 0.025
7 3 0.075 0.038 0.090 0.028 0.073 0.069
8 4 0.170 0.094 0.184 0.078 0.171 0.138
9 5 0.297 0.197 0.313 0.169 0.289 0.247
10 6 0.446 0.338 0.461 0.306 0.426 0.378
11 7 0.614 0.491 0.626 0.459 0.580 0.522
12 8 0.789 0.650 0.799 0.634 0.751 0.675
13 9 0.968 0.819 0.980 0.819 0.935 0.838
14 10 1.153 0.994 1.168 1.009 1.129 1.013
15 11 1.343 1.178 1.359 1.206 1.331 1.203
16 12 1.543 1.372 1.560 1.406 1.536 1.403

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 116


Table: 5.19. In vitro release after 90 days of stability studies of controlled release matrix tablets of Perindopril Erbumine
(PE7, PE8, PE9) at 400C/75% RH (n=3).

PE 7 PE 7 90 day PE 8 PE 8 90 day PE 9 PE 9 90 day

s.no time % cum amt % cum amt % cum amt % cum amt % cum amt % cum amt
1 0 0 0 0 0 0 0
2 0.16 0 0 0 0 0 0
3 0.5 0 0 0 0 0 0
4 0.75 0 0 0 0 0 0
5 1 0.003 0 0.007 0 0.005 0
6 2 0.022 0.009 0.029 0.006 0.014 0.025
7 3 0.075 0.038 0.090 0.028 0.073 0.069
8 4 0.170 0.094 0.184 0.078 0.171 0.138
9 5 0.297 0.197 0.313 0.169 0.289 0.247
10 6 0.446 0.338 0.461 0.306 0.426 0.378
11 7 0.614 0.491 0.626 0.459 0.580 0.522
12 8 0.789 0.650 0.799 0.634 0.751 0.675
13 9 0.968 0.819 0.980 0.819 0.935 0.838
14 10 1.153 0.994 1.168 1.009 1.129 1.013
15 11 1.343 1.178 1.359 1.206 1.331 1.203
16 12 1.543 1.372 1.560 1.406 1.536 1.403

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 117


6. DISCUSSION

Drug like Perindopril Erbumine has been selected as model drug because the

drug shows promising pharmacokinetics and physico-chemical properties required for

control release dosages. Perindopril Erbumine is a dipeptide monoacid monoester

with a perhydro indole group and no sulphydryl radical; chemical name, tert-

butylammonium (2S, 3aS, 7aS)-1-(N-[(S)-1-ethoxycarbonyl butyl]-L-alanyl)

perhydroindole-2-carboxylate and used for the treatment of patients with

hypertension and symptomatic heart failure. Perindopril Erbumine has molecular

weight of 368.49 gm/mol, oral bioavailability <90%, protein binding is 10-20% and

elimination half life is 1.2 hrs. Thus, it was considered as a potential drug for

controlled drug delivery.

Perindopril erbumine was characterized for physico-chemical properties. The

UV absorption maximum was found at 215.5 nm which was nearer to the literature

value 215-217 nm and the method developed during the course of this work was

validated for linearity, accuracy and precision. The method so developed was found

to be linear, accurate, reproducible and precise. The melting point of Perindopril

erbumine was found to be 126.5 C (n=3) which corroborates with previously

reported literature value 126-128 C.

From the study it was ascertained that the Perindopril erbumine sample

received as gift sample was fairly pure and the validated analytical method shows

that the analytical method developed follows Beer‟s law. The study of Partition

coefficient of the drug indicates that Perindopril erbumine is hydrophilic. The

solubility of Perindopril erbumine was found to decrease with increase in pH.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 118


In this present work, we developed controlled matrix tablets containing

Perindopril erbumine using microcrystalline cellulose as a direct compressible

vehicle. Xanthan gum, Eudragit RL100 and Almond gum polymers were used as

Matrix carriers at thee levels (20 % w/w, 25 % w/w, and 30 % w/w).

The pre-compressional characteristics were investigated for all the

formulations. Compressional characteristics of the controlled matrix tablets were also

investigated. In vitro release studies were conducted in a USP XXIV dissolution

apparatus using pH 6.8 as dissolution media at 370 C and 60 rpm using 900 mL of

dissolution fluid. In vitro studies were conducted to understand whether drug is being

released from the matrix slab. The study was conducted to know the actual release

profile of the drug for a period of 12 h. During the study 1 mL were samples were

withdrawn periodically and sink conditions were maintained. Further stability studies

according ICH guidelines, for accelerated stability of pharmaceuticals, were

conducted at 400 C/ 75% RH for a period of 90 days and the data of stability study is

given in tables 5.24 to 5.35.

Perindopril erbumine tablets were prepared by dry granulation compression

using different ratios of polymers like Xanthan gum, eudragit RL 100, almond gum

using microcrystalline cellulose as a hydrophilic matrix carrier. All the ingredients

were weighed, passed though #120 and blended to fineness in a clean and dry mortar

and pestle for 10mins. After sufficient mixing of drug as well as other ingredients talc

(lubricant) and magnesium stearate (anti-adherent) was added at 2:1 ratio and further

mixed for additional 3-5mins and then compressed into in to 100 mg tablets on a pilot

press machine (PP1D, Chamunda Pharma, India) using 6 mm diameter, flat faced

punches at a pressure of approximately 5-6 kgs /cm2. Totally 9 formulations


Deptartment of Pharmaceutics, V.L.C.P, Raichur. 119
(PE1-PE9) were developed and studied. Also, the powder bed prior to compression

was characterized for rheological properties.

Study of powder rheology indicated a Carr‟s compressibility index of less

than 15 % and therefore it was ascertained that, the powder bed is compressible. The

study of repose angle showed less than 300. This indicated the powder beds of all the

formulations are freely flowable and easily compressible and further improvement

was seen when glidants/ lubricants were added. It was understood that the powder is

less bulky as the bulk density and tapped density of powder bed was found to be less

than 1 (table 5.2).

The thickness, diameter and weight of the tablets were found to be uniform

and consistent as indicated by their low SD values (table 5.3). The hardness of the

compressed tablets determined using hardness tester (Pfizer, India) indicates that the

tablets are of adequate strength. The percent friability of the tablets was very low and

therefore it was understood that the tablets are of sufficient strength to withstand the

stress of transportation. Drug content of all tablet formulations was found to be

uniform. The study on swelling behaviour revealed that, amount of matrix carriers in

the formulation influences the swelling rather than the amount of diluent. Also, all the

formulations containing matrix carriers swell considerably for a significant period,

without losing the shape or integrity of the tablet. Swelling was observed to be

increasing with increase in matrix polymer content in the formulation.

The in vitro dissolution study was conducted using USP XXIV dissolution

testing apparatus in pH 6.8 at 50 rpm and temperature was maintained at 37 0.5 C.

The dissolution was carried out for 12 h and samples of 1 mL were withdrawn

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 120


periodically and analyzed spectrophotometrically at 215.5 nm. Average of triplicates

readings was considered and the data have been tabulated and shown in tables 5.5 to

5.13. In vitro release data obtained was statistically analyzed by ANOVA and a value

of p < 0.05 was considered to be significant. The raw data was also subjected to

regression analyses by least squares method.

Matrix tablets containing gelatin were studied for in vitro release. PE1

containing 20 % w/w xanthun gum, released 40.56 % of drug at end of 12 h. And,

Formulation PE2, containing 25% w/w xanthun gum released 42.53 %. Similarly PE3

containing 30 % w/w xanthun gum released 45.11% of the drug at end of 12 h. As the

content of xanthun gum in the tablet is increased, the rate of release is retarded as

indicated by their slope values. The linearity of the curve ( r= 0.984) indicates that the

drug release could be following zero order kinetics with a rate of 0.125 , 0.133, and

0.142 , respectively.

PE4 containing 20 % w/w eudragit RL100 released 44.39% of drug at end of

12 h. And, Formulation PE5, containing 25 % w/w eudragit RL100 released 45.18 %.

Similarly PE6 containing 30 % w/w eudragit RL100 released 44.78 % of the drug at

end of 12 h. As the content of eudragit RL100 in the tablet is increased, the rate of

release is retarded as indicated by their slope values. The linearity of the curve

(r=0.987) indicates that the drug release could be following zero order kinetics with a

rate of 0.145, 0.146, and 0.143, respectively.

PE7 containing 20 % w/w almond gum released 46.90 % of drug at end of

12 h. And, Formulation PE8, containing 25 % w/w almond gum 47.58 %. Similarly

PE 9 containing 30 % w/w xanthan gum 48.02 % of the drug at end of 12 h. As the


Deptartment of Pharmaceutics, V.L.C.P, Raichur. 121
content of almond gum in the tablet is increased, the amount of drug released is

retarded the rate of release is retarded as indicated by their slope. The linearity of the

curve (r= 0.987) indicates that the drug release could be following zero order kinetics

with a rate of 0.153 , 0.156, and 0.155, respectively.

Stability studies were conducted according to ICH protocol at 40 °C /75% RH

for a period of 90 days. The data of stability study shown in table 5.14-5.19 indicates

that there was no decrease in the drug content over a period of 90 days. At the end of

90 days the tablets were subjected to dissolution studies, the data is presented in table

5.19, and the profiles so obtained were compared with the profiles of in vitro release

obtained without stress (400 C/ 75% RH). It was found that the dissolution profiles

were comparable and there was no significant difference observed between the means

of dissolution profile conducted without stress and with stress at p<0.05.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 122


7. CONCLUSIONS

In the present work, an attempt was made to develop Controlled matrix tablets

of Perindopril Erbumine as an improved and better patient compliant dosage form.

From the study conducted, the following conclusions are drawn.

Controlled formulations of Perindopril Erbumine in the form of controlled

matrix tablets were developed to a satisfactory level, in terms of drug release, content

uniformity, swelling index, friability, hardness and weight variation.

 The analytical method developed during the course of the work resulted in a

λ max of 215.5 nm which corroborates with the literature value of 215-216 nm.36

 The method was found to be linear, precise and accurate.

 Melting point of Perindopril erbumine was found to be 129.5 °C which

corroborates with the literature value of 126-128 °C. 37

 The solubility of drug was found to be 355 μg/mL in distilled water.

 Partition coefficient of the pure drug was determined in n-Octanol: Water

(50:50), and it was found to be 0.464 (n=3), corroborating with the reported

literature value of 0.489. Indicating its lipophilic nature.

 Bulk density of powder bed for formulation was in the range of 0.293 gm/mL -

0.415 gm/mL, which was found to be less than 1 mg/mL for all formulations.

Similarly, the tapped density of powder bed for formulations was in the range of

0.498 mg/mL - 0.312 mg/mL which was less than 1 mg/mL for all formulations.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 123


 The compressibility index was 3.03% - 12.6% for Controlled matrix tablets

which was found to be less than 15 % indicating that the powder bed is

compressed into a compact mass of tablet.

 Angle of repose (°θ) for the formulations before adding glidants was 21.8°-

28.9°. Angle of repose (°θ) for the formulations after adding glidants was

19.98°- 24.11°. A considerable decrease in angle of repose upon addition of

glidants (Talc: Magnesium Stearate = 2:1) signifies improved flow ability.

 The weight of Controlled matrix tablets containing Perindopril erbumine was

found to be fairly uniform ranging between, 100.3 ± 1.04 to 100.9 ± 1.61 mg for

a 100 mg tablet (n=10).

 The thickness of the tablets of all the formulation was in the range of 3.008 ±

0.002 mm to 3.010 ± 0.002 (n=3).

 The hardness was found to be fairly consistent and uniform, ranging between

3.26 ± 0.115 Kg/cm2 to 4.93 ± 0.23 Kg/cm2 (n=3).

 The drug content of all the formulations having dose, of 4 mg were found to be

fairly uniform, reproducible and consistent, ranging between 3.806± 0.001 mg

to 3.94 ± 0.001mg per tablet of 100 mg.

 The friability of all the formulations were determined in a friabilator operated

for 4 min at 25 rpm, and the percent friability was found to be ranging 0.4 % to

0.9%.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 124


 The Controlled matrix tablets showed a disintegration time ranging from 15 ± 1

to 70 ± 0 seconds, owing to the variation in amount and type of

superdisintegrant incorporated.

 Percent swelling index was to be increasing with time and with increase in

hydrophilic polymer content. Controlled matrix tablets containing Xanthan gum

showed better swelling index and mechanical properties followed by Controlled

matrix tablets containing almond gum and eudragit RL 100.

 The cumulative percentage release of drug from PE1, PE2 and PE3 were found

to be 40.56% (r = 0.978), 42.53% (r = 0.984) and 45.112 % (r = 0.984) within

12 h of study respectively.

 The cumulative percentage release of drug from PE4, PE5 and PE6 were found

to be 44.39% (r = 0.986), 45.18% (r = 0.987) and 44.78 % (r = 0.983) within

12 h of study respectively.

 The cumulative percentage release of drug from PE7, PE8 and PE9 were found

to be 46.90 % (r = 0.986), 47.58% (r= 0.987) and 48.02% (r = 0.986) within 12

h of study respectively.

 Stability studies conducted as per ICH guidelines at 40 °C / 75 % RH indicated

that there is no decrease in drug content or no significant difference between the

means of profiles without stress and with stress at p<0.05 observed for a period

of 3 months . Thus, the manufactured fast dissolving tablets were found to be

stable with respect to physicochemical and release characteristics.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 125


The investigations so far and the results obtained there of, for various

formulations fulfilled the objectives of the current investigation however, further

studies could be carried out to know the effect of varying compressional parameters

on actual production and also actual in vivo benefit to a patient.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 126


8. SUMMARY

Controlled release tablets are dosage forms which remain in the

gastro- intestinal tract for a long period, improve the bioavailability of those

drugs whose therapeutic window is in the stomach or proximal part of small

intestine. Such controlled release dosages are therapeutically highly beneficial

in the treatment of hypertension.

The present investigation was planned to formulate controlled release

tablets to improve perindopril erbumine bioavailability. The starch as binder and

talc and magnesium stearate (2:1) as glidants were optimized. Also the effect of

various matrix carrier excipients like Microcrystalline cellulose, Xanthan gum,

Almond gum, Eudragit RL 100 was studied. Chapter 1 gives an over view of the

controlled release dosage forms and hypertension. Current literature cites various

approaches and different drugs that have been successfully formulated and

evaluated, has been described in chapter 3. Throughout the work, standard

techniques and methods, as outlined in various text books, research articles and

other sources were reviewed and used in this investigation. The investigation

carried out on various formulations resulted in totally all formulation obeying

fickian diffusion or zero order release kinetics. Chapter 5 and 6, describes the

results obtained from various experiments conducted and discusses the results

and mechanism of the release. The study on rheological characteristics of

powder/granule bed indicated that, all the granules were freely flowing and

compressible. Studies on compression characteristics indicated that the tablet

remain intact over the period of more than 9 h. drug content was found to be more

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 127


than 93 % and was fairly uniform and consistent.

In vitro results indicated that, Microcrystalline cellulose, a l m o n d g u m ,

xanthun gum and eudragit RL 100 tablets containing starch as binder follow fickian

diffusion or zero order release kinetics. The increasing the amount of glidants does

not significantly alter the drug release kinetics, it improve the flow ability of the

granule bed. Stability studies at 40ºC and 75 % RH for a p e r iod of 90 days. The

drug content found at the end of everyday shows that there is little decrease in

drug content which is statistically not significant (p<0.05).

Conclusions drawn from the results are mentioned in chapter-7. The present

study conclusively proved that Perindopril erbumine matrix tablets could be

successfully developed using various polymers. The prepared tablets gave promising

results with respect to swelling strength and in vitro release from the dosage form.

Vancouver style was followed to quote the references in the study and is listed in the

chapter of Bibliography (chapter 9).

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 128


9. BIBLIOGRAPHY
1. John A.Gater. Drugs affecting renal and cardiovascular function:

Antihypertensive, Agents and the drug therapy of Hypertension In chapter 33

of, Goodman and Gilman‟s Pharmacological Basis of Therapeutics, 9th

edition., Editers. Joel G.Hardman, and Lee E. Limbard, Mc Graw Hill – health

professions division, New York: 1996; 781-808.

2. Satoskar RS, Bhandarkar SD and Ainapure SS. Cardiovascular drugs.

In chapter 7 of, Pharmacology and Pharmacotherapeutics, 14th edition.,

Popular prakashan, Bombay, India : 1995; 325-410.

3. http://publishing.eur.nl/ir/repub/asset/7584/060315_schelleman-h.pdf/24/

02/2011.

4. Harsh Mohan, The kidney and lower urinary tract. In chapter 19, Texbook

of pathology, 4th edition, Jayppe Brothers Medical Pubhlishers New Delhi:

2000;670-672.

5. Neal L, Benowitz MD. Antihypertensive Agents. In chapter 11, Basic

and clinical pharmacology, 6th edition, editor Bertram G. Katzung

Appleton and Lange: 1995; 147: 165-166.

6. Rang HP, Dale MM. The vascular system. In Pharmacology. 5th

edition,Churchill Livingstone, New York: 2003; 298-302.

7. Tripathi KD. Anti hypertensive drugs. Essentials of medical pharmacology.

5thedition, Jayppe brothers medical publishers, New Delhi; 2003.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 129


8. Chien YW. Concepts and system design for rate controlled drug delivery. In

chapter 1 of, Novel Drug Delivery Systems. 2nd edition, Marcel Decker

Inc.,New York: 1992; pp1

9. Brahmankar DM and Jaiswal SB. Biopharmaceutics and pharmacokinetics -

Atreatise. 1st edition. Vallabh Prakashan, Delhi: 2000.

10. Chien YW. Parenteral drug delivery and delivery systems. In chapter 8

of,Novel Drug Delivery Systems. 2nd edition, Marcel Dekker Inc., New

York:1992; 381.

11. Vyas SP and Khar RK. Essentials of controlled drug delivery. In chapter 1

of,Controlled drug delivery concepts and advances,1st edition.

VallabhPrakashan, Delhi: 2005.

12. http://dissertations.ub.rug.nl/Files/faculties/science/2005/r.steendam/c2.pdf

24/02/ 2010.

13. Reza MS, Quadir MA and Haider SS. Comparative evaluation of

plastic,hydrophobic and hydrophilic polymers as matrices for controlled-

release drugdelivery. J Pharma Sci 2003;6(2):282-91.

14. Takka S, Rajbhandari S and Sakr A. Effect of anionic polymers on the

releaseof propranolol hydrochloride from matrix tablets. Eur J Pharm

Biopharm2001;52:75-82.

15. Bandari S, Mittapalli RK, Gannu R, Rao MY. Orodispersible tablets: An

overview. Asian Journal of Pharmaceutics, 2008; 2 (1): 2-11.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 130


16. Mudbidri A. Tablet compression principles. Pharma Times. 2010; 42 (11):

44-47.

17. Shirwaikar A A, Jacob S and Grove V. Formulation and evaluation of

sustained release tablets using an insoluble rosin matrix system Ind. J.

Pharm.Sci., 2005; 67 (1): 80-83.

18. Nevin Erk. Comparison of spectrophotometric and an LC method for the

determination Perindopril and indapamide in pharmaceutical formulations. J

of Pharmaceutical and Biomedical Analysis. 2001. 26. 43–52.

19. Prameela Rani. A, Bala Sekaran. A validated RP-HPLC method for the

determination of perindopril erbumine in pharmaceutical formulations.

International Journal of PharmTech and Research. 2009;1(3):575-578.

20. Sarojini .S, Deepthi S. Kunam . Effect of natural almond gum as a binder in

the formulation of diclofenac sodium tablets. International journal of

PharmTech and Research.2010;1(3) : 55-60.

21. Ankit. B. Chaudhary, Rakesh. K. patel. Determination of Losartan Potassium

and Perindopril Erbumine in Tablet Formulations byReversed-Phase HPLC.

International Journal of ChemTech Research,2010 ; 2(2) : 1141-1146.

22. Raja sekharan. T, Palanichamy.S. Formulation and Evaluation of

Theophylline Controlled ReleaseMatrix Tablets using Xanthan gum. Der

Pharmacia Lettre, 2009; 1 (2) 93-101.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 131


23. Bhanja Satyabrata, Mohanty Chandan. Design and in vitro Evaluation of

Mucoadhesive Buccal Tablets of Perindopril Prepared by Sintering Technique.

International Journal of PharmTech Research. 2010;2(3): 1810-1823.

24. Madhusudan Rao Y , Ranjith Kumar.M . Factors Influencing the Design and

Performance of Oral Sustained/Controlled Release Dosage Forms.

International Journal of Pharmaceutical Sciences and Nanotechnology. 2009;

2(3): 583-594.

25. Thawatchai Phaechamud , Garnpimol . C.R., Controlled release of Propronalol

HCL from chitosan-laclose-xanthan gum in matrix tablets.

26. Harry GB, Morris KR, Bugay DE, Thakur AB and Serajuddin ATM. Solid

state NMR and IR for the analysis of pharmaceutical solids: polymorphs of

fosinopril sodium. International Journal Pharm and Biomed Analysis

1993;11(11):1063-1069.

27. Basak S C, Srinivas Rao Y, Manavalan R and Rama Rao P. Controlled release

HPMC matrix tablets of propranolol hydrochloride. Ind. J. Pharm.Sci., 2004;

66(6): 827-830.

28. Sunil Jambhekar, Casella R and Maher T. The physicochemical characteristics

and bioavailability of indomethacin from cyclodextrin,

hydroxyethylcyclodextrin, and hydroxypropyl cyclodextrin complexes. Int J

Pharm 2004;270:149–166.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 132


29. Medenica. M, Ivanovic.D, Evaluation of impurities level of perindopril tert-

butylamine in tablets. Journal of Pharmaceutical and Biomedical Analysis,

2009; 44: 1087–1094.

30. Hisham E. Abdellatef, Magda M. Ayad, Spectrophotometric and atomic

absorption spectrometric determination of ramipril and perindopril through

ternary complex formation with eosin and Cu(II). Journal of Pharmaceutical

and Biomedical Analysis 1999; 18 : 1021-1027.

31. Hisham E. Abdellatef, Utility of certain p-acceptors for the

Spectrophotometric determination of perindopril. Journal of Pharmaceutical

and Biomedical Analysis 1998; 17: 1267-1271.

32. Kenneth I. Ozoemena , Raluca-Ioana Stefan. Enantioanalysis of S-perindopril

using different cyclodextrin-based potentiometric sensors. 2005; 425–429.

33. Yeole PG, Galgatte UC, Babla I.B and Nakhat PD. Design and evaluation of

Xanthan gum based sustained release matrix tablets of diclofenac sodium. Ind

J Pharm Sci 2006;8(2):185-189.

34. Reynolds DT, Gehrke SH, Hussain AS and Shenouda LS. Polymer Erosionn

and Drug Release Characterization of HydroxypropylmethylcelluloseMatrices.

J. Pharm. Sci 1998: 87(9);1115-1123.

35. Lutfi Genc. Studies on controlled release dimenhydrinate from matrix tablet

formulations. Pharma. Acta. 1999:74;43-49.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 133


36. Damle inalini C, Dewani Mohit G. Development and validation of stability

indicating hptlc method for determination of perindopril erbumine, 2010;1(1):

428-438.

37. Alderman DA, A review of cellulose ethers hydrophilic matrices for oral

controlled- release dosage forms. International Journal of Pharm.Tech. Prod.

Manuf.1984;5.

38. www.druginfosys.com/Drug.aspx/drugCode961/perindoprilerbumine24/03/10.

39. www.chemblink.com/products/107133-36-8/html.

40. www.tradeindia.com/fp518124/html.

41. www.en.wikipedia.org/wiki/Perindopril.

42. www.medicinescomplete.com/search.html/perindopril erbumine

43. Jeffrey Atkinson Cardiovascular Drug Reviews Vol. 10. No. 4. pp. 446-471

44. www.mims.com/Page.aspx/menuid&name/perindopril erbumine.

45. www.pharmaceutical-drugmanufacturers.com/pharmaceuticaldrugs/

perindopril erbumine.html.

46. www.cimsasia.com/Page.aspx/menuid&name/perindopril erbumine.

47. Araujo OE. Emulsifying properties of new polysaccharide gum. J Phrm

Sci. 1967; 56: 1141-1145.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 134


48. Booth AN, Hendrickson AP, Deds F. Physiologic effects of three microbial

polysaccharides on rats. Toxicol Appl Pharmaco. 1963; 1(5): 478-84.

49. Charm SE,McComis W. Physical measurements of gums. Food Technology.

1965; 19: 948-953.

50. Dea ICM, McKinnon AA and Rees DA.Tertiary and quarternary structure in

aqueous polysaccharide systems which model cell wall cohesion. Reversible

changes in conformation and association of agarose, carrageenan,and

galactomannas. J Mol Bio. 1972; 68(1): 153-172.

51. Dintzis FR, Babcock GE,and Tobin R. Studies on dilute solutions and

dispersions of polysaccharide from Xanthomonas campestris. NRRL B – 459.

Carbohydrate Research. 1970; 13: 257-267.

52. Composition of Almond-Tree Gum. R. Huerre. Joum. Pharm. Chinz., 1908;

27: 561-569

53. Wang S, Xie Bi-xia, Zhong Qiu-ping, DU Hong-yan. Rheological properties

in supernatant of peach gum from almond (Prunus dulcis). J. Cent. South

Univ. Technol. 2008; 15(s1): 509−515.

54. Degussa Creating essentials. Specification and test methods for Eudragit

RL100 and Eudragit RS 100.2004-2009:1-4.

55. http://eudragit.evonik.com/product/eudragit/en/products-services/eudragit

products/protective-formulations/e-100/pages/default.aspx.

56. www.medicinenet.com/mc/starch/pharmaceutical excipients.html 02/02/11.


Deptartment of Pharmaceutics, V.L.C.P, Raichur. 135
57. Rowe RC, Sheskey PJ, Owen SC. Handbook of pharmaceutical excipients 5th

ed. Joint publication of American Pharmaceutical Association and

Pharmaceutical Society of Great Britain. 2006; 430-433.

58. http://commons.wikimedia.org/wiki/File:Magnesium_stearate.png. accessed

on 02.02.2011.

59. Rowe RC, Sheskey PJ, Owen SC. Handbook of pharmaceutical excipients 5th

ed. Joint publication of American Pharmaceutical Association and

Pharmaceutical Society of Great Britain. 2006; 767-769.

60. Potts RO, Guy RH. Predicting skin permeability. Pharm. Res. 1992; 9(5): 663-

669.

61. Aulton ME. “Powder flow”, Ch. 14, in „Pharmaceutics-The science of dosage

form design‟ 2nd ed. Churchill Livingstone, Spain. 2002; 205-208.

62. Banker GS, Rhodes CT. “Tablet dosage form”, Ch. 10, in „Modern

pharmaceutics‟ 2nd ed. Marcel Dekker Inc, Newyork. 1990; 40: 416-420.

63. Diez Colom H, Moreno Obach R. Comparative in vitro study of transdermal

absorption of series of calcium channel antagonists. J Pharm Sci. 1991; 80

(10): 932-934.

64. Mahapatra AK, Murthy PN, Sahoo J, Biswal S, Sahoo SK. Formulation design

and optimization of mouth dissolving tablets of levocetrizine hydrochloride

using sublimation technique. Indian J. Pharm. Educ. Res. 2009; 43 (1): 39-45.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 136


65. http://www.pharmasm.com/pdf_files/poongothai.pdf. accessed on 01.02.2011

66. Matthews B R, Wall G M. Regulatory aspects of stability testing in Europe.

Drug Dev Ind Pharm. 1999; 25 (7): 831-856.

67. Mahajan BK. Methods in biostastics. 6th ed. Jaypee Bros. Medical publishers

Pvt. Ltd, New Delhi. 1997; 66-75.

68. Smith RV, Stewart JT. Textbook of biopharmaceutic analysis. Lea and

Febiger, Philadelphia. 1981; 90-92.

Deptartment of Pharmaceutics, V.L.C.P, Raichur. 137

You might also like