07.06.2013 Views

DEVELOPMENT AND EVALUATION OF PULSATILE DRUG ...

DEVELOPMENT AND EVALUATION OF PULSATILE DRUG ...

DEVELOPMENT AND EVALUATION OF PULSATILE DRUG ...

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

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

<strong>PULSATILE</strong> <strong>DRUG</strong> DELIVERY SYSTEM <strong>OF</strong><br />

FLURBIPR<strong>OF</strong>EN<br />

By<br />

PATEL VISHAL ASHOK<br />

Reg. No. 08 PU 251<br />

Dissertation submitted to the<br />

Rajiv Gandhi University of Health Sciences, Karnataka, Bangalore.<br />

In partial fulfillment<br />

of the requirements for the degree of<br />

MASTER <strong>OF</strong> PHARMACY<br />

in<br />

PHARMACEUTICS<br />

Under the guidance of<br />

Mr. M. NAJMUDDIN<br />

M. Pharm. (Ph. D.)<br />

Associate Professor<br />

DEPARTMENT <strong>OF</strong> PHARMACEUTICS<br />

LUQMAN COLLEGE <strong>OF</strong> PHARMACY<br />

GULBARGA. 585102 (KARNATAKA)<br />

2010<br />

i


RAJIV G<strong>AND</strong>HI UNIVERSITY <strong>OF</strong> HEALTH SCIENCES,<br />

KARNATAKA, BANGALORE<br />

DECLARATION BY THE C<strong>AND</strong>IDATE<br />

I hereby declare that this dissertation/thesis entitled<br />

“Development and Evaluation of Pulsatile Drug Delivery System<br />

of Flurbiprofen” is a bonafide and genuine research work carried out<br />

by me under the guidance of Mr. M. Najmuddin, Associate professor<br />

of Luqman college of Pharmacy Gulbarga.<br />

Date:<br />

Place: Gulbarga <br />

ii


RAJIV G<strong>AND</strong>HI UNIVERSITY <strong>OF</strong> HEALTH SCIENCES,<br />

KARNATAKA, BANGALORE<br />

CERTIFICATE BY THE GUIDE<br />

This is to certify that the dissertation entitled “Development<br />

and Evaluation of Pulsatile Drug Delivery System of<br />

Flurbiprofen” is a bonafide research work done by Mr. Patel Vishal<br />

Ashok in partial fulfillment of the requirement for the degree of<br />

“Master of Pharmacy” in Pharmaceutics.<br />

Date: M. Najmuddin<br />

M. Pharm (Ph.D.)<br />

Place: Gulbarga Associate Professor<br />

Luqman College of Pharmacy,<br />

Gulbarga.5851012<br />

iii


RAJIV G<strong>AND</strong>HI UNIVERSITY <strong>OF</strong> HEALTH SCIENCES,<br />

KARNATAKA, BANGALORE<br />

CERTIFICATE BY THE CO-GUIDE<br />

This is to certify that the dissertation entitled “Development<br />

and Evaluation of Pulsatile Drug Delivery System of<br />

Flurbiprofen” is a bonafide research work done by Mr. Patel Vishal<br />

Ashok in partial fulfillment of the requirement for the degree of<br />

“Master of Pharmacy” in Pharmaceutics.<br />

Date: Mr. Aejaz Ahmed<br />

M. Pharm<br />

Place: Gulbarga Lecturer<br />

Luqman College of Pharmacy,<br />

Gulbarga.5851012<br />

iv


RAJIV G<strong>AND</strong>HI UNIVERSITY <strong>OF</strong> HEALTH SCIENCES,<br />

KARNATAKA, BANGALORE<br />

ENDORSEMENT BY THE HOD, PRINCIPAL/<br />

HEAD <strong>OF</strong> THE INSTITUTION<br />

This is to certify that the dissertation entitled “Development<br />

and Evaluation of Pulsatile Drug Delivery System of<br />

Flurbiprofen” is a bonafide research work done by Mr. Patel Vishal<br />

Ashok under the guidance of Mr. M. Najmuddin, Research guide<br />

Luqman College of pharmacy Gulbarga.<br />

Date: Prof. Syed. Sanaullah<br />

Place: GULBARGA M. Pharm (PhD.)<br />

Luqman College of Pharmacy,<br />

Gulbarga. 585102<br />

v


RAJIV G<strong>AND</strong>HI UNIVERSITY <strong>OF</strong> HEALTH SCIENCES,<br />

KARNATAKA, BANGALORE<br />

COPYRIGHT<br />

DECLARATION BY THE C<strong>AND</strong>IDATE<br />

I hereby declare that the Rajiv Gandhi University of Health Sciences,<br />

Karnataka shall have the right to preserve, use, and disseminate this<br />

dissertation/ thesis in print or electronic format for academic/<br />

research purpose.<br />

Date:<br />

Place: GULBARGA Patel Vishal Ashok<br />

© Rajiv Gandhi University of Health Sciences, Karnataka<br />

vi


No work big or small can fructify without the grace and mercy of Almighty<br />

“JAI SAI NATH”, “The Honoror”. The most essentially humble solicitation and<br />

a lot of thanks to the Supreme Power for manifesting himself through the various<br />

helpful people I came across in my life. I bow my head to Him and ask for His<br />

blessings to be with me forever.<br />

I consider myself most lucky to work under the guidance of<br />

Mr. M.Najmuddin, Associate Professor, Luqman College of Pharmacy,<br />

Gulbarga. I take this opportunity to express my heartfelt gratitude to my reverend<br />

guide. His discipline, principles, simplicity, caring attitude and provision of<br />

fearless work environment will be cherished in all walks of my life. I am very<br />

much grateful to him for his invaluable guidance and ever-lasting encouragement<br />

throughout my course.<br />

Very special thanks to Dr. M.G Purohit, for their constant support in<br />

analytical work.<br />

I am immensely thankful to Dr. Syed Sanaullah, Principal,Luqman College of<br />

pharmacy, Dr. Mujeeb, Treasurer, Luqman college of pharmacy, Gulbarga.<br />

I owe my warmest and humble thanks to Dr. N.G. Raghvendra Rao, Dr.<br />

S.S.Bushetti, Mrs. Syeda Humaira, Mr. M. A. Saleem, Mr. Mohan Joshi,<br />

Prashant sir, Mr.Raghunandan Deshpande, Mr.Mahesh Bedre, Mr. Aejaz<br />

Ahmed, Mr. Sunil Firangi and other staff members of Luqman College of<br />

Pharmacy, Gulbarga, for their timely help, encouragement, boosting my<br />

confidence in the progress of my academics.<br />

I express my deepest and very special thanks to my batch mates Ram,<br />

Ganesh, Sachin, Ketan, Aniket, Vijay, Tousif, Azuruddin, Shahid, for their kind<br />

co-operation, help and encouragement throughout my course.<br />

I convey my thanks and well wishes to all my juniors Sudhir, Shrishail,<br />

Anaf, Vishal, Dinesh, Subhan, Izhar, Vahid, Mangesh, Hadi Jaspal, Jinesh and<br />

others who have contributed directly or indirectly during my dissertation<br />

I extended my special thanks to my dear friends & Brothers Ishwer,<br />

Nandakishor, Praful, Manoj, Anwar, Sunil, Amol Basgunde, Goutam, Rohit,<br />

Sachin, Rohit.<br />

vii


I render my grateful respect and sincere thanks to my beloved Parents. I<br />

would like to extend my gratitude to my ‘Jijaji’ Mr. Shailendra Patil and my<br />

sister Mrs. Deepu, Ms. Banti without whom it could not have been possible for<br />

me to complete this project work.<br />

I express my sincere thankful to non teaching staff Narendra, Peer Pasha,<br />

Suresh and Librarian Ms. Pratibha of Luqman college of pharmacy, Gulbarga,<br />

for their co-operation.<br />

Last but not the least, I would like to thank SUPER Computers, Gulbarga<br />

for making this thesis work in a reproducible manner.<br />

Date:<br />

Thankful I ever remain……………<br />

Place: Gulbarga. VISHAL ASHOK PATEL<br />

viii


ix


Abs Absorbance<br />

ºC Temperature on Celsius Scale<br />

CAP Cellulose Acetate phthalate<br />

cm Centimetre<br />

cm 2<br />

Centimetre Square<br />

cps Centipoises<br />

EC Ethyl Cellulose<br />

o<br />

F Temperature on Faranide Scale<br />

FDA Food and Drug Administration<br />

gms Grams<br />

HPMC Hydroxypropyl methyl cellulose<br />

Hrs Hours<br />

IP Indian Pharmacopoeia<br />

IR Infrared<br />

kg/cm 2<br />

Kilogram per centimetre square<br />

L.R. Laboratory Grade<br />

mg Milligram<br />

g/mcg Microgram<br />

min Minutes<br />

mm Millimetre<br />

NASID Non-steroidal anti-inflammatory drug<br />

nm Nanometre<br />

P.G. Pharmaceutical Grade<br />

‘r’ Regression Coefficient<br />

rpm Resolutions per minute<br />

SEM Scanning Electron Microscopy<br />

SD Standard Deviation<br />

Sod. Alg. Sodium Alginate<br />

UV Ultraviolet<br />

v/v Volume/Volume<br />

w/w Weight/Weight<br />

% w/v Percentage weight by volume<br />

% w/w Percentage weight by weight<br />

x


FM-1 Flurbiprofen: Eudragit L-100: Eudragit S-100<br />

FM-2 Flurbiprofen: Eudragit L-100: Eudragit S-100<br />

FM-3 Flurbiprofen: Eudragit L-100: Eudragit S-100<br />

FM-4 Flurbiprofen: Eudragit L-100: Eudragit S-100<br />

F-1 Flurbiprofen: Eudragit L-100: Eudragit S-100: Guar gum (20mg)<br />

F-2 Flurbiprofen: Eudragit L-100: Eudragit S-100: Guar gum (30mg)<br />

F-3 Flurbiprofen: Eudragit L-100: Eudragit S-100: Guar gum (40mg)<br />

F-4 Flurbiprofen: Eudragit L-100: Eudragit S-100: HPMC (20mg)<br />

F-5 Flurbiprofen: Eudragit L-100: Eudragit S-100: HPMC (30mg)<br />

F-6 Flurbiprofen: Eudragit L-100: Eudragit S-100: HPMC (40mg)<br />

F-7 Flurbiprofen: Eudragit L-100: Eudragit S-100: Sodium alginate (20mg)<br />

F-8 Flurbiprofen: Eudragit L-100: Eudragit S-100: Sodium alginate (30mg)<br />

F-9 Flurbiprofen: Eudragit L-100: Eudragit S-100: Sodium alginate (30mg)<br />

xi


In this study, investigation of an oral colon specific, pulsatile device to achieve<br />

time and/or site specific release of Flurbiprofen, based on chronopharmaceutical<br />

consideration. The basic design consists of an insoluble hard gelatin capsule body,<br />

filled with eudragit microcapsules of Flurbiprofen and sealed with a hydrogel plug.<br />

The entire device was enteric coated, so that the variability in gastric emptying time<br />

can be overcome and a colon-specific release can be achieved. The Flurbiprofen<br />

microcapsules were prepared by solvent evaporation method with Eudragit L-100 and<br />

S-100 (1:2) by varying drug to polymer ratio and evaluated for the particle size, angle<br />

of repose, percentage yield, drug content, SEM, IR and in-vitro release study. The<br />

drug content in the range of 75.9 ± 0.78 to 95.59 ± 0.68. The in-vitro, drug release<br />

studies were carried out using pH 6.8 phosphate buffer for 12 hrs. At the end of 12 th<br />

hrs the drug release in the range of 88.51 ± 1.53 to 95.58 ± 0.24 and from the obtained<br />

results; FM-3 was selected as an optimized formulation for designing pulsatile device.<br />

Different hydrogel polymers (Guar gum, HPMC, Sodium alginate) were used as plugs<br />

in different ratios, to maintain a suitable lag period. The entire device was coated with<br />

5% CAP. The formulated pulsatile device was evaluated weight variation, thickness<br />

of CAP, IR, and in-vitro release study. The in-vitro release study were carried out<br />

using pH 1.2 buffer for a period of 2 hrs then 7.4pH phosphate buffer for a period of<br />

3hrs then 6.8 pH phosphate buffer for a period of 10 hrs. At the end of 15 th hrs the<br />

drug releases were in the ranges of 62.28 ± 0.479 to 74.61 ± 0.408, 59.79 ± 0.363 to<br />

76.46 ± 0.802 and 70.25 ± 0.155 to 82.95 ± 0.239 with Guar gum, HPMC and Sodium<br />

alginate plugs respectively. From obtained results, it was found that the order of<br />

sustaining capacity of pulsatile device is, HPMC > Guar gum > Sodium alginate.<br />

Keywords: Pulsatile; Colon-specific device; Chronotherapeutics; Rheumatoid<br />

arthritis; Eudragit microcapsules<br />

xii


CHAPTER<br />

NO.<br />

<br />

CONTENTS<br />

PAGE<br />

NO.<br />

CHAPTER– 1 INTRODUCTION 01–32<br />

CHAPTER –2 OBJECTIVES 33–36<br />

CHAPTER – 3 REVIEW <strong>OF</strong> LITERATURE 37–66<br />

CHAPTER – 4 METHODOLOGY 67–82<br />

CHAPTER – 5 RESULTS 83–117<br />

CHAPTER – 6 DISCUSSION 118–125<br />

CHAPTER – 7 CONCLUSIONS 126–127<br />

CHAPTER – 8 SUMMARY 128–130<br />

CHAPTER – 9 BIBLIOGRAPHY 131–141<br />

ANNEXURES 142 – 147<br />

xiii


Sl.<br />

No.<br />

<br />

Title<br />

1. Circadian rhythm and manifestation of clinical diseases 05<br />

2. Average pH in the GI tract 26<br />

3. Average GI transit time 26<br />

4. Summary of colon-specific drug delivery strategies 30<br />

5. Materials / Chemicals used 67<br />

6. Equipments used and source 68<br />

7. Formulation of Flurbiprofen Microcapsules using Eudragit – L 100<br />

and Eudragit – S100<br />

8. Composition for modified pulsatile device on the basis of design<br />

summary<br />

9. Standard calibration data of Flurbiprofen in Ethanol 83<br />

10. Standard calibration data of Flurbiprofen in pH 1.2 buffer 84<br />

11. Standard calibration data of Flurbiprofen in pH 6.8 buffer 85<br />

12. Standard calibration data of Flurbiprofen in pH 7.4 buffer 86<br />

13. Micromeritic properties of Flurbiprofen Microcapsules 88<br />

14. Percentage yield and Drug content of Flurbiprofen microcapsules 88<br />

15. In-vitro release profile of Flurbiprofen microcapsules for FM-1 92<br />

16. In-vitro release profile of Flurbiprofen microcapsules for FM-2 93<br />

17. In-vitro release profile of Flurbiprofen microcapsules for FM-3 94<br />

18. In-vitro release profile of Flurbiprofen microcapsules for FM-4 95<br />

19. Kinetic values obtained from in-vitro release profile for<br />

microcapsules<br />

Page<br />

No.<br />

20. Coating thickness 100<br />

72<br />

80<br />

96<br />

xiv


Sl.<br />

No.<br />

Title<br />

Page<br />

No.<br />

21. In-vitro release rate profile of F1 containing 20mg Guar Gum 102<br />

22. In-vitro release rate profile of F2 containing 30mg Guar Gum 103<br />

23. In-vitro release rate profile of F3 containing 40mg Guar Gum 104<br />

24. In-vitro release rate profile of F4 containing 20mg HPMC 105<br />

25. In-vitro release rate profile of F5 containing 30mg HPMC 106<br />

26. In-vitro release rate profile of F6 containing 40mg HPMC 107<br />

27. In-vitro release rate profile of F7 containing 20mg Sodium Alginate 108<br />

28. In-vitro release rate profile of F8 containing 30mg Sodium Alginate 109<br />

29. In-vitro release rate profile of F9 containing 40mg Sodium Alginate 110<br />

30. Kinetic values obtained from in-vitro release profile for<br />

microcapsules<br />

111<br />

xv


Sl.<br />

No.<br />

<br />

Title<br />

1 A 24-hours clock diagram of the peak time of selected human<br />

circadian rhythms with reference to the day-night cycle.<br />

2 Drug release profile of pulsatile drug delivery systems 20<br />

3 Different stages in drug release from pulsincap 22<br />

4 Structure of colon 25<br />

5 Classification of microparticles 32<br />

6 Overview of designed pulsatile device 79<br />

7 Standard calibration curve of Flurbiprofen in Ethanol 83<br />

8 Standard calibration curve of Flurbiprofen in pH 1.2 buffer 84<br />

9 Standard calibration curve of Flurbiprofen in pH 6.8 buffer 85<br />

10 Standard calibration curve of Flurbiprofen in pH 7.4 buffer 86<br />

11 Images of Flurbiprofen microcapsules for FM-1, FM-2, FM-3, FM-4 87<br />

12 Scanning electron microphotographs of FM-3 formulation 89<br />

13 I.R. Spectrum of Flurbiprofen (pure drug) 90<br />

14 I.R. Spectrum of Eudragit L-100 90<br />

15 I.R. Spectrum of Eudragit S-100 91<br />

16 I.R. Spectrum of Flurbiprofen microcapsules (FM-3) 91<br />

17 Comparative zero order plots for Flurbiprofen microcapsules 97<br />

18 Comparative first order plots for Flurbiprofen microcapsules 97<br />

19. Comparative Higuchi matrix model for Flurbiprofen microcapsules 98<br />

20. Comparative Peppas model for Flurbiprofen microcapsules 98<br />

Page<br />

No.<br />

21. I.R. Spectrum of Flurbiprofen + Sodium Alginate 101<br />

04<br />

xvi


Sl.<br />

No.<br />

Title<br />

Page<br />

No.<br />

22. I.R. Spectrum of Flurbiprofen + HPMC 101<br />

23. I.R. Spectrum of Flurbiprofen + Guar gum 101<br />

24. Comparative zero order plots of formulation containing Guar Gum as<br />

hydrogel plug<br />

25. Comparative first order plots of formulation containing Guar Gum as<br />

hydrogel plug<br />

26. Comparative Higuchi matrix model of formulation containing Guar<br />

Gum as hydrogel plug<br />

27. Comparative Peppas model of formulation containing Guar Gum as<br />

hydrogel plug<br />

28. Comparative zero order plots of formulation containing HPMC as<br />

hydrogel plug<br />

29. Comparative first order plots of formulation containing HPMC as<br />

hydrogel plug<br />

30. Comparative Higuchi matrix model of formulation containing HPMC<br />

as hydrogel plug<br />

31. Comparative Peppas model of formulation containing HPMC as<br />

hydrogel plug<br />

32. Comparative zero order plots of formulation containing Sodium<br />

Alginate as hydrogel plug<br />

33. Comparative first order plots of formulation containing Sodium<br />

Alginate as hydrogel plug<br />

34. Comparative Higuchi matrix model of formulation containing<br />

Sodium Alginate as hydrogel plug<br />

35 Comparative Peppas model of formulation containing Sodium<br />

Alginate as hydrogel plug<br />

112<br />

112<br />

113<br />

113<br />

114<br />

114<br />

115<br />

115<br />

116<br />

116<br />

117<br />

117<br />

xvii


CHAPTER-1<br />

<br />

Introduction<br />

Controlled drug delivery systems have acquired a centre stage in the area<br />

of pharmaceutical R &D sector. Such systems offer temporal &/or spatial control<br />

over the release of drug and grant a new lease of life to a drug molecule in terms<br />

of controlled drug delivery systems for obvious advantages of oral route of drug<br />

administration. These dosage forms offer many advantages, such as nearly<br />

constant drug level at the site of action, prevention of peak-valley fluctuation,<br />

reduction in dose of drug, reduced dosage frequency, avoidance of side effects and<br />

improved patient compliance. In such systems the drug release commences as<br />

soon as the dosage form is administered as in the case of conventional dosage<br />

forms. However, there are certain conditions, which demand release of drug after<br />

a lag time. Such a release pattern is known as “pulsatile release”. 1<br />

Traditionally, drug delivery has meant getting a simple chemical absorbed<br />

predictably from the gut or from site of injection. A second-generation drug<br />

delivery goal has been the perfection of continuous, constant rate (zero order)<br />

delivery of bioactive agents. However, living organisms are not “zero order” in<br />

their requirement or response to drugs. They are predictable resonating dynamic<br />

systems, which require different amounts of drug at predictably different times<br />

within the circadian cycle in order to maximize desired and minimize undesired<br />

drug effects. Due to advances in chronobiology, chronopharmacology and global<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

1


Introduction<br />

market constraints, the traditional goal of pharmaceutics (e.g. design drug delivery<br />

system with a constant drug release rate) is becoming obsolete. However, the<br />

major bottleneck in the development of drug delivery systems that match<br />

circadian rhythms (chronopharmaceutical drug delivery system: ChrDDS) may be<br />

the availability of appropriate technology. The diseases currently targeted for<br />

chronopharmaceutical formulations are those for which there are enough scientific<br />

backgrounds to justify ChrDDS compared to the conventional drug administration<br />

approach. These include asthma, arthritis, duodenal ulcer, cancer, diabetes,<br />

cardiovascular diseases, hypercholesterolemia, ulcer and neurological diseases. 2<br />

If the organization in time of living system including man is borne in<br />

mind, it is easy to conceive that not only must the right amount of the right<br />

substance be at right place but also this must occur at the right time. In the last<br />

decade numerous studies in animals as well as clinical studies have provided<br />

convicing evidence, that the pharmacokinetics &/or the drug’s effects-side effects<br />

can be modified by the circadian time &/or the timing of drug application within<br />

24 hrs of a day. 3<br />

Circadian variation in pain, stiffenss and manual and manual deyterity in<br />

patients with osteo and rheumatoid arthritis have been studied and has implication<br />

for timing antirheumatide drug treatment. 4 Morning stiffness associated with pain<br />

at the time of awakening is a diagnostic criterion of the rheumatoid arthritis and<br />

these clinical circadian symptoms are supposed to be outcome of altered<br />

functioning of hypothalamic–pitutary–adrenocortical axis.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

2


Introduction<br />

Chronopharmacotherapy for rheumatoid arthritis has been recommended<br />

to ensure that the highest blood levels of the drug coincide with peak pain and<br />

stiffness. 5 A pulsatile drug delivery system that can be administered at night<br />

(before sleep) but that release drug in early morning would be a promising<br />

chronopharmaceutic system. 5,6<br />

Drug targeting to colon would prove useful where intentional delayed drug<br />

absorption is desired from therapeutic point of view in the treatment of disease<br />

that have peak symptoms in the early morning such as nocturnal asthma, angina,<br />

arthritis. 1,4,7,8,<br />

Some orally administered drugs (E.g. Diclofenac, Theophyllin, Ibuprofen<br />

Isosorbide) may exhibit poor uptake in the upper regions of GIT or degrade in the<br />

presence of GIT enzymes. Better bioavailability can be achieved through colon-<br />

specific drug delivery. Colonic targeting is also advantageous where delay in<br />

systemic absorption is therapeutically desirable. 4,7<br />

1.1 Circadian rhythms and their implications:<br />

Circadian rhythms are self-sustaining, endogenous oscillation, exhibiting<br />

periodicities of about one day or 24 hours. Normally, circadian rhythms are<br />

synchronized according to the body ’ s pacemaker clock, located in the<br />

suprachiasmic nucleus of the hypothalamus.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

3


Introduction<br />

The physiology and biochemistry of human being is not constant during<br />

the 24 hours, but variable in a predictable manner as defined by the timing of the<br />

peak and through of each of the body’s circadian processes and functions. The<br />

peak in the rhythms of basal gastric and secretion, white blood cells (WBC),<br />

lymphocytes, prolactin, melatonin, eosinophils, adrenal corticotrophic hormone<br />

(ACTH), follicle stimulating hormone (FSH), and leuteinizing hormone (LH), is<br />

manifested at specific times during the nocturnal sleep span. The peak in serum<br />

cortisol, aldosterone, testosterone plus platelet adhesiveness and blood viscosity<br />

follows later during the initial hours of diurnal activity. Hematocrit is the greatest<br />

and airway caliber the best around the middle and afternoon hours, platelet<br />

numbers and uric acid peak later during the day and evening. Hence, several<br />

physiological processes in humans vary in a rhythmic manner, in synchrony with<br />

the internal biological clock, as shown in fig.1. 4,9<br />

Fig.1: A 24-hours clock diagram of the peak time of selected human circadian<br />

rhythms with reference to the day-night cycle.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

4


Introduction<br />

Through a number of clinical trials and epidemiological studies, it has<br />

become evident that the levels of disease activity of number of clinical disorders<br />

have a pattern associated with the body’s inherent clock set according to circadian<br />

rhythms. Infect just as the time of day influences normal biologic processes, so it<br />

affects the pathophysiology of disease and its treatment. Examples of some of the<br />

diseases are shown in Table 1. 4<br />

Table 1: Circadian rhythm and manifestation of clinical diseases<br />

Disease or syndrome Circadian rhythmicity<br />

Allergic Rhinitis Worse in the morning/upon rising<br />

Asthma Exacerbation more common during the sleep period<br />

Rheumatoid Arthritis Symptoms more common during the sleep period<br />

Osteoarthritis Symptoms worse in the middle/later portion of the day<br />

Angina Pectoris Chest pain and ECG changes more common in early<br />

morning<br />

Myocardial Infraction Incidence greatest in early morning<br />

Stroke Incidence higher in the morning<br />

Sudden cardiac death Incidence higher in the morning after awakening<br />

Peptic ulcer disease Worse in late evening and early morning hours<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

5


Introduction<br />

1.2 Chronotherapeutic: Therapy in synchrony with biorhythms<br />

Chronotherapy coordinates drug delivery with human biological rhythms<br />

and holds huge promise in areas of pain management and treatment of asthma,<br />

heart disease and cancer. The coordination of medical treatment and drug delivery<br />

with such biological clocks and rhythms is termed chronotherapy. 10<br />

Chronotherapeutics, or delivery of medication in concentrations that vary<br />

according to physiological need at different times during the dosing period, is a<br />

relatively new practice in clinical medicine and thus many physicians are<br />

unfamiliar with this intriguing area of medicine. It is important that physicians<br />

understand the advantages of chronotherapy so that they can make well-informed<br />

decisions on which therapeutic strategies are best for their patients-traditional ones<br />

or chronotherapies.<br />

The goal of chronotherapeutics is to synchronize the timing of treatment<br />

with the intrinsic timing of illness. Theoretically, optimum therapy is more likely<br />

to result when the right amount of drug is delivered to the correct target organ at<br />

the most appropriate time. In contrast, many side effects can be minimized if a<br />

drug is not given when it is not needed. Unlike homeostatic formulations, which<br />

provide relatively constant plasma drug levels over 24 hours,, chronotherapeutic<br />

formulations may use various release mechanisms. e.g., time-delay coatings<br />

(Covera-HS TM ), osmotic pump mechanisms (COER -24 TM ), and matrix systems<br />

(Geminex TM), that provide for varying levels throughout the day.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

6


Introduction<br />

A major objective of chronotherapy in the treatment of several diseases is<br />

to deliver the drug in higher concentrations during the time of greatest need<br />

according to the circadian onset of the disease or syndrome. The chronotherapy of<br />

a medication may be accomplished by the judicious timing of conventionally<br />

formulated tablets and capsules. In most cases, however, special drug delivery<br />

technology must be relied upon to synchronize drug concentrations to rhythms in<br />

disease activity. 4<br />

1.3 Arthritis<br />

11, 12, 13<br />

The term arthritis is used to describe changes in the joints which may be<br />

either inflammatory or degenerative in character. If only one joint is affected the<br />

condition is referred to as monoarticular arthritis; if several joints are involved<br />

it is called polyarticular arthritis or polyarthritis (Greek: poly= many).<br />

It is imperative for all adults to have at least a basic understanding of<br />

arthritis since most adults face some form of Arthritis. It is an extremely common<br />

and is among the oldest known disease among men and women. Only in recent<br />

years however, has the magnitude of the health problems associated with Arthritis<br />

been appreciated in the United States and throughout the world. As a result, a<br />

great deal of new knowledge, understanding, and treatment is available today.<br />

Medical scientists have discovered new leads in search for the causes of various<br />

forms of arthritis. Though cure is not yet available, treatment is generally<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

7


Introduction<br />

satisfactory, especially if started within the first six months after onset.<br />

Fortunately, very few people today need to be crippled by arthritis.<br />

A Joint consists of the ends of the articulating bones which are covered<br />

with cartilage. It is surrounded and kept in position by a capsule and special<br />

ligaments which are lined by a membrane called the synovial membrane.<br />

Symptoms of arthritis:<br />

The joints ache and swell<br />

Pain and muscle-spasm are common features; and in the late stages very<br />

severe pain and gross destruction and deformity of the joints may develop.<br />

In children the condition tends to develop suddenly, many joints being<br />

affected from the beginning the small joint soft the hands and feet being as<br />

a rule affected first.<br />

Severe muscle wasting might also take place<br />

Patients of arthritis can only walk a certain distance after which they tend<br />

to feel pain and stiffness in their joints.<br />

Arthritis can also develop slowly starting as a slight restriction of<br />

movement in certain directions, with stiffness first thing in the morning<br />

and aches after exercises.<br />

Arthritis can be both of the infective variety as well as the chronic.<br />

In infective arthritis the patient feels ill and toxic with a swinging<br />

temperature and a furred tongue. Leucocytes are always evident, and a<br />

blood culture may confirm the presence of the septicaemia.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

8


Some common forms of Arthritis:<br />

Rheumatic arthritis(fibromyalgia)<br />

Rheumatoid arthritis(Still's Disease)<br />

Degenerative, e.g. osteo-arthritis<br />

Psoriatic arthritis<br />

Ankylosing Spondylitis<br />

Introduction<br />

The above mentioned are all different form of arthritis and they all have<br />

different cures and therapies as well as symptoms. The common strand binding<br />

them is that they are all marked by severe pain in the joints.<br />

1.3.1 Rheumatoid Arthritis:<br />

Rheumatoid arthritis is a form of chronic arthritis. This disease affects<br />

chiefly young adults, mainly women, and one or many joints may be involved; it<br />

also occurs in children (Still's Disease). It is generalized affection of joints, and<br />

their synovial membranes, cartilages, capsules and the muscles supplying them;<br />

but other connective tissues elsewhere in the body might also be affected.<br />

Rheumatoid arthritis is now a major cause of crippling in European countries, but<br />

it is not common in the tropics.<br />

Causes of Rheumatoid Arthritis:<br />

The causes of the conditions are not yet fully understood. While infection<br />

form the focus of the respiratory, alimentary, urinary or genital tracts is sometimes<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

9


Introduction<br />

a factor, recent research has also demonstrated the importance of the endocrine<br />

glands as both as a cause and a therapy.<br />

1.3.2 Osteoarthritis<br />

This is a chronic and mild form of arthritis which is distinguished by<br />

degenerative changes affecting primarily the articular cartilage and adjacent bone<br />

which can usually be seen on x-ray. Sometime, only one joint is affected, and it is<br />

usually a large one such as the hip. Osteoarthritis is extremely common and<br />

usually constitutes a little more than a considerable and continuing nuisance. The<br />

chance of getting it increases with age. The weight-bearing joints are more<br />

commonly involved, but one hereditary form of osteoarthritis involves the end and<br />

middle joints of fingers.<br />

Causes of osteoarthritis:<br />

This disease is caused due to a gradual destruction of cartilage. Its precise<br />

cause is unknown but it is no longer known as a simple wear and tear<br />

disease.<br />

Hereditary seems to play a very important role in osteoarthritis.<br />

How can an osteoarthritis patient be treated?<br />

The treatment of osteoarthritis is simpler than that of the more severe<br />

forms of arthritis.<br />

The step in treatment is to assure the patient that it is not crippling,<br />

deforming disease.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

10


Introduction<br />

This is often a great relief to the patient since the patients usually think that<br />

they have a destructive form of arthritis. They often harbours more fear<br />

about their future regarding the course the disease would take in their life<br />

more than the disease warrants.<br />

Joint rest is important and special exercises are needed.<br />

Postural exercises are important aids in treatment.<br />

The body weight should be kept down<br />

Drug therapy is kept to a minimum. The principal drug is Aspirin and<br />

Paracetamol; the dose is to be decided by the doctor.<br />

Walking shoes with raised heel often help. The patient should also use a<br />

walking stick.<br />

Among the various surgeries available for arthritis, Osteotomy is the most<br />

successful.<br />

1.3.3 Fibromyalgia<br />

Fibromyalgia is a common condition. However it can get severe enough to<br />

start intruding into your day to day life. Fibromyalgia literally means, fibrous<br />

tissues (fibro -) and the muscles ( -my-) being affected by pain ( -algia). In this<br />

disease the whole body feels affected since the tendons and the ligaments both get<br />

affected. Fibromyalgia in fact affects the muscles and not the joints at all. Also,<br />

this form of arthritis never causes permanent damage to tissues though the<br />

symptoms may last for months or years.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

11


Introduction<br />

Fibromyalgia does not have any outward manifestation; therefore others<br />

might not understand your physical pain and tiredness. In the past fibromyalgia,<br />

was diagnosed as a form of rheumatism or generally also wrongly diagnosed as<br />

being a degenerative joint disease. It is only recently that we have a clearer picture<br />

of fibromyalgia. Subsequently medical practitioners have also streamlined various<br />

symptoms, signs and therapies specially suited to fibromyalgia. Tennis elbow is a<br />

localized condition of this same disease. Fibromyalgia however can lead to<br />

tenderness in various points of the body.<br />

Causes fibromyalgia:<br />

Fibromyalgia is a functional disturbance which implies that its causes<br />

cannot purely be categorized as being physical or mental.<br />

Both the person's mind as well as body is involved in this ailment.<br />

Sleep being vital to both the good health of the body as well as the mind, it<br />

comes as no surprise that a major cause of fibromyalgia is sleep disorders.<br />

If a person has more light sleep than deep sleep then he is likely to be<br />

inflicted by Fibromyalgia.<br />

Any stress that the person is feeling might interfere with his/her deep or<br />

restorative sleep and therefore end up in fibromyalgia.<br />

Sleeping in uncomfortable positions and places could also be a leading<br />

cause of this disease.<br />

Once fibromyalgia sets in, it becomes a vicious cycle; since the person now<br />

already afflicted by pain loses sleep as a result of which his/her condition<br />

automatically intensifies the condition.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

12


1.3.4 Ankylosing Spondylitis<br />

Introduction<br />

This is a chronic inflammatory form of arthritis which affects the spinal<br />

joints. The main and the most important feature is that in this form of arthritis the<br />

joints between the spine and the pelvis get inflicted. This form of arthritis causes<br />

back ache and stiffness in the back as well as a hunched up body posture. In very<br />

severe cases there might be inflammations around the tendons and ligaments that<br />

connect and provide support to joints that lead to pain and tenderness in the<br />

shoulder blades and the spinal cord. Ankylosing Spondylitis can also impair<br />

mobility by creating inflammation of the vertebra.<br />

This ailment can have confusing symptoms since there can be varied kinds<br />

of manifestations. While some individuals suffer gouts of transient back ache only<br />

while others suffer chronic back aches that lead to differing degrees of back ache<br />

and stiffness.<br />

Colloquially, Ankylosing Spondylitis is referred to as poker back and<br />

rheumatoid Spondylitis. It is only about five decades back that this ailment got the<br />

name that is now prevalent. Since this ailment belongs to the family of diseases<br />

that attack the spine it is also referred to as, in medical jargon as<br />

spondylarthropathies.<br />

You must keep in mind that men are three times more at a risk of acquiring<br />

this disease than women. This ailment also attacks youngster’s more than older<br />

people, contrary to popular belief.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

13


Causes Ankylosing Spondylitis:<br />

Introduction<br />

Ankylosing spondylitis is believed to be hereditary since it depends on<br />

tissue type and we genetically inherit tissue types.<br />

The exact cause of this ailment is however still not known.<br />

Some researches also believe that environmental interaction with certain<br />

tissue types might result in Ankylosing spondylitis.<br />

What are the signs and symptoms of Ankylosing spondylitis?<br />

Recurring inflammation in the eyes which cause pain, redness, blurred<br />

vision and sensitivity to bright light. People also experience inflammation<br />

of the joints.<br />

Chronic back ache that lasts for years<br />

Pain and tenderness in the ribs, shoulder blades, hips, thighs and along the<br />

shins.<br />

Pronounced back pain<br />

Youngsters who are afflicted will experience pain in the heels and waist.<br />

1.3.5 Psoriatic arthritis<br />

Psotiaric arthritis is a less common form of arthritis which affects, men<br />

and women in an equal ratio, and usually strikes when they are between the ages<br />

of 20 to 50. It is marked by scaly growth of rough tissues around the joints. There<br />

are several types of psoriatic arthritis, with symptoms that range from mild to<br />

severe. In general, the disease isn't as crippling as other forms of arthritis, but if<br />

left untreated it can cause discomfort, disability and deformity. Although no cure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

14


Introduction<br />

exists for psoriatic arthritis, medication, physical therapy and lifestyle changes<br />

often can relieve pain and slow the progression of joint damage. Psotiaric arthritis<br />

causes swelling in the joints. It affects a number of joints including the fingers,<br />

wrists, toes, knees, ankles, elbows and shoulder joints, the spine and joints in the<br />

lower back (called sacroiliac joints). Pso riatic arthritis also affects tissues<br />

surrounding the joints including tendons and ligaments and causes inflammation<br />

and swelling and pain in and around the joints. This ailment usually affects the<br />

wrists, knees, ankles, fingers and toes. It also affects the back.<br />

One of these conditions is psoriatic arthritis, which may affect as many as<br />

1 million of the approximately 6 million Americans who have psoriasis. In fact<br />

30% of the people who have psoriasis later go onto have psoriatic arthritis.<br />

Causes of Psoriatic arthritis:<br />

The exact cause of psoriatic arthritis is unknown<br />

As mentioned before psoriasis generally develops into psoriatic arthritis<br />

It has been observed that people who have psoriatic arthritis patients in the<br />

house generally get inflicted by this ailment.<br />

Exposure to infection, stress, alcohol, poor nutrition<br />

Reaction to a medication or vaccine<br />

Overexposure to the sun or prolonged exposure to irritating chemical such<br />

as disinfectants and pain thinners.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

15


1.4 Chronopharmaceutics: 2<br />

Introduction<br />

Chronopharmaceutics is a branch of pharmaceutics devoted to design and<br />

evaluation of drug delivery system that release a bioactive agent at a rhythm that<br />

ideally matches the biological requirement of a given disease therapy. Ideally<br />

chronopharmaceutical drug delivery system (ChrDDS) should embody time -<br />

controlled and site specific drug delivery system. Evidence suggests that an ideal<br />

ChrDDS should:<br />

Be non-toxic within approved limits of use,<br />

Have a real-time and specific triggering biomarker for a given disease<br />

state.<br />

Have a feed-back control system (ex: self -regulated and adaptive<br />

capability to circadian rhythm and individual patient to differentiate<br />

between awake-sleep status),<br />

Be biocompatible and biodegradable, especially for parentral<br />

administration,<br />

Be easy to manufacture at economic cost and<br />

Be easy to administer to patients and enhances compliance to dosage<br />

regimen.<br />

When treating human diseases, the overall goal is to cure or manage the<br />

disease while minimizing the negative impact of side effects associated with<br />

therapy. In this respect, chronopharmaceutics will be a clinically relevant and<br />

reliable discipline if pharmaceutical scientists could delineate a formal and<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

16


Introduction<br />

systemic approach to design and evaluate drug delivery system that matches the<br />

biological requirement.<br />

1.5 Pulsatile drug delivery systems<br />

1.5.1 New global trends in drug discovery and development<br />

In this century, the pharmaceutical industry is caught between pressure to<br />

keep prices down and the increasing cost of successful drug discovery and<br />

development. The average cost and time for the development of a new chemical<br />

entity are much higher (app $500 million and 10-12 years) than those required to<br />

develop a novel drug delivery system (NDDS or ChrDSS) ($20-$50 million and 3<br />

to 4 years). In the form of an NDDS or ChrDDs, an existing drug molecule can<br />

“get a new life” thereby increasing its market value and competitiveness and<br />

extending patent life. 2<br />

Among modified-release oral dosage forms, increasing interest has<br />

currently turned to systems designed to achieve time specific (delayed, pulsatile)<br />

and site-specific delivery of drugs. In particular, systems for delayed release are<br />

meant to deliver the active principle after a programmed time period following<br />

administration. These systems constitute a relatively new class of device the<br />

importance of which is especially connected with the recent advances in<br />

chronopharmacology. It is by now well-known that the symptomatology of a large<br />

number of pathologies as well as the pharmacokinetics and pharmacodynamics of<br />

several drugs follow temporal rhythms, often resulting in circadian variations.<br />

Therefore, the possibility of exploiting delayed release to perform chronotherapy<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

17


Introduction<br />

is quite appealing for those diseases, the symptoms of which recur mainly at night<br />

time or in the early morning, such as bronchial asthma, angina pectoris and<br />

rheumatoid arthritis. The delay in the onset of release has so far mainly been<br />

achieved through osmotic mechanisms, hydrophilic or hydrophobic layers, coating<br />

a drug-loaded core and swellable or erodible plugs sealing a drug containing<br />

insoluble capsule body. 14<br />

Delivery systems with a pulsatile release pattern are receiving increasing<br />

interest for the development of dosage forms, because conventional systems with<br />

a continuous release are not ideal. Most conventional oral controlled release drug<br />

delivery systems release the drug with constant or variable release rates. A<br />

pulsatile release profile is characterized by a time period of no release rates (lag<br />

time) followed by a rapid and complete release. These dosage forms offer many<br />

advantages such as<br />

Nearly constant drug levels at the site of action.<br />

Avoidance of undesirable side effects.<br />

Reduced dose and<br />

Improved patient compliance.<br />

Used for drugs with chronopharmacological behaviour, a high first pass<br />

effect, the requirement of night-time dosing and site-specific absorption in<br />

GIT.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

18


The conditions that demand pulsatile release include:<br />

Introduction<br />

Many body functions that follow circadian rhythm i.e. their waxes and<br />

wanes with time. Ex: hormonal secretions.<br />

Diseases like bronchial asthma, myocardial infraction, angina pectoris,<br />

rheumatoid diseases, ulcer and hypertension display time dependence.<br />

Drugs that produce biological tolerance demand for a system that will<br />

prevent continuous present at the biophase as this tend to reduce their<br />

therapeutic effect.<br />

The lag time is essential for the drugs that undergo degradation in gastric<br />

acidic medium (ex: peptide drugs) irritate the gastric mucosa or induce<br />

nausea and vomiting.<br />

Targeting to distal organs of GIT like the colon requires that the drug<br />

release is prevented in the upper two-third portion of the GIT.<br />

All of these conditions demand for a time-programmed therapeutic scheme<br />

releasing the right amount of drug at the right time. This requirement is fulfilled<br />

by pulsatile drug delivery system, which is characterized by a lag time that is an<br />

interval of no drug release followed by rapid drug release. 1<br />

Pulsatile systems are basically time-controlled drug delivery systems in<br />

which the system controls the lag time independent of environmental factors like<br />

pH, enzymes, gastrointestinal motility, etc. these time-controlled systems can be<br />

classified as single unit (tablet or capsule) or multiple unit (e.g., pellets)<br />

systems. 1,15<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

19


Fig 2: Drug release profile of pulsatile drug delivery systems<br />

A: Ideal sigmoidal release<br />

1) Single Unit Systems 15,16,17,18<br />

B & C: Delayed release after initial lag time<br />

i) Drug delivery systems with eroding or soluble barrier coatings<br />

Introduction<br />

Most pulsatile delivery systems are reservoir devices coated with a barrier<br />

layer. The barrier dissolves or erodes after a specify lag period, after which the<br />

drug is released rapidly from the reservoir core. In general, the lag time prior to<br />

drug release from a reservoir type device can be controlled by the thickness of the<br />

coating layer. E.g. The Time Clock ® system (West Pharmaceutical Services Drug<br />

Delivery & Clinical Research Centre), and chronotropic ® system consists of a<br />

drug containing core coated by hydrophilic swellable hydroxypropylmethyl<br />

cellulose (HPMC), which is responsible for a lag phase in the onset of release.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

20


ii) Drug delivery systems with rupturable coatings<br />

Introduction<br />

In this the drug is released from a core (tablet or capsule) after rupturing<br />

the surrounding polymeric layer, caused by inbuilt pressure within the system.<br />

The pressure necessary to rupture the coating can be achieved with gas-producing<br />

effervescent excipients, osmotic pressure or swelling agents.<br />

iii) Capsular shaped systems<br />

Several single unit pulsatile dosage forms with a capsular design have<br />

been developed. Most of them consist of an insoluble capsule body, containing the<br />

drug and a plug, which gets removed after a predetermined lag time because of<br />

swelling, erosion or dissolution. E.g., Pulsincap ® system and Port ® system.<br />

The Pulsincap ® system consists of a water-insoluble capsule body<br />

(exposing the body to formaldehyde vapor which may be produced by the addition<br />

of trioxymethylene tablets or potassium permanganate to formalin or any other<br />

method), filled with the drug formulation and plugged with a swellable hydrogel<br />

at the open end. Upon contact with dissolution media or gastrointestinal fluid, the<br />

plug swells and comes out of the capsule after a lag time, followed by a rapid<br />

release of the contents. The lag time prior to the drug release can be controlled by<br />

the dimension and the position of the drug. In order to assure a rapid release of the<br />

drug content, effervescent agents or disintegrants were added to the drug<br />

formulation, especially with water-insoluble drug. Studies in animals and healthy<br />

volunteers proved the tolerability of the formulation (e.g., absence of<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

21


Introduction<br />

gastrointestinal irritation). In order to overcome the potential problem of variable<br />

gastric residence time of a single unit dosage forms, the Pulsincap ® system was<br />

coated with an enteric layer, which dissolved upon reaching the higher pH regions<br />

of the small intestine.<br />

The plug consists of<br />

Swellable materials coated with insoluble, but permeable polymers (e.g.,<br />

polymethacrylates)<br />

Erodible compressed materials (e.g., HPMC, polyvinyl alcohol,<br />

polyethylene oxide)<br />

Congealed melted polymers (e.g., saturated polyglycoated glycerides or<br />

glyceryl monooleate).<br />

Fig. 3: Different stages in drug release from pulsincap<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

22


2) Multiparticulate Pulsatile Drug Delivery Systems:<br />

Introduction<br />

These have various advantages, when compared to single unit dosage<br />

forms, which include, a reproducible gastric residence time, no risk of dose<br />

dumping and the flexibility to blend with different compositions or release<br />

patterns e.g., pellets.<br />

However, drug loading in these systems is low because of higher need of<br />

excipients (e.g. sugar cores). Multiparticulate with pulsatile release pr ofiles are<br />

usually reservoir-type devices with a coating, which either ruptures or changes its<br />

permeability.<br />

1.6 Colon specific drug delivery systems (CSDDS) 16,17<br />

The colonic region of the gastrointestinal tract is one area that would<br />

benefit from the development and use of such modified release technologies.<br />

Although considered by many to be an innocuous organ that has simple functions<br />

in the form of water and electrolyte absorption and the formation, storage and<br />

expulsion of faecal material, the colon is vulnerable to a number of disorders<br />

including ulcerative colitis, crohn’s disease IBS and carcinomas. In addition,<br />

systemic absorption from the colon also be used as a means of achieving<br />

chronotherapy for diseases that are sensitive to circadian rhythms such as asthma,<br />

angina and arthritis.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

23


1.6.1 Structure and function of the Colon: 16<br />

Introduction<br />

The colon forms the lower part of the gastrointestinal tract and extends<br />

from the ileocecal junction at the anus. The colon is upper five feet of the large<br />

intestine and the rectum is the lower six inches. While the colon is mainly situated<br />

in the abdomen, the rectum is primarily a pelvic organ. The first portion of the<br />

colon is spherical and is called cecum. The appendix hangs off the cecum. The<br />

next portion of the colon, in the order in which contents flow, is the ascending<br />

(proximal) colon, just under the liver, the angle or bend is known as the hepatic<br />

flexure, located just beneath the rib cage. The colon then turns to a long horizontal<br />

segment, the transverse colon. Beneath the left rib cage, the colon turns downward<br />

at the haustra, to become the descending (distal) colon. In the left lower portion of<br />

the abdomen, the colon makes an S-shaped curve from the hip over the midline<br />

known as the sigmoid colon. The colon and rectum have an anatomic blood<br />

supply. Along these blood vessels are lymph nodes. Lymph nodes are structures<br />

found in the circulating lymphatic system of the body that produce and store cells<br />

that fight infection, inflammation, foreign proteins and cancer.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

24


1.6.2 pH in colon: 7,16<br />

Fig. 4: Structure of colon<br />

Introduction<br />

Radiotelemetry shows the highest pH levels (7.5 0.5) in the terminal<br />

ileum. On entry into the colon, the pH drops to 6.40.6. The pH in the mid colon<br />

is 6.60.8 and in the left colon 7.00.7. There is a fall in pH on entry into the<br />

colon due to the presence of short chain fatty acids arising from bacterial<br />

fermentation polysaccharides. For example lactose is fermented by the colonic<br />

bacteria to produce large amounts of lactic acid resulting in pH drop to about 5.0.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

25


Table 2: Average pH in the GI tract<br />

Location pH<br />

Oral cavity 6.2-7.4<br />

Oseophagus 5.0-6.0<br />

Stomach Fasted condition:1.5-2.0<br />

Fed condition: 3.0-7.5<br />

Small intestine Jejunum:5.0-6.5<br />

Ileum: 6.0-7.5<br />

Large intestine Right colon: 6.4<br />

1.6.3 Gastrointestinal transit: 7<br />

Mild colon and left colon:6.0-7.6<br />

Introduction<br />

Gastric emptying of dosage form is highly variable and depends primarily<br />

on whether the subject is fed or fasted and on the properties of the dosage forms<br />

such as size and density. The arrival of an oral dosage form at the colon is<br />

determined by the rate of gastric emptying and the small intestinal transit time.<br />

Although the surface area in the colon is low compared to the small intestine, this<br />

is compensated by the markedly slower rate of transit.<br />

Table 3: Average GI transit time<br />

Oral Transit time (hr)<br />

Stomach 3(fed)<br />

Small intestine 3-4<br />

Large intestine 20-30<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

26


1.6.4 Methods for targeting drug into the colon: 7,14,16,19,20<br />

Introduction<br />

By definition, an oral colonic delivery system should retard drug release in<br />

the stomach and small intestine but allow complete release in the colon. The fact<br />

that such a system will be exposed to a diverse range of gastrointestinal conditions<br />

on passage through the gut makes colonic delivery via the oral route a challenging<br />

proposition. Nevertheless a number of approaches have been used and systems<br />

have been developed for the purpose of achieving colonic targeting. These<br />

applications are either drug specific (prodrug) or formulations specific (coated or<br />

matrix preparations). The most commonly used targeting mechanisms are:<br />

1 pH-dependent delivery<br />

2 Time dependent delivery<br />

3 Pressure dependent delivery<br />

4 Bacteria- dependent delivery<br />

1) pH- Triggering Drug Delivery Systems:<br />

Use of pH-dependent polymers is based on the differences in pH levels<br />

throughout GIT. The polymers described as pH- dependent in colon specific drug<br />

delivery are insoluble at low pH levels but become increasingly soluble as pH<br />

rises. The principle group of polymers utilized for the preparation of colon<br />

targeted dosage forms has been the Eudragits (registered trademark of Rohm<br />

Pharma, Darmstadt, Germany), more specifically Eudragit L100 and S100 are<br />

copolymers of methacrylic acid and methyl methacrylate. The ratio of the<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

27


Introduction<br />

carboxyl ester groups is approximately 1:1 in Eudragit L100 and 1:2 in Eudragit<br />

S100. The polymers form salts and dissolve above pH 6 and 7 respectively. This<br />

approach is based on the assumption that gastrointestinal pH increases<br />

progressively from the small intestine to colon. In fact, the pH in the distal small<br />

intestine is usually around 7.5, while the pH in the proximal colon is closer to 6.<br />

The pH-sensitive delivery systems commercially available for mesalazine<br />

(5-aminosalicylic acid) (Asacol ® and Salofalk ® ) and budesonide (Budenofalk ® and<br />

Entocort ® ) for the treatment of ulcerative colitis and Chron’s disease respectively.<br />

2) Time-Dependent Delivery System:<br />

As discussed earlier, although gastric emptying tends to be highly variable,<br />

small intestinal transit times are less (3 1 h). So various attempts are made to<br />

prevent the release of drug until 3-4 h after leaving the stomach. These systems<br />

release their drug load after a pre-programmed time delay. To attain colonic<br />

release, the lag time should equate to the time taken for the system to reach the<br />

colon. This time is difficult to predict in advance, although a lag time of five hours<br />

is usually considered sufficient, given that small intestinal transit time is reported<br />

to be relatively constant at three to four hours.<br />

The drug delivery systems, Pulsincap ® and Time Clock ® are time<br />

dependent formulations.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

28


3) Pressure-Controlled Drug Delivery Systems:<br />

Introduction<br />

Gastrointestinal pressure has also been utilized to trigger drug release in<br />

the distal gut. This pressure, which is generated via muscular contractions of the<br />

gut wall for grinding and propulsion of intestinal contents, varies in intensity and<br />

duration throughout the gastrointestinal tract, with the colon considered to have a<br />

higher luminal pressure due to the processes that occur during stool formation.<br />

Systems have therefore been developed to resist the pressures of the upper<br />

gastrointestinal tract but rupture in response to the raised pressure of the colon.<br />

The system can be modified to withstand and rupture at different pressures by<br />

changing the size of the capsule and thickness of the capsule shell wall.<br />

Takaya et al. have developed pressure-controlled colon-delivery capsules<br />

prepared using an ethyl cellulose, which is insoluble in water.<br />

4) Microflora-Activated Drug Delivery Systems:<br />

The resident gastrointestinal bacteria provide a further means of effecting<br />

drug release in the colon. These bacteria predominantly colonize the distal regions<br />

of the gastrointestinal tract where the bacterial count in the colon is 10 11 per<br />

gramme, as compared with 10 4 per gramme in the upper small intestine. Moreover<br />

400 different species are present. Colonic bacteria are predominantly anaerobic in<br />

nature and produce enzymes that are capable of metabolizing endogenous and<br />

exogenous substrates, such as carbohydrates and proteins that escape digestion in<br />

the upper gastrointestinal tract. Both prodrugs and dosage forms from which the<br />

release of drug is triggered by the action of colonic bacteria enzymes have<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

29


Introduction<br />

devised. Enzymes produced by the colonic bacterial are capable of catalyzing a<br />

number of metabolic reactions, which includes reduction (of double bonds, nitro<br />

groups, azo groups, aldehydes, sulphoxides, ketones, alcohols, N-oxides and<br />

arsenic acid), hydrolysis (of glycosides, sulphates, amides, esters, nitrates and<br />

sulphonates), deamination, decarboxylation, dealkylation, acetylation, nitrosamine<br />

formation, heterolytic ring fission and esterification.<br />

Design<br />

strategy<br />

Table 4: Summary of colon-specific drug delivery strategies 16,19,20<br />

Drug release triggering<br />

mechanisms<br />

Prodrugs Cleavage of the linkage<br />

bond between drug and<br />

carrier via reduction and<br />

hydrolysis by enzyme from<br />

colon bacteria. Typical<br />

enzymes include<br />

azoreductase, glycosidase,<br />

and glucuronidase.<br />

pHdepedent<br />

systems<br />

Timedependent<br />

system<br />

Microfloro<br />

activated<br />

system<br />

Combination of polymers<br />

with pH-dependent<br />

solubility to take advantage<br />

of the pH changes along the<br />

GI tract.<br />

The onset of drug release is<br />

aligned with positioning the<br />

delivery system in the colon<br />

by incorporating a time<br />

factor simulating the system<br />

transit in upper GIT.<br />

Primarily fermentation of<br />

non-starch polysaccharides<br />

by colon anaerobic bacteria.<br />

The polysaccharides are<br />

incorporated into the<br />

delivery system via film<br />

coating and matrix<br />

formation.<br />

Advantages Disadvantages<br />

Able to achieve<br />

site specificity.<br />

Formulation<br />

well protected<br />

in the stomach<br />

Small intestine<br />

transit time<br />

fairly<br />

consistent.<br />

Good site<br />

specificity with<br />

prodrugs and<br />

polysaccharides.<br />

It will be considered as a<br />

new chemical entity<br />

from regulatory<br />

prespective. So far this<br />

approch has been<br />

primarily constricted to<br />

achives releated to the<br />

treatment of IBD.<br />

Unpredictable sitespecificity<br />

of drug<br />

release because of<br />

inter/intra subject<br />

variation of pH between<br />

small intestine and the<br />

colon.<br />

Substantial variation in<br />

gastric retention times<br />

make it complicated in<br />

predicting the accurate<br />

location of drug release.<br />

Diet and disease can<br />

affect colonic<br />

microflora; enzymatic<br />

degredation may be<br />

excessively slow.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

30


1.7 Microencapsulation: 7<br />

Introduction<br />

A microcapsule is a tiny capsule and its preparation produced, called<br />

microcapsulation, can endow various traits to core material in order to add<br />

secondary functions and/or compensate for shortcomings.<br />

Microencapsulation is a technique to prepare tiny packaged materials<br />

called microcapsules that have many interesting features. This technique has been<br />

employed in a diverse range of fields from chemicals and pharmaceuticals to<br />

cosmetics and printing. For this region widespread interest has developed in<br />

microencapsulation technology. Microcapsules are tiny microparticles with<br />

diameters in the range of nanometres or millimetres that consist of core materials<br />

and convering membranes (sometime also called walls). The most significant<br />

features of microcapsules is their size that allows for a huge surface or interface<br />

area. Through selection of the composition materials (sometimes also called<br />

walls). The most significant feature of microcapsules is their microscopic size that<br />

allows for a huge surface or interface area. Through selection of composition<br />

materials (core material and membrane), we can endow microcapsule with a<br />

variety of functions. Generally, membrane materials are chosen in order to<br />

pronounce the effects of microencapsulation. Therefore, not only synthetic and<br />

natural polymers but also lipids and inorganic materials are used for the<br />

preparation of microcapsules.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

31


Introduction<br />

Microcapsules have a number of interesting advantages and the main<br />

reasons for microencapsulation cap be exemplified as<br />

1. Controlled release of en encapsulating drugs,<br />

2. Protection of the encapsulated materials against oxidation or deactivation due<br />

to reaction in the environment<br />

3. Masking of odour and/or taste of encapsulating materials.<br />

4. Isolation of encapsulating materials from undesirable phenomena, and<br />

5. Easy handling as powder-like materials.<br />

Microcapsule can also be classified into three basic categories according to<br />

their morphology as mono-cored, poly-cored and matrix type as shown in Fig 8<br />

Morphological control is important and much effort has been given to controlling<br />

internal structures, which largely depend on protocol and the microcapsulation<br />

methods employed.<br />

Fig 5: Classification of microparticles.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

32


2.1 Need for the study:<br />

CHAPTER-2<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Objectives<br />

The development of oral sustained and controlled release formulation<br />

offer benefits like controlled administration of therapeutic dose at the delivery<br />

rate, constant blood levels of the drug, reduction of side effects minimizations<br />

of dosing frequency and enhancement of patient compliance. 21<br />

Among modified-release oral dosage form increasing interest has<br />

currently turned to system designed to achieve time-specific (delayed, pulsatile)<br />

and site-specific delivery of drug. The possibility of exploiting delayed release<br />

to perform chronotherapy, is quite appealing for those diseases, the symptoms<br />

of which recur mainly at night time or in the early morning, such as bronchial<br />

asthma, angina pectoris and rheumatoid arthritis. 22,23,24<br />

Flurbiprofen [1,1’-biphenyl]-4-acetic acid, 2-fluoro-alpha-methyl-, is an<br />

important analgesic and non-steroidal anti-inflammatory drug (NSAID) also<br />

with anti-pyretic properties whose mechanism of action is the inhibition of<br />

prostaglandin synthesis. It is used in the therapy of rheumatoid disorders.<br />

Flurbiprofen is rapidly eliminated from the blood, its plasma elimination half-<br />

life is 3-6 hours and in order to maintain therapeutic plasma levels. The drug<br />

must be administered approximately 150-200mg daily by oral in divided<br />

doses. 25<br />

33


DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Objectives<br />

The aim of this work is to design site-specific (pulsatile) drug delivery<br />

system with pH sensitive methacrylic acid co-polymers and Flurbiprofen as<br />

model drug, whose drug delivery system.<br />

2.2 Objective of the study<br />

To design and characterize an oral, drug delivery system of Flurbiprofen<br />

intended to approximate the chronobiology of arthrities, proposed for colonic<br />

targeting. It is a chronopharmaceutical approach for the better treatment of<br />

rheumatoid arthritics.<br />

Based on the concept that a formulation on leaving the stomach, arrives at<br />

the ileocaecal junction in about 6 hours after administration and difference in pH<br />

throughout GIT, a time and pH dependent pulsatile ( or modified pulsincap),<br />

controlled drug delivery system was designed.<br />

This capsule consists of a non-disintegrating body and a soluble cap. The<br />

drug formulations is contained within the capsule body and separated from the<br />

water-soluble cap by a hydrogel polymer plug. The entire capsule is enteric coated<br />

to prevent variable gastric emptying. The enteric coating prevents disintegration<br />

of the soluble cap in the gastric fluid. On reaching the small intestine the capsule<br />

will lose its enteric coating and the polymer plug inside the capsule swells to<br />

create a lag phase that equals the small intestinal transit time. This plug ejects on<br />

swelling and releases the drug from the capsule in the colon.<br />

34


2.3 Plan of Research Work<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Objectives<br />

1. Selection of polymers and its combinations suitable for the colonic drug<br />

delivery.<br />

2. Preparation of Flurbiprofen microcapsules.<br />

3. Evaluation of microcapsules:<br />

a) Particle size<br />

b) Study of flow properties<br />

c) Percentage yield<br />

d) Drug content<br />

c) Drug-Polymer interaction<br />

d) External morphology<br />

c) In-vitro drug release kinetics<br />

4) Preparation of cross-linked gelatine capsules.<br />

a) Formaldehyde treatment of capsule bodies<br />

b) Test for formaldehyde treated empty capsule<br />

Physical test<br />

Identification attributes<br />

Visual defect<br />

Dimensions<br />

Solubility studies of treated capsules<br />

35


Chemical test<br />

Qualitative test for free formaldehyde<br />

5) Development of pulsatile drug delivery system.<br />

6) Evaluation of pulsatile drug delivery system<br />

a) Thickness of cellulose acetate phthalate coating<br />

b) Weight variation<br />

c) In-vitro drug release kinetics<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Objectives<br />

36


3.1 Literature Survey<br />

CHAPTER-3<br />

Review Of Literature<br />

<br />

Listair CR, et al. 26 developed a chronopharmaceutical capsule drug<br />

delivery system capable of releasing drug after predetermined time delays. The<br />

drug formulation is sealed inside the capsule body by an erodible table (ET). The<br />

release time is determined by an ET erosion rate and increases as the content of an<br />

insoluble excipient (dibasic calcium phosphate) and of gel forming excipient<br />

(HPMC) increases. Programmable pulsatile release has been achieved from a<br />

capsule over a 2-12 hrs period, consistent with the demands of chronotherapeutic<br />

drug delivery.<br />

Samantha MK, et al. 27 designed and evaluated Pulsincap drug delivery<br />

system of Salbutamol sulphate for drug targeting to colon in disease condition like<br />

asthma. Empty gelatin capsule was coated with ethycellulose keeping the cap<br />

portion as such. A hydrogel plug made of gelatin was suitably coated with<br />

Cellulose Acetate Phthalate in such a way that it was fixed to the body under the<br />

cap. Eudragit microcapsule containing Salbutamol sulphate were prepared and<br />

incorporated into this specialized capsule shell. The in-vitro dissolution studies<br />

indicated that the onset of drug release was after 7 to 8 hrs of the experiment and<br />

revealed its better sustaining efficacy over a period of 24hrs.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

37


Review Of Literature<br />

Young-II Jeong, et al. 28 evaluated pressure-controlled colon delivery<br />

(PCDCS) capsules prepared by a dipping method. Empty PCDCS are composed<br />

of two polymer membranes. The inner one was a water-insoluble polymer<br />

membrane, ethycellulose (EC) and the outer one was an enter ic polymer<br />

membrane. Hydroxypropyl methylcellulose phthalate (HPMCAS). By<br />

consequently dipping in an ethanolic EC solution and alkalized enteric polymer<br />

solution, PCDCS were obtained after both the capsule body and cap were adjusted<br />

to the size of #2 capsules.<br />

Seshasayan A, et al. 29 studied release of Rifampicin from modified<br />

pulsincap preparation, using different preparation of various hydrophilic polymer<br />

such as guar gum, cabapol-940, sodium alginate, hydroxypropyl methylcellulose,<br />

gum karaya and poly vinyl alcohol. Among all the polymers tested guar gum<br />

showed better sustaining capacity even at low concentration.<br />

Julie Binns, et al. 18 studied the tolerability of multiple oral dose of<br />

pulsincap ® capsule in a healthy volunteers. Twelve healthy subjects entered a<br />

double-blind placebo-controlled parallel group study to evaluate the tolerability of<br />

a 28 days course of twice daily dosage with pulsincap ® capsules having the plug<br />

set for separation after 6 hrs.<br />

Sangalli ME, et al. 30 performed in-vitro and in-vivo evaluation of an oral<br />

system (Chronotropic TM ) designed to achieve time & /or site specific release.<br />

Cores containing antipyrine as the model drug were prepared by tableting and<br />

both the retarding and enteric coatings were applied in fluid bed. The release test<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

38


Review Of Literature<br />

was carried out in a USP 24 paddle apparatus. The in-vitro testing, performed on<br />

healthy volunteers, envisaged the HPLC determination of antipyrine salivary<br />

concentration and γ-scintigraphic investigation.<br />

Zahirul Khan MI, et al. 31 formulated a pH-dependent colon targeted oral<br />

drug delivery system using methacrylic acid copolymers. Drug release was<br />

manipulated using Eudragit ® 100-55 and Eudragit ® S100 combinations. The<br />

coated tablets were tested in-vitro for their sutability for pH dependent colon<br />

targeted oral delivery.<br />

Howard Stevens NE, et al. 32 designed pulsincap ® formulations to delivery<br />

a dose of drug following 5 h delay and evaluated the capability of the formulation<br />

to deliver defetilide to the lower GIT. The combination of scintigraphic and<br />

pharmacokinetic analysis permits identification of the site of drug release from the<br />

dosage form and pharmacokinetic parameters to be studied in man in a non-<br />

invasive manner.<br />

Libo Yang, et al. 33 reviewed the new approaches in-vitro and in-vivo<br />

evaluation of colon specific drug delivery systems.<br />

Ying-huan Li, et al. 34 developed a multifunctional and multiple unit<br />

system, which contains versatile mini-tablets in a hard gelatin capsule, as Rapid-<br />

release Mini-tablets (RMTs), Sustained -release Mini-Tablets (SMTs), Pulsatile<br />

Mini-Tablets (PMTs) and Delayed onset Sustained- release Mini tablets (DSMTs),<br />

each with various lag times of release.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

39


Review Of Literature<br />

Tomohiro Takaya, et al. 35 studied the importance of dissolution process<br />

on systemic availability of drugs delivered by colon delivery system. The<br />

relationship between in-vitro drug release characteristics from colon delivery<br />

systems and in-vivo drug absorption was investigated using three kinds of<br />

delayed-release systems. Pressure controlled colon delivery capsules for liquid<br />

preparations, time controlled colon delivery capsules for liquid and solid<br />

preparations and Eudragit S coated tablets for solid preparations were used in this<br />

study.<br />

Takashi Ishibashi, et al. 36 evaluated colonic absorbability of drugs dog<br />

using a novel colon-targeted delivery capsule (CTDC). A series of dog studies<br />

were performed to examine the in- vitro/in-vivo relationship of drug release<br />

behaviour of the newly developed CTDC. The 4 kinds of CTDCs containing<br />

theophylline, each of which has a different in-vitro dissolution lag time, were<br />

orally administered to 4 beagle dogs under fasted condition and the onset time of<br />

drug absorption were compared.<br />

Jonathan CDS, et al. 37 investigated the coating-dependent release<br />

mechanism of a pulsatile capsule using Nuclear Resonance microscopy.<br />

Chrononopharmaceutical capsules, ethycellulose-coated to prevent water ingress,<br />

exhibited clearly different characteristic when coated by organic or aqueous<br />

processes.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

40


Review Of Literature<br />

Mastiholimath, et al. 38 attempt was made to that of the small intestine<br />

(7.0–7.8). So, by using the deliver theophylline into colon by taking the advantage<br />

of the fact that colon has a lower pH value (6.8) than mixture of the polymers, i.e.<br />

Eudragit L and Eudragit S in proper proportion, pH dependent release in the colon<br />

was obtained.<br />

Abraham S, et al. 39 reported modified pulsincap dosage form of<br />

metronidazle to target drug release in colon by using extrusion-spheronization<br />

method.<br />

Sungthongjeen S, et al. 40 reported development of pulsatile release tablets<br />

with swelling and rupturable layers. Finally concluded that pulsatile release tablets<br />

with a swelling layer and rupturable ethyl cellulose coating were developed. The<br />

system released the drug rapidly after a certain lag time due to the rupture of ethyl<br />

cellulose film.<br />

Krishnaiah YSR, et al. 41 development of colon targeted drug delivery<br />

systems for mebendazole The tablets were evaluated for drug content uniformity,<br />

and were subjected to in vitro drug release studies. Differential scanning<br />

calorimetry (DSC).<br />

Satyanarayana S, et al. 42 presented the delivery of drugs to the colon for<br />

local effects, including drug candidates, methods used for studying colonic drug<br />

absorption and colon-specific drug delivery systems, role of absorption enhancers<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

41


Review Of Literature<br />

in colonic drug delivery, and the use of prodrugs, drugs coated with pH dependent<br />

or pH independent biodegradable polymers, or polysaccharide matrices<br />

Ram Prasad YV, et al. 43 assessed the utility of guar gum as a carrier for<br />

colon specific drug delivery using indomethacin as model drug from an oral<br />

matrix tablet formulation containing 40 mg of drug and 340 mg of guar gum in 0.1<br />

M HCl for 2 hrs and pH 7.4 phosphate buffer for 3 hrs in albino rat cecal content<br />

after 5 hrs of testing under conditions mimicking mouth to colon transit, guar gum<br />

was found to be susceptible to colon bacterial enzyme action with the consequent<br />

release of indomethacin.<br />

Krishnaiah YSR, et al. 44 used guar gum as a carrier for colon specific and<br />

studied influence of metronidazole and tinidazole on in-vitro release of<br />

albendazole from guar gum matrix tablet and it was found that the release of drug<br />

from guar gum formulations increased with a decrease in the dose of<br />

metronidazole / tinidazole.<br />

Leopold CS, et al. 45 carried out in-vitro study for the assessment of poly<br />

(Laspartic acid) as a drug carrier for colon specific drug delivery, using<br />

dexamethasone as model drug. It was concluded that poly (L -aspartic acid)<br />

appears to be a suitable drug carrier for colon specific drug delivery.<br />

Rubinstein et al. 46 (1995) showed that calcium pectinate -indomethacin<br />

tablets of both types, i.e. compression coated and matrix tablets give no release of<br />

indomethacin at a pH-1.5 for 2 hrs.When these tablets were shaken at a pH-7.4 a<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

42


Review Of Literature<br />

drug leak was seen in plain matrix tablets but not in compression coated tablets. In<br />

the presence of pectinolytic enzymes, a sudden release of indomethacin was seen<br />

in both, the types of tablets but the rate and the percentage of release were lower<br />

(only 57.6#2.5% ) in compression coated tablets as compared to plain matrix<br />

tablets (74.2#4%) after 12hrs.<br />

Turkoglu, et al. 47 evaluated the pectin-HPMC system for compression<br />

coated core tablets of 5-aminosalicylic acid (5 -ASA) for colonic delivery. Drug<br />

dissolution/system erosion/degradation studies were carried out in pH 1.2 and 6.8<br />

phosphate buffers using a pectinolytic enzyme. The system was designed based on<br />

the gastrointestinal transit time concept, under the assumption of colon arrival<br />

times of 6 hrs. It was found that pectin alone was not sufficient to protect ate core<br />

tablets and HPMC addition was required to control the solubility of pectin. The<br />

optimum HPMC concentration was 20% and such system would protect the<br />

course up to 6 h that corresponded to 25-35% erosion and after that under the<br />

influence of pectinase the system would degrade faster and delivering 5-ASA to<br />

the colon. The pectin-HPMC envelope was found to be a promising drug delivery<br />

system for those drugs to be delivered to the colon.<br />

Rama Prasad et al. 48 evaluated the suitability of guar gum as a carrier in<br />

colonic drug delivery. In this study, matrix tablet of indomethacin with guar gum<br />

were prepared. These tablets were found to retain their integrity in 0.1M HCl for 2<br />

hrs and in Sorensen’s phosphate buffer (pH-7.4) for 3 hr releasing only 21% of the<br />

drug in these 5 hrs. However, in presence of 2% rat cecal contents the drug<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

43


Review Of Literature<br />

release increased and further increased with 4% concentration of cecal contents.<br />

The drug release improved to about 91% in 4% cecal content medium after<br />

enzyme induction of rats. This study suggests the specificity of these matrices for<br />

enzyme trigger in the colon to release the drug.<br />

Krishnaiah, et al. 49 evaluated guar gum as a compression coating to protect<br />

the drug core of 5- ASA in upper GIT. Percent drug release from tablet increased<br />

considerably from 11 hr and the tablets were completely disintegrated in 26 hr in<br />

the presence of the rat cecal medium. The results of drug release studies on<br />

compression coated tablets suggested that the thickness of guar gum coating in the<br />

range of 0.61-0.91 mm was sufficient to deliver the drugs selectively to the colon.<br />

Krishnaiah et. al. 50 studied to develop colon targeted drug delivery systems<br />

for metronidazole using guar gum as a carrier. Matrix, multilayer and compression<br />

coated tablets of metronidazole containing various proportions of guar gum were<br />

prepared. Matrix tablets and multilayer tablets of metronidazole failed to control<br />

the drug release within 5 hrs of the dissolution study in the physiological<br />

environment of stomach and small intestine. The compression coated formulations<br />

released less than 1% of metronidazole in the physiological environment of<br />

stomach and small intestine. The results of the study show that compression<br />

coated metronidazole tablets with either 275 or 350 mg of guar gum coat is most<br />

likely to provide targeting of metronidazole for local action in the colon owing to<br />

its minimal release of the drug in the first 5 hrs.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

44


Review Of Literature<br />

Shinha, et al. 51 evaluated polysaccharide (combined xanthan gum and guar<br />

gum) matrices for microbially triggered drug delivery to the colon. Matrix tablets<br />

were prepared using xanthan gum and guar gum in vary in proportions using<br />

indomethacin as model drug. The ability of the prepared matrices to retard drug<br />

release in the upper gastrointestinal tract and to undergo enzymatic hydrolysis by<br />

the colonic bacteria was evaluated in the presence of rat cecal content. Guar gum<br />

alone as a drug release retarding excipient in the matrices does not achieve the<br />

desired retardation. Presence of xanthan gum in the tablets not only retard the<br />

initial drug release from the tablets, but due to high swelling, make them more<br />

vulnerable to digestion by the microbial enzymes in the colon.<br />

Lorenzo-lamosa, et al. 52 prepared and demonstrated the efficacy of a<br />

system, which combines specific biodegradability and pH dependent release<br />

behaviour. The systemconsists of chitosan microcores entrapped within acrylic<br />

microspheres containing diclofenac sodium as model drug. The drug was<br />

efficiently entrapped within the chitosan microcores using spray drying and then<br />

microencapsulated into Eudragit L100 using and Eudragit S100 using an oil-in-oil<br />

solvent evaporation method. Release of the drug from chitosan multi-reservoir<br />

system was adjusted by changing the chitosan molecular weight or the type of<br />

chitosan salt. No release was seen in acidic pH for 3hr, but at higher pH Eudragit<br />

dissolved and swelling of chitosan started leading to continuous drug diffusion<br />

which completed in the next 4hr. Furthermore, by coating the chitosan microcores<br />

with Eudragit ® perfect pH-dependent release profiles were attained.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

45


Review Of Literature<br />

Brondsted et al. 53 investigated the application of glutaraldehyde cross<br />

linked dextran as a capsule material for colon-specific drug delivery. These<br />

capsules were loaded with hydrocortisone and drug release studies were<br />

conducted in vitro. Ten percent of drug was released in initial 3h and only about<br />

35% in 24 h at pH 5.4. Addition of dextranase enzyme after 24 h resulted in a<br />

rapid degradation of the capsule leading to fast and complete release of<br />

hydrocortisone. However, these results reflect only an experimental condition and<br />

not the in vivo situation.<br />

Vervoort, et al. 54,55 synthesized methacrylated insulin and aqueous<br />

solutions of methacrylated insulin upon free radical polymerization, were<br />

converted to cross-linked hydrogels. Rheological studies and characterization of<br />

there hydrogels showed that higher substituted insulin had better network and<br />

higher mechanical strength. These hydrogels were there studied for their swelling<br />

properties and degradation in vitro. Degradation studies carried out in presence of<br />

insulinase showed that increasing enzyme concentration and incubation time<br />

degraded insulin faster. However, increasing the substitution on insulin molecules<br />

resulted in stronger hydrogels with less enzyme diffusion thereby less<br />

degradation.<br />

Rubinstein, et al 56 developed cross-linked chondroitin as a drug carrier for<br />

colon-specific delivery. Chondroitin sulphate was cross-linked with 1,12-<br />

diaminododecane via dicyclohexylcarbodiimide activation. Cross-linked<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

46


Review Of Literature<br />

chondroitin sulphate was used to form a matrix tablet with indomethacin. Release<br />

ofindomethacin from the tablet was studied in the presence of rat cecal contents<br />

ascompared to release in phosphate buffer saline. A significant difference in drug<br />

release was observed after 14 hrs in the two dissolution media. Also, different<br />

degreesof cross-linked chondroitin sulphate were used to study their effect on<br />

drug releasefrom the matrices. The cumulative percent release of indomethacin<br />

from cross-linked chondroitin matrix tablet showed that release was increased in<br />

the presence of rat cecal contents. Studies on rat cecal contents with various cross-<br />

linked chondroitin sulphate showed greater cumulative drug release when cross-<br />

linking was less and as crosslinking increased the cumulative release decreased<br />

i.e. a linear relationship was found between the degree of cross-linking of polymer<br />

and the amount of drug released in rat cecal content.<br />

Milojevic, et al. 57 evaluated Amylose-Ethocel ® system for their potential<br />

as microbial degradable colon drug carrier. Amylose-Ethocel ® coating system<br />

resistant to gastric acid and small intestinal enzymes, but degradable by colonic<br />

bacteria. Varying concentration of Amylose and Ethocel ® in the form of aqueous<br />

dispersions were use to coat 5-ASA pellets. Coating formulation comprising<br />

Amylose and Ethocel ® in the ratio of 1:4 w/w showed optimum drug release<br />

retarding properties in gastric and intestinal fluids.<br />

Shantha KL, et al. 58 developed azo polymeric hydrogels for colon targeted<br />

drug delivery and studied various physicochemical properties such as FT-IR,<br />

SEM, Equilibrium swelling studies, In-vitro release.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

47


Review Of Literature<br />

Takashi Ishibashi, et al. 59 developed new capsule-type dosage form for<br />

colon-targeted delivery of drugs The system was designed by imparting a timed-<br />

release function and a pH-sensing function to a hard gelatin capsule. The technical<br />

characteristics of the system are to contain an organic acid together with an active<br />

ingredient in a capsule coated with a three-layered film consisting of an acid-<br />

soluble polymer, a water-soluble polymer, and an enteric polymer. In order to find<br />

the suitable formulation, various formulation factors were investigated through a<br />

series of in vitro dissolution studies. As a result, it was found that: (1) various<br />

organic acids can be used for this system: (2) a predictable timed -release<br />

mechanism of a drug can be attained by adjusting the thickness of the Eudragit ® E<br />

layer: and (3) the outer enteric coating with HPMC ® -AS provided acceptable acid-<br />

resistibility. All these results suggested that this approach can provide a useful and<br />

practical means for colon-targeted delivery of drugs.<br />

Anita L et. al. 60 reviewed the ability to deliver therapeutic agents to a<br />

patient in a pulsatile or staggered release profile has been a major goal in drug<br />

delivery research recently. This review will cover methods that have been<br />

developed to control drug delivery profile with different polymeric systems.<br />

Externally and internally controlled system will be considered including a range<br />

of technologies like pre-programmed systems as well as systems that are sensitive<br />

to modulated enzymatic or hydrolytic degradation, pH, magnetic fields,<br />

ultrasounds, electric fields, temperature, light and mechanical stimulation. These<br />

systems have the potential to improve the quality of life for patients undergoing<br />

therapy with a variable dosing regimen.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

48


Review Of Literature<br />

Morta Rodriguez, et al. 61 presented a new multiparticulate system to<br />

deliver active molecules to colonic region, which combines pH-dependent and<br />

controlled drug release properties. This system was constituted by drug loaded<br />

cellulose acetate butyrate (CAB) microspheres coated by an enteric polymer<br />

(eudragit ® S). Both, CAB cores and pH-sensitive microcapsules were prepared by<br />

the emulsion solvent evaporation technique in an oil phase. The in-vitro drug<br />

release studies of pH-sensitive microcapsules containing the hydrophobic cores<br />

showed that no drug was released below pH 7.<br />

Lorenzo- Lamosa ML, et al. 62 designed the microcapsulated chitoson<br />

microspheres for colonic drug delivery. Sodium diclofenac(SD) used as a model<br />

drug, was effectively entrapped within chitoson (CS) microcores using spray -<br />

drying and then microcapsulated into Eudragit ® L-100 and S-100 using an oil-in-<br />

oil solvent evaporation method. A combined mechanism of release is proposed,<br />

which considers the dissolution of eudragit coating, the swelling of the CS<br />

microcores and the dissolution of SD and its further diffusion through the CS gel<br />

cores.<br />

Shivkumar HN, et al. 63 designed the multiparticulate system comprising of<br />

nonpareil seeds coated with a mixture of Eudragit S-100 and L-100 for<br />

chronopharmaceutical delivery of diclofenac sodium. The in-vitro dissolution test<br />

showed that the release of drug from coated pellets depended on the pH of the<br />

dissolution fluid and the coat weights applied.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

49


Review Of Literature<br />

Kristmudsodottir T, et al. 64 prepared microcapsule containing diltiazem<br />

hydrochloride by the Spray drying technique using Acrylate-methaacrylate<br />

copolymers, Eudragit RS and RL as coating materials and studied the drug release<br />

rate and concluded that the drug release rate can be controlled by choice of<br />

polymer type and production condition during spray-drying.<br />

Hideki Ichikawa, et al. 65 designed the prolonged-release microcapsules<br />

containing diclofenac sodium for oral suspension and their preparation by the<br />

wurster process.<br />

Giovanni FP, et al. 66 prepared controlled release of ketoprofen by emulsion<br />

solvent evaporation method. The prepared microspheres were characterized by<br />

scanning electron microscopy, x-ray diffractometry, and in-vitro dissolution<br />

studies and then used for the preparation of tablets.<br />

Alf Lamprecht, et al. 67 designed the microsphere for the colonic delivery<br />

of 5-fluorouracil prepared by a simple and fast o/w emulsification method. A<br />

relatively high drug load even for the hydrophilic drug 5-FU can be achieved. A<br />

pH-dependent release can be provided in a range of pH 6.8, where the main drug<br />

load is retained, and pH 7.4, where a fast dissolution of the carrier occurs. The<br />

enlargement of the particle results in a slight retardation effect on the drug release.<br />

When polymer combinations were applied for the microsphere preparation<br />

(Eudragit RS100 and Eudragit P-4581F) at different mixing ratios, only minor<br />

influences on particle size and drug load are observed. These simple polymer<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

50


Review Of Literature<br />

mixtures can prolong the release only for a relatively short period due to the<br />

exceptional structural arrangements of the carrier system.<br />

Lars hovgaard, et al. 68 developed dextran hydrogel for colon-specific drug<br />

delivery. Degradation of the hydrogels has been studied in vitro using dextrose, in<br />

vivo in rate and in human fermentation model. It was found that by changing the<br />

chemical composition of hydrogels it is possible to control the equilibrium degree<br />

of swelling, mechanical strength and degradability. The dextran hydrogel were<br />

degraded in vivo in the cecum of rat but not in stomach. Furthermore, the dextran<br />

hydrogel were degraded in human colonic fermentation model, indicating that<br />

dextranases are indeed present in human colonic contents. Finally, releases of<br />

hydrocortisone from the hydrogels were evaluated. It was found to depend on the<br />

presence of dextranases in the release medium. the result suggest that the dextran<br />

hydrogels are promising as drug carries for colon-specific drug deliver.<br />

Mine Orlu, et. al. 69 designed and evaluated colon specific drug delivery<br />

system containing flurbiprofen microsponges Microsponges containing FLB<br />

(Flurbiprofen) and EudragitRS100 were prepared by quasi-emulsion solvent<br />

diffusion method. Additionally, FLB was entrapped into a commercial<br />

Microsponge ® 5640 system using entrapment method. Afterwards, the effects of<br />

drug: polymer ratio, inner phase solvent amount, stirring time and speed and<br />

stirrer type on the physical characteristics of microsponges were investigated. The<br />

thermal behaviour, surface morphology, particle size and pore structure of<br />

microsponges were examined.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

51


3.2 Drug profile: 25,70,71<br />

Flurbiprofen<br />

Molecular Formula : C15H13FO2<br />

Average Molecular Weight : 244.2609<br />

Drug Category: Analgesics<br />

Chemical Structure:<br />

Anti-Inflammatory Agents<br />

Non-Steroidal Anti-inflammatory Agents<br />

Carbonic Anhydrase Inhibitors<br />

Cyclooxygenase Inhibitors<br />

Chemical Name: 2-(3-fluoro-4-phenylphenyl) propanoic acid<br />

Description:<br />

Physical state and appearance: Solid. (Solid crystalline powder.)<br />

Odor : Practically odourless<br />

Taste : Not available.<br />

Review Of Literature<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

52


Molecular Weight : 244.27 g/mole<br />

Color : White to yellowish.<br />

Melting Point : 110°C (230°F)<br />

Solubility:<br />

Partially soluble in methanol.<br />

Very slightly soluble in cold water.<br />

Insoluble in diethyl ether.<br />

Stability: The product is stable.<br />

Review Of Literature<br />

Storage: Keep container tightly closed. Keep container in a cool, well-ventilated<br />

area.<br />

Indication: Flurbiprofen tablets are indicated for the acute or long-term treatment<br />

of the signs and symptoms of rheumatoid arthritis and osteoarthritis<br />

Pharmacology:<br />

Flurbiprofen, a non- steroidal anti-inflammatory drug (NSAID) of the<br />

propionic acid class, is used for the relief of pain an inflammatioassociated with<br />

rheumatoid arthritis and osteoarthritis and for the inhibition of intraoperative<br />

miosis. Flurbiprofen exhibits anti-inflammatory, analgesic, and antipyretic<br />

activities<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

53


Mechanism of Action:<br />

Review Of Literature<br />

The anti-inflammatory effect of flurbiprofen may result from the<br />

reversible inhibition of cyclooxygenase, causing the peripheral inhibition of<br />

prostaglandin synthesis. Flurbiprofen also inhibits the migration of leukocytes into<br />

sites of inflammation and prevents the formation of thromboxane A2, an<br />

aggregating agent, by the platelets<br />

Absorption:<br />

The mean oral bioavailability of flurbiprofen from tablets is 96% relative<br />

to an oral solution.<br />

Toxicity : LD50=10 mg/kg (orally in dogs).<br />

Protein Binding : > 99%<br />

Biotransformation:<br />

Hepatic. Cytochrome P450 2C9 plays an important role in the metabolism<br />

of flurbiprofen to its major metabolite, 4’-hydroxy-flurbiprofen. The 4’-hydroxy-<br />

flurbiprofen metabolite showed little anti-inflammatory activity in animal models<br />

of inflammation<br />

Dose : 150-200mg daily by oral in divided doses.<br />

Half Life : 4.7-5.7 hours<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

54


Interactions:<br />

Drug Interaction<br />

Review Of Literature<br />

Acenocoumarol The NSAID increases the anticoagulant effect<br />

Alendronate Increased risk of gastric toxicity<br />

Anisindione The NSAID increases the anticoagulant effect<br />

Cyclosporine Monitor for nephrotoxicity<br />

Dicumarol The NSAID increases the anticoagulant effect<br />

Methotrexate<br />

The NSAID increases the effect and toxicity of<br />

methotrexate<br />

Warfarin The NSAID increases the anticoagulant effect<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

55


3.3 Polymer Profile<br />

1. EUDRAGIT L 100 <strong>AND</strong> EUDRAGIT S 100: 72<br />

Synonyms : Methacrylic acid, Eudragit<br />

Functional category : Film former, tablet binder<br />

Review Of Literature<br />

Chemical name: Copolymers synthesized from dimethylaminoethyl methacrylate<br />

and other neutral methacrylic esters<br />

Basic structure:<br />

Eudrgit ® L 100 and S 100 are anionic copolymers based on methacrylic<br />

acid and methyl methacrylate. The ratio of the free carboxyl groups to the ester<br />

groups is approx. 1:1 in eudragit ® L 100 and approx. 1:2 in eudrgit ® S 100.The<br />

average molecular weight is approx. 135,000.<br />

Description : White powders with a faint characteristic odour<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

56


Review Of Literature<br />

Solubility: 1 g of eudragit ® L 100 or eudragit ® S 100 dissolves in 7 g methanol,<br />

ethanol, in aqueous isopropyl alcohol and acetone (containing approx. 3 % water),<br />

as well as in 1 N sodium hydroxide to give clear to slightly cloudy solutions.<br />

EUDRAGIT ® L 100 and S 100 are practically insoluble in ethyl acetate,<br />

methylene chloride, petroleum ether and water.<br />

Stability and storage condition:<br />

Eudragit S 100 and L 100 polymers are stable at room temperature.<br />

Safety: Acute toxicity studies have been performed in rats, rabbits and dogs. No<br />

toxic effects were observed. Chronictoxicity studies were performed in rats over a<br />

period of 3 months. No significant changes were found in the animal organs.<br />

Applications in Pharmaceutical Formulation or Technology:<br />

Eudragit L 100 and S 100 are employed as film coating agents resistant<br />

to gastric fluid with solubility above pH 6.0 and 7.0 respectively, for enteric<br />

coating of formulations.<br />

Comments:<br />

For spray coating, the lacquer solutions and dispersions and must be<br />

thinned with suitable solvents. Suitable solvents are ethanol, isopropyl alcohol,<br />

diethyl ether, acetone, methylene chloride and water. Suitable plasticizers are<br />

glyceryl triacetate, phthalic acid esters, polyethyiene glycols, triacetin, dibutyl<br />

phthalate and citric acid esters.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

57


2. CELLULOSE ACETATE PHTHALATE 73,74,75<br />

Review Of Literature<br />

Cellulose Acetate phthalate is cellulose, some of the hydroxyl groups of<br />

which are esterified by acetyl groups and others by hydrogen phthaloyl groups.<br />

Nonproprietary: Cellacephate; Cellacefate<br />

Synonyms: CAP, Cellulose acetophthalate<br />

Category: Pharmaceutical aid (for enteric coating of tablets).<br />

Structure:<br />

Description:<br />

White, free-flowing powder or colorless flakes; odorless or with a faint<br />

odor of acetic acid; hygroscopic<br />

Solubility:<br />

Freely soluble in acetone; soluble in diethylene glycol and in dioxan;<br />

practically insoluble in water, in ethanol (95), in toluene and in chlorinated and<br />

non-chlorinated aliphatic hydrocarbons. It dissolves in dilute solutions of alkalis.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

58


Storage:<br />

Viscosity:<br />

Review Of Literature<br />

Store in tightly-closed containers at a temperature between 8 and 15 o C.<br />

A 15% solution in acetone with a moisture content of 0.4% has viscosity<br />

of 50-90 cps. This is a good coating solution with honey-like consistency, but the<br />

viscosity is influenced by the purity of the solvent.<br />

Incompatibilities:<br />

CAP is incompatible with ferrous sulfate, ferric chloride, silver nitrate,<br />

sodium citrate, aluminum sulfate, calcium chloride, mercuric chloride, barium<br />

nitrate, basic lead acetate, strong alkalis and acids.<br />

Applications in pharmaceutical formulation:<br />

CAP is used as an enteric coating material for tablets or capsules. Such<br />

coating resists simulated gastric fluid, and dissolves in the intestinal fluid. The<br />

addition of plasticizers improves the water resistance of this coating material and<br />

such plasticizers are more effective than when CAP is used alone.<br />

Comments:<br />

The CAP should always be added to solvent; not the reverse.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

59


3. HYDROXYPROPYL METHYLCELLULOSE 74,76<br />

Non-proprietary names:<br />

Review Of Literature<br />

Hypomellus, hydroxyl propyl methylcellulose 2208, 2906, 2910.<br />

Synonyms: Methyl hydroxypropylcellulose, Propylene glycol ether of<br />

methylcellulose, methylcellulose propylene glycol ether.<br />

Chemical Name & CAS Registry number: Cellulose, 2-hydroxypropylmethyl<br />

ether Cellulose Hydroxypropylmethylether [9004-65-3].<br />

Empirical formula : C8H15 – (C10H18O6) n – C8 H15 O5<br />

Molecular weight : Approximately 86,000<br />

Structure:<br />

Description: An odourless, tasteless, whit or creamy coloured fibrous or granular<br />

powder.<br />

Functional category:<br />

Coating agent, film former, tablet binder, stabilizing agent, suspending<br />

agent, viscosity agent and emulsion stabilizer.<br />

Apparent Density: 0.25 – 0.75 g/cm 3<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

60


Solubility:<br />

Review Of Literature<br />

Soluble in cold water forming viscous colloidal solution, insoluble in<br />

chloroform, alcohol and ether, but soluble in mixture of methanol and methylene<br />

chloride.<br />

Viscosity: HPMC K4M: 4,000 cps<br />

Stability and Storage Condition:<br />

Very stable in dry conditions, Solutions are stable at pH 3.0 – 11.0. Store<br />

in a tight container, in a cool place.<br />

Incompatibility: Extreme pH conditions, oxidizing materials.<br />

Safety: Human and animal feeding studies have shown Hydroxypropyl<br />

methylcellulose to be safe.<br />

4. GUAR GUM 74,77<br />

Synonyms: Guar flour, Buratoin V-7-E; Supercol Guar Gums; jaguar.<br />

Functional Category:<br />

USP : Tablet binder; suspending and/or viscosity- increasing agent<br />

Other : Tablet disintegrant.<br />

Chemical Name & CAS Registry number:<br />

Chemical type : Galactomannan polysaccharide [9000-30-0]<br />

Botanical origin : Cyamopsis tetragonolobus (L) Taub.<br />

Empirical formula: (C6H12O6) n<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

61


Structure:<br />

Description: White to yellowish- white powder. or nearly so with<br />

Physical state and appearance: Solid.<br />

Odour : Odorless<br />

Taste : bland test.<br />

Melting Point : Decomposes.<br />

Review Of Literature<br />

Properties: It forms viscous colloidal dispersion when hydrated in cold water.<br />

Incompatibility: Acetone, alcohol, tannins, strong acids and alkalis, borate ions,<br />

if present in the dispersing water, will prevent hydration of guar gum. The<br />

addition borate ions to hydrated solutions produce cohesive gels, which prevents<br />

further hydration. The gel can be liquefied by reducing the pH below 7.<br />

Safety:<br />

Guar gum is recognized by the FDA as a substance added directly to<br />

human food and has been affirmed as generally as safe. Specific limits in yours<br />

foods are established; however alkylated guar gums are not covered by<br />

appropriate FDA regulations.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

62


5. SODIUM ALGINATE 74,78<br />

Review Of Literature<br />

Synonyms: Alginc acid; Sodium salt; sodium polymannuronate satialgine S 20;<br />

Album S 160 and S 15/600; Kelcosol, Keltone, Kelco-gel LV, HV; Sodium<br />

Alginate.<br />

Functional Category:<br />

binder.<br />

Suspending and/or viscosity- increasing agent, disintegrating agent; tablet<br />

Chemical Name & CAS Registry number:<br />

Sodium alginate is the purified carbhydrate product extracted form brown<br />

sea weed by the use of dilute alkali. It consists chiefly of the sodium salt of alginic<br />

acid, a polyuronic acid composed of β-D-mannuronic acid, residues linked so that<br />

the carboxyl group of each unit is free while the aldehyde group is shielded by a<br />

glycosidic linkage. [9005-38-3]<br />

Structure:<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

63


Description:<br />

Physical state and appearance: Solid. (Powdered solid.)<br />

Odor : Odorless.<br />

Taste : Tasteless.<br />

Color : White to Off-white (cream)<br />

Boiling Point : Not available.<br />

Melting Point : >300°C (572°F)<br />

Solubility:<br />

Review Of Literature<br />

It is slowly soluble in water, forming a viscous, colloidal solution. It is<br />

insoluble in alcohol and in hydro-alcoholic solutions in which the alcohol content<br />

is grater than 30% by weight. It is also insoluble in other organic solvents.<br />

Stability and Storage conditions:<br />

It is hygroscopic. Dry storage stability is excellent when the powder is<br />

stored in a well-closed container at temperatures of 25 o C or less.<br />

Precautions:<br />

Keep away from heat. Keep away from sources of ignition. Empty<br />

containers pose a fire risk, evaporate the residue under a fume hood. Ground all<br />

equipment containing material. Do not ingest. Do not breathe dust. If ingested,<br />

seek medical advice immediately and show the container or the label. Keep away<br />

from incompatibles such as oxidizing agents, acids, alkalis.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

64


Review Of Literature<br />

Storage: Keep container tightly closed. Keep container in a cool, well-ventilated<br />

area.<br />

Safety:<br />

The incorporation of 5 to 15% sodium alginate in the diet of purebreed<br />

beagle dogs for one year caused no harmful effects. It is found not allergenic.<br />

6. ETHYL CELLULOSE 74,79<br />

Synonyms : Ethyl cellulose; Ehocel.<br />

Functional Category : Tablet binder; coating agent.<br />

Chemical Name & CAS Registry number:<br />

Structure:<br />

Cellulose, ethyl ether<br />

Cellulose ethyl ether [9004-57-3]<br />

Description: A tasteless, free-flowing, white to light tan powder.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

65


Solubility:<br />

Review Of Literature<br />

Insoluble in water, glycerine and propylene glycol, but soluble in varying<br />

degrees in certain organic solvents, depending upon the ethoxyl content. The<br />

addition of 10-20% of a lower aliphatic alcohol to solvents, such as ketons, esters<br />

and hydrocarbons, can improve the solubility.<br />

Stability and Storage conditions:<br />

It is resistant to alkalis, both dilute and concentrated and to salt solutions.<br />

It is more sensitive to acidic materials than are cellulose esters. It should be stored<br />

between 7 o and 32 o C in a dry area away from all sources of heat. Store in a well<br />

closed container.<br />

Incompatibility: Incompatible with paraffin wax and microcrystalline.<br />

Safety: Essentially non-toxic.<br />

Application in pharmaceutical formulation or technology:<br />

Binder in tablet, coating materials for tablets, coating material for<br />

stabilization, coating to prevent unpleasant taste, thickness agent in creams,<br />

lotions and gels.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

66


4.1 Materials<br />

follows:<br />

Sr.<br />

No.<br />

CHAPTER-4<br />

<br />

Methodology<br />

The following materials/chemicals were used in the work which are as<br />

Table-5: Materials / Chemicals used<br />

Materials/ Chemicals Grade Name Of Supplier<br />

1. Flurbiprofen -<br />

2. Eudragit L-100 Degussa India Pvt. Ltd., Mumbai<br />

3. Eudragit S-100 Degussa India Pvt. Ltd., Mumbai<br />

4. Guar gum L.R. S.D. Fine Chem. Ltd., Mumbai.<br />

5. Sodium alginate L.R. S.D. Fine Chem. Ltd., Mumbai.<br />

6. HPMC (K4M L.R. Ozone international Mumbai.<br />

7. Cellulose acetate phthalate L.R. Sisco Research Lab. Pvt. Mumbai<br />

8. Ethyle cellulose L.R. Loba Chemical, India.<br />

9. Dibutyl phthalate L.R. S.D. Fine Chem. Ltd., Mumbai.<br />

10. Span-80 L.R. Loba Chemical, India.<br />

11. Acetone L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

12. Liquid paraffin(heavy) L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

13. Petrolium ether 60-80 C L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

14. Potassium dihydrogen<br />

phosphate<br />

L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

15. Sodium hydroxide L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

16. Potassium Chloride L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

17. Hydrochloric acid L.R. S.D. Fine Chem. Ltd. Mumbai.<br />

18. Empty gelatin capsules - -<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

67


4.2 Equipments<br />

The equipments used in the present work are as follows:<br />

Table-6: Equipments used and source<br />

Sr. No. Instruments Source<br />

1. Electronic balance<br />

2. UV/Visible spectrophotometer<br />

Methodology<br />

Shimadzu Corporation--BL-220H,<br />

Japan<br />

Shimadzu 1700, Shimadzu<br />

Corporation. Japan<br />

3. Scanning electron microscopy JEOL–6360A, Japan.<br />

4. FTIR spectrophotometer<br />

Shimadzu, Shimadzu Corporation.<br />

Japan<br />

5. Magnetic stirrer Remi Motor Equipments, Mumbai<br />

6.<br />

Electrolab dissolution apparatus<br />

(USPXXIII)<br />

7. Oven<br />

Electro lab.<br />

Shital Scientific Industries,<br />

Mumbai<br />

8. pH meter Elico II-122<br />

9. Distillation unit Biotech India, Mumbai<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

68


4.3 Evaluation of Flurbiprofen<br />

4.3.1 Preparation of Calibration Curve in Ethanol<br />

Methodology<br />

An accurately weighted amount of Flurbiprofen equivalent to 100 mg was<br />

dissolved in 100ml of ethanol. A series of standard solution containing Beer’s<br />

Lambert’s range of concentration from 2 to 16 µg/ml of Flurbiprofen were<br />

prepared and absorbance was measured at 247 nm against reagent blank. All<br />

spectral absorbance measurement was on Shimadzu 1700 UV-visible<br />

spectrophotometer.<br />

4.3.2 Preparation of Calibration Curve in 1.2 pH buffer<br />

An accurately weighted amount of Flurbiprofen equivalent to 100 mg was<br />

dissolved in small volume of ethanol, in 100 ml volumetric flask and the volume<br />

was adjusted to 100 ml with 1.2 pH buffer and further dilution were made with 1.2<br />

pH buffer. A series of standard solution containing Beer’s Lambert’s range of<br />

concentration from 2 to 16 µg/ml of Flurbiprofen were prepared and absorbance<br />

was measured at 247 nm against reagent blank. All spectral absorbance<br />

measurement was on Shimadzu 1700 UV-visible spectrophotometer.<br />

4.3.3 Preparation of Calibration Curve in 6.8 pH phosphate buffer<br />

An accurately weighted amount of Flurbiprofen equivalent to 100 mg was<br />

dissolved in small volume of ethanol, in 100 ml volumetric flask and the volume<br />

was adjusted to 100 ml with 6.8 pH phosphate buffer and further dilution were<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

69


Methodology<br />

made with 6.8 pH phosphate buffer. A series of standard solution containing<br />

Beer’s Lambert’s range of concentration from 2 to 16 µg/ml of Flurbiprofen were<br />

prepared and absorbance was measured at 247 nm against reagent blank. All<br />

spectral absorbance measurement was on Shimadzu 1700 UV-visible<br />

spectrophotometer.<br />

4.3.4 Preparation of Calibration Curve in 7.4 pH phosphate buffer<br />

An accurately weighted amount of Flurbiprofen equivalent to 100 mg was<br />

dissolved in small volume of ethanol, in 100 ml volumetric flask and the volume<br />

was adjusted to 100 ml with 7.4 pH phosphate buffer and further dilution were<br />

made with 7.4 pH phosphate buffer. A series of standard solution containing<br />

Beer’s Lambert’s range of concentration from 2 to 16 µg/ml of Flurbiprofen were<br />

prepared and absorbance was measured at 247 nm against reagent blank. All<br />

spectral absorbance measurement was on Shimadzu 1700 UV-visible<br />

spectrophotometer.<br />

4.4. Microcapsulation of Flurbiprofen<br />

4.4.1 Rationale for selection of ingredients and process: 39,40<br />

From literature review, it was evident that the pH in the proximal colon<br />

ranges from 6.6 to 7.0. So the Eudragit L-100 and S-100 were combined in<br />

different ratios and solubility of these combinations was found that Eudragit L-<br />

100 and S-100 in the ratio 1:2 was soluble in pH range of 6.6 to 7.0. Hence this<br />

combination was selected for preparation of microcapsules<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

70


Methodology<br />

Pure acetone did not dissolve Eudragit; however acetone with 2% water fitted<br />

the criterion well. Liquid paraffin was used was used as the dispersion media or<br />

external phase. Petroleum ether was used to clean the microparticles since it<br />

removes liquid paraffin without the integrity of the microparticles.<br />

Solvent evaporation O/O (oil in oil) emulsification method of<br />

microencapsulation was chosen since it yields more uniform particles. The<br />

method is correctly referred as O/O instead of W/O (water in oil) since a<br />

polymeric solution in organic solvent is considered as oil in microencapsulation<br />

terminology.<br />

4.4.2 Preparation of Flurbiprofen microcapsule: 80,81<br />

Emulsification-solvent evaporation.<br />

Accurately weighted amount of Drug, Eudragit L-100 and S-100 as shown<br />

in Table-7 were dissolved in 50ml of acetone to form a homogenous polymers<br />

solution. Core material, i.e. Flurbiprofen was dispersed in it and mixed<br />

thoroughly. This organic phase was slowly poured at 15°C into liquid paraffin<br />

(100 ml) containing 1% (w/w) of Span -80 with stirring at 1000 rpm to form a<br />

uniform emulsion. Thereafter, it was allowed to attain room temperature and<br />

stirring was continued until residual acetone evaporated and smooth-walled, rigid<br />

and discrete microcapsules were formed. The microcapsules were collected by<br />

decantation and the product was washed with petroleum ether (40 –60°C) and<br />

dried at room temperature for 3 hrs. The microcapsules were then stored in a<br />

desiccators over fused calcium chloride.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

71


Sl.<br />

No.<br />

Table-7: Formulation of Flurbiprofen Microcapsules using Eudragit – L 100 and Eudragit – S100<br />

Ingredients<br />

Formulation Code<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Methodology<br />

FM-1 FM-2 FM-3 FM-4<br />

1. Drug (mg) 1000 1000 1000 1000<br />

2. Eudragit L-100 (mg) 166 333 500 666<br />

3. Eudragit S-100 (mg) 332 666 1000 1332<br />

4. Span-80 (ml) 0.5 0.5 0.5 0.5<br />

5. Acetone (ml) 50 50 50 50<br />

72


4.5 Evaluation of Flurbiprofen Microcapsule<br />

parameters.<br />

Methodology<br />

Prepared microcapsules of Flurbiprofen were evaluated for the following<br />

4.5.1 Particle size 82<br />

Determination of average particle size of Flurbiprofen microcapsule was<br />

carried out by using optical microscopy. A minute quantity of microcapsules was<br />

spread on a clean glass slide and average size of 600 microcapsules was<br />

determined in each batch.<br />

4.5.2 Study of flow properties of microcapsules 83<br />

Angle of repose (θ) of the microspheres, which measures the resistance to<br />

particle flow, was determined by a fixed funnel method. The height of the funnel<br />

was adjusted in such a way that the tip of the funnel just touches the heap of the<br />

blends. Accurately weighed microspheres were allowed to pass through the funnel<br />

freely on to the surface. The height and diameter of the powder cone was<br />

measured and angle of repose was calculated using the following equation<br />

θ = tan -1<br />

h<br />

r<br />

Where h/r is the surface area of the free standing height of the<br />

microspheres heap that is formed on a graph paper after making the microspheres<br />

flow from the glass funnel.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

73


4.5.3 Percentage yield 84<br />

Methodology<br />

The measured weight was divided by total amount of all non-volatile<br />

components which were used for the preparation of microcapsule.<br />

% yield =<br />

Actual weight of product<br />

Total weight of excipient and drug<br />

4.5.4 Drug Content Uniformity 85<br />

x 100<br />

In 100ml volumetric flask 25mg of crushed microcapsules were taken and<br />

dissolved with small quantity of ethanol and the volume was made up to mark<br />

with pH 6.8 and stirred for 12 hrs. After stirring the solution was filtered through<br />

whatman filter paper and from the filtrate appropriate dilutions were made and<br />

absorbance was measured at 247 nm by using Shimadzu 1700 UV-<br />

spectrophotometer.<br />

4.5.5 Scanning Electron Microscopy 86<br />

It has been used to determine particle size distribution, surface topography,<br />

texture and examine the morphology of fractured or sectioned surface.<br />

The samples for SEM analysis were prepared by following method. Dry<br />

microcapsules brass stub a coated with gold in an ion sputter. Then picture of<br />

microcapsules were taken by random scanning of the stub. The SEM analysis or<br />

the microcapsules was carried out by using JEOL–6360A analytical scanning<br />

electron microscope. The microcapsules were viewed at an accelerating voltage of<br />

20KV.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

74


4.5.6 Drug-polymer Interaction 87<br />

Methodology<br />

FT-IR spectra of the Flurbiprofen, FM3 formulation, Eudragit L-100,<br />

Eudragit S-100 were determined by using KBr pellet technique. Samples were<br />

scanned over the 500-4000cm -1 Spectral region at a resolution of 4cm -1 . The ratio<br />

of the sample in KBr disc was 1% (shimadzu FT-IR spectrometer). To ensure no<br />

interaction has been occurred between the drug and polymers.<br />

4.5.7 In-vitro dissolution study 38,80<br />

In vitro dissolution profile of each formulation was determined by<br />

employing USP XXIII rotating basket method (900 ml of p H 6.8-phosphate<br />

buffer, 100 rpm and 37±0.5 o C). Microcapsules equivalent to 150 mg of<br />

Flurbiprofen was loaded into the basket of the dissolution apparatus. Dissolution<br />

study carried out for 12 hrs. 5 ml of the sample was withdrawn from the<br />

dissolution media at suitable time intervals and the same amount was replaced<br />

with fresh buffer. The absorbance was measured at 247 nm by using Shimadzu<br />

1700 UV–Vis spectrophotometer, against pH 6.8 as blank.<br />

4.6 Preparation of Cross-Linked Gelatine Capsules<br />

Formaldehyde treatment<br />

38, 39<br />

Formalin treatment has been employed to modify the solubility of gelatine<br />

capsules. Exposure to formalin vapours results in an unpredictable decreases in<br />

solubility of gelatine owing to the crosslinkage of the amino group in the gelatine<br />

molecular chain aldehyde group of formaldehyde by Schiff’s base condensation.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

75


Methodology<br />

Procedure: Hard gelatine capsule of size 00 and 100 in number taken. Their<br />

bodies were separated from the caps. 25 ml of 15% (v/v) formaldehyde was taken<br />

into desiccators and a pinch of potassium permanganate was added to it, to<br />

generate formalin vapours. The wire mesh containing the empty bodies of capsule<br />

was then exposed to formaldehyde vapours. The caps were not exposed leaving<br />

them water-soluble. The desiccators were tightly closed. The reaction was carried<br />

out for 12 h after which the bodies were removed and dried at 50 O C for 30 min to<br />

ensure completion of reaction between gelatine and formaldehyde vapours. The<br />

bodies were then dried at room temperature to facilitate removal of residual<br />

formaldehyde.<br />

polythene bag.<br />

These capsule bodies were capped with untreated caps and stored in a<br />

4.7 Tests for Formaldehyde Treated Empty Capsules<br />

Various physical and chemical test were carried out simultaneously for<br />

formaldehyde treated and untreated capsules.<br />

4.7.1 Physical tests:<br />

Identification attributes: The ‘00’ capsule were one with a red cap and red<br />

colored body. They were lockable type, odorless, softy and sticky when<br />

treated with wet fingers. After formaldehyde treatment, there were no<br />

significant changes in the capsules. They were non-tacky when touched with<br />

wet fingers.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

76


Methodology<br />

Visual defect: In about 100 capsule bodies treated with formaldehyde, about<br />

ten were found to be shrunk or distorted.<br />

Dimensions: Variations in dimensions between formaldehyde, treated and<br />

untreated capsules were studied. The length and diameter of the capsules were<br />

measured before and after formaldehyde treatment, using dial caliper.<br />

Solubility studies of treated capsules: The solubility test was carried out for<br />

normal capsules and formaldehyde treated capsules for 24 hrs. Ten capsules<br />

were randomly selected and then subjected to solubility studies at room<br />

temperatures in buffers of pH 1.2, 7.4 and 6.8. 100 ml solution and stirred for<br />

24 hrs. The time at which the capsule dissolves or forms a soft fluffy mass was<br />

noted.<br />

4.7.2 Chemical test:<br />

Qualitative test for free formaldehyde: 73<br />

Standard formaldehyde solution used is formaldehyde solution (0.002,<br />

w/v) and sample solution is formaldehyde treated bodies (about 25 in number)<br />

were cut into small pieces and taken into a beaker containing distilled water. This<br />

was stirred for 1 hrs with a magnetic stirrer, to solubilize the free formaldehyde.<br />

The solution was then filtered into a 50 ml volumetric flask, washed with distilled<br />

water and volume was made up to 50 ml with the washings.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

77


Procedure:<br />

Methodology<br />

1ml of sample solution, 9ml of water was added. One millilitre of resulting<br />

solution was taken into a test tube and mixed with 4ml of water and 5ml of<br />

acetone reagent. The test tube was warmed in a water bath at 40 o C and allowed to<br />

stand for 40 min. The solution was not more intensely colored than a reference<br />

solution prepared at the same time and in the same manner using 1ml of standard<br />

solution in place of the sample solution. The comparison should be made by<br />

examining tubes down their vertical axis.<br />

4.8 Formulation of Pulsatile (modified pulsincap) Drug Delivery<br />

System: 18,38,39,88<br />

Microcapsule equivalent to 150mg of Flurbiprofen were accurately weighted<br />

and filled into the previously formaldehyde treated bodies by hand filling.<br />

The bodies containing the microsphere were then plugged with different<br />

amounts of polymers like guar gum, hydroxylpropylmethylcellulose and<br />

sodium alginate as shown in Table-8. Then join the capsule body and cap and<br />

sealed with a small amount of the 5% ethyl cellulose ethanolic solution.<br />

The sealed capsules were completely coated with 5% Cellulose Acetate<br />

Phthalate (CAP) to prevent variable gastric emptying. The whole system thus<br />

produced is modified pulsincap.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

78


4.8.1 Coating of pulsincap<br />

Methodology<br />

5 % w/w solution of CAP was prepared by using acetone: ethanol (8.:2) as a<br />

solvent and dibutyl phthalate as plasticizer (0.75%) as a plasticizer. Dip coating<br />

method was fallowed to develop the pulsincap. The capsules were alternatively<br />

dipped in 5 % CAP solution and dried. Coating was repeated until an expected<br />

weight gain of.8-12% was obtained and the capsule resists disintegration in 0.1 N<br />

HCL for a minimum period of 2 hrs.<br />

Fig. 6: Overview of designed pulsatile device<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

79


Formulation<br />

code<br />

Table- 8: Composition for modified pulsatile device on the basis of design summary<br />

Wt.of empty<br />

body<br />

(mg*)<br />

Wt.of micro<br />

capsule (mg)<br />

Polymer used<br />

Wt.of polymer<br />

used (mg)<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Total weight<br />

with cap (mg)<br />

Methodology<br />

Wt. after CAP<br />

coating (mg)<br />

F-1 68.8 355 Guar gum 20 443.8 454.7<br />

F-2 67.9 355 Guar gum 30 452.9 462<br />

F-3 68.5 355 Guar gum 40 463.5 470.18<br />

F-4 67.5 355 HPMC 20 442.5 451.34<br />

F-5 67.4 355 HPMC 30 452.4 460.8<br />

F-6 68.5 355 HPMC 40 463.5 473.25<br />

F-7 68.0 355 Sod. Alg. 20 443.0 454<br />

F-8 67.7 355 Sod. Alg. 30 452.7 462.35<br />

F-9 67.6 355 Sod. Alg. 40 462.6 471.95<br />

HPMC: Hydroxy Propyl Methylcellulose.<br />

Sod.Alg: Sodium Alginate<br />

* Microcapsule equivalent to 150 mg of drug used<br />

80


4.9 Evaluation of Modified Pulsincap 39,40<br />

4.9.1 Thickness of cellulose acetate phthalate coating<br />

Methodology<br />

The thickness of cellulose acetate phthalate coating was measured using<br />

screw gauge and expressed in mm.<br />

4.9.2 Weight variation<br />

10 capsules were selected randomly from each batch and weight<br />

individually for weight variation.<br />

4.9.3 Drug Polymer Interaction<br />

FT-IR spectra of physical mixture of Flurbiprofen+Guargum,<br />

Flurbiprofen+HPMC, Flurbiprofen+Sodium alginate was carried out by using KBr<br />

pellet technique. Samples were scanned over the 500-4000cm -1 Spectral region at<br />

a resolution of 4cm -1 . The ratio of the sample in KBr disc was 1% (shimadzu FT-<br />

IR spectrometer).<br />

4.9.4 In vitro release profile<br />

Dissolution studies were carried out by using USP XXIII dissolution<br />

test apparatus (Basket) method. Capsules were placed in a basket so that the<br />

capsule should be immersed completely in dissolution media but not float. In<br />

order to simulate the pH changes along the GI tract, three dissolution media with<br />

pH 1.2, 7.4 and 6.8 were sequentially used referred to as sequential pH change<br />

method. When performing experiments, the pH 1.2 medium was first used for 2<br />

hrs (since the average gastric emptying time is 2 hrs) then removed and the fresh<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

81


Methodology<br />

pH 7.4 phosphate buffer saline (PBS) was added. After 3 hrs (average small<br />

intestinal transit time is 3 hrs) the medium was removed and fresh pH 6.8<br />

dissolution medium was added for subsequent hrs. 900ml of the dissolution<br />

medium was used at each time. Rotation speed was 100 rpm and temperature was<br />

maintained at 37±0.5 ◦C. five millilitres of dissolution media was withdrawn at<br />

predetermined time intervals and fresh dissolution media was replaced. The<br />

withdrawn samples were analyzed at 247 nm, by UV absorption spectroscopy.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

82


CHAPTER-5<br />

5.1 Evaluation of Flurbiprofen<br />

<br />

<br />

Standard Calibration Curve of Flurbiprofen<br />

Solvent………………………………….Ethanol<br />

Wavelength……………………………..247nm<br />

Unit for concentration………………......mcg/ml<br />

Table-9: Standard calibration data of Flurbiprofen in Ethanol<br />

SI.NO.<br />

Concentration<br />

(mcg/ml)<br />

Absorbance<br />

(nm)<br />

1 0.000 0.000<br />

2 2.000 0.118<br />

3 4.000 0244<br />

4 6.000 0.371<br />

5 8.000 0.496<br />

6 10.000 0.609<br />

7 12.000 0734<br />

8 14.000 0858<br />

9 16.00 0.976<br />

*Average of triplicate sets<br />

Fig-7: Standard calibration curve of Flurbiprofen in Ethanol<br />

<br />

<br />

<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

<br />

<br />

<br />

<br />

<br />

83


5.2 Evaluation of Flurbiprofen<br />

<br />

Standard Calibration Curve of Flurbiprofen<br />

Solvent………………………….............pH 1.2 buffer<br />

Wavelength……………………………..247nm<br />

Unit for concentration………………......mcg/ml<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

Table-10: Standard calibration data of Flurbiprofen in pH 1.2 buffer<br />

SI.NO.<br />

Concentration<br />

(mcg/ml)<br />

Absorbance<br />

(nm)<br />

1 0.000 0.000<br />

2 2.000 0.085<br />

3 4.000 0.165<br />

4 6.000 0.242<br />

5 8.000 0.335<br />

6 10.000 0.419<br />

7 12.000 0.505<br />

8 14.000 0.583<br />

9 16.00 0.654<br />

*Average of triplicate sets<br />

Fig-8: Standard calibration curve of Flurbiprofen in pH 1.2 buffer<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

84


5.3 Evaluation of Flurbiprofen<br />

<br />

Standard Calibration Curve of Flurbiprofen<br />

Solvent………………………….............pH 6.8 phosphate buffer<br />

Wavelength……………………………..247nm<br />

Unit for concentration………………......mcg/ml<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

Table-11: Standard calibration data of Flurbiprofen in pH 6.8 buffer<br />

SI.NO.<br />

Concentration<br />

(mcg/ml)<br />

Absorbance<br />

(nm)<br />

1 0.000 0.000<br />

2 2.000 0.078<br />

3 4.000 0.152<br />

4 6.000 0.228<br />

5 8.000 0.308<br />

6 10.000 0.386<br />

7 12.000 0.454<br />

8 14.000 0.538<br />

9 16.00 0.612<br />

*Average of triplicate sets<br />

Fig-9: Standard calibration curve of Flurbiprofen in pH 6.8 buffer<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

85


5.4 Evaluation of Flurbiprofen<br />

<br />

Standard Calibration Curve of Flurbiprofen<br />

Solvent………………………….............pH 7.4 phosphate buffer<br />

Wavelength……………………………..247nm<br />

Unit for concentration………………......mcg/ml<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

Table-12: Standard calibration data of Flurbiprofen in pH 7.4 buffer<br />

SI.NO.<br />

Concentration<br />

(mcg/ml)<br />

Absorbance<br />

(nm)<br />

1 0.000 0.000<br />

2 2.000 0.115<br />

3 4.000 0.23<br />

4 6.000 0.345<br />

5 8.000 0.455<br />

6 10.000 0.576<br />

7 12.000 0.688<br />

8 14.000 0.803<br />

9 16.00 0.917<br />

*Average of triplicate sets<br />

Fig-10: Standard calibration curve of Flurbiprofen in pH 7.4 buffer<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

86


Fig-11 Image showing microcapsule of Flurbiprofen formulations<br />

(A) FM-1 (B) FM-2 (C) FM-3 (D) FM-4<br />

A B<br />

C D<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

87


5.5 Evaluation of Flurbiprofen Microcapsules:<br />

5.5.1 Micrometric properties<br />

Table-13: Micromeritic properties of Flurbiprofen Microcapsules<br />

Formulation code Mean particles size (m) Angle of repose<br />

FM-1 163.68 1.14 18° 80” 3.89<br />

FM-2 167.32 1.01 18° 55” 1.93<br />

FM-3 196.77 3.49 21° 39” 2.78<br />

FM-4 251.84 2.01 22° 76” 3.39<br />

All values are represented as mean standard deviation (n=3)<br />

5.5.2 Percentage yield and drug content<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

Table-14: Percentage yield and Drug content of Flurbiprofen microcapsules<br />

Formulation code Percentage yield Drug content uniformity<br />

FM-1 87.21 0.50 82.89% 0.68<br />

FM-2 88.41 0.37 89.23% 0.85<br />

FM-3 90. 24 0.08 95.59% 0.68<br />

FM-4 85.85 0.29 75.9% 0.78<br />

All values are represented as mean standard deviation (n=3)<br />

88


5.5.3 Scanning Electron Microscopy (SEM)<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

Morphology of microcapsules was examined by scanning electron<br />

microscopy. The view of the microcapsules showed smooth surface morphology<br />

exhibited range of sizes within each batch. The outer surface of microcapsules<br />

was smooth and dense, while the internal surface was porous. The shell of<br />

microcapsules also showed some porous structure it may be caused by<br />

evaporation of solvent entrapped within the shell of microcapsules after forming<br />

smooth and dense layer.<br />

Fig. 12: Scanning electron microphotographs of FM-3 formulation.<br />

A B<br />

C D<br />

89


5.5.4 Infrared spectroscopy:<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

The FT-IR spectra study showed no change in the finger print of pure drug<br />

spectra, thus confirming absence of drug and polymer interaction. The result<br />

shown in Fig. 13 to 156<br />

Fig. 13: I.R. Spectrum of Flurbiprofen (pure drug)<br />

Fig. 14: I.R. Spectrum of Eudragit L-100<br />

90


Fig. 15: I.R. Spectrum of Eudragit S-100<br />

Fig. 16: I.R. Spectrum of Flurbiprofen microcapsules (FM-3)<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

91


Time (T)<br />

Hrs<br />

5.5.5 Evaluation of In-vitro drug release of Prepared Flurbiprofen Microcapsules<br />

Square root<br />

of Time<br />

Table-15: In-vitro release profile of Flurbiprofen microcapsules for FM-1<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

0.5 0.70 -0.301 43.93 29.29 ± 1.48 70.71 1.466 1.849<br />

1 1.00 0.000 52.14 34.96 ± 1.41 65.04 1.543 1.813<br />

2 1.41 0.301 62.82 41.88 ± 1.50 58.12 1.622 1.764<br />

3 1.73 0.477 72.26 48.17 ± 0.36 51.83 1.682 1.714<br />

4 2.00 0.602 84.62 56.41 ± 0.70 43.59 1.751 1.639<br />

5 2.23 0.698 92.85 60.92 ± 1.20 39.08 1.784 1.591<br />

6 2.44 0.778 96.15 64.10 ± 0.84 35.90 1.806 1.555<br />

7 2.64 0.845 102.3 68.22 ± 0.71 31.79 1.833 1.502<br />

8 2.82 0.903 112.2 74.83 ± 1.42 25.17 1.874 1.400<br />

9 3.00 0.954 119.1 79.42 ± 0.53 20.58 1.899 1.313<br />

10 3.16 1.000 128.5 85.17 ± 0.58 14.29 1.933 1.155<br />

11 3.31 1.041 136.3 90.91 ± 0.85 9.090 1.958 0.958<br />

12 3.46 1.079 143.3 95.58 ± 0.24 4.420 1.980 0.645<br />

All values are represented as mean standard deviation (n=3)<br />

92


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table-16: In-vitro release profile of Flurbiprofen microcapsules for FM-2<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

0.5 0.70 -0.301 41.14 27.43 ± 1.88 72.57 1.438 1.860<br />

1 1.00 0.000 53.38 35.58 ± 0.35 64.42 1.551 1.809<br />

2 1.41 0.301 60.73 40.48 ± 1.50 59.52 1.607 1.774<br />

3 1.73 0.477 67.40 44.93 ± 0.46 55.07 1.652 1.740<br />

4 2.00 0.602 72.33 48.22 ± 0.98 51.78 1.683 1.714<br />

5 2.23 0.698 81.46 54.31 ± 0.61 45.69 1.734 1.659<br />

6 2.44 0.778 94.64 63.09 ± 2.35 36.91 1.799 1.567<br />

7 2.64 0.845 101.9 67.99 ± 2.16 32.01 1.832 1.505<br />

8 2.82 0.903 113.1 75.45 ± 1.54 24.55 1.877 1.390<br />

9 3.00 0.954 125.5 83.69 ± 1.06 16.13 1.922 1.212<br />

10 3.16 1.000 133.5 89.05 ± 1.07 10.95 1.949 1.039<br />

11 3.31 1.041 136.1 90.76 ± 1.35 9.240 1.957 0.965<br />

12 3.46 1.079 138.7 91.96 ± 0.51 8.040 1.963 0.905<br />

All values are represented as mean standard deviation (n=3)<br />

93


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table-17: In-vitro release profile of Flurbiprofen microcapsules for FM-3<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

0.5 0.70 -0.301 40.44 26.96 ± 0.15 73.04 1.430 1.863<br />

1 1.00 0.000 45.10 30.07 ± 1.16 69.93 1.478 1.844<br />

2 1.41 0.301 52.90 35.27 ± 0.81 64.73 1.547 1.811<br />

3 1.73 0.477 61.65 41.10 ± 0.13 58.90 1.613 1.770<br />

4 2.00 0.602 70.60 47.08 ± 0.83 52.92 1.672 1.723<br />

5 2.23 0.698 80.19 53.16 ± 0.97 46.54 1.728 1.667<br />

6 2.44 0.778 89.39 59.60 ± 0.25 40.4 1.775 1.606<br />

7 2.64 0.845 94.53 63.02 ± 0.26 36.98 1.799 1.567<br />

8 2.82 0.903 102.0 66.37 ± 0.41 33.63 1.821 1.526<br />

9 3.00 0.954 107.2 71.49 ± 0.25 28.51 1.854 1.406<br />

10 3.16 1.000 119.2 79.50 ± 0.47 20.5 1.900 1.311<br />

11 3.31 1.041 124.6 83.06 ± 0.89 16.93 1.919 1.228<br />

12 3.46 1.079 133.3 88.87 ± 1.68 11.13 1.948 1.046<br />

All values are represented as mean standard deviation (n=3)<br />

94


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table-18: In-vitro release profile of Flurbiprofen microcapsules for FM-4<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.00 0.000 0.000<br />

0.5 0.70 -0.301 37.05 24.70 ± 0.46 75.3 1.392 1.876<br />

1 1.00 0.000 46.15 30.77 ± 0.39 69.23 1.488 1.840<br />

2 1.41 0.301 54.77 36.36 ± 0.39 63.64 1.560 1.803<br />

3 1.73 0.477 62.28 41.52 ± 0.19 58.48 1.618 1.767<br />

4 2.00 0.602 70.63 47.09 ± 0.84 52.91 1.672 1.723<br />

5 2.23 0.698 80.42 53.61 ± 0.47 46.39 1.729 1.666<br />

6 2.44 0.778 91.26 60.84 ± 0.47 39.16 1.784 1.592<br />

7 2.64 0.845 100.7 67.14 ± 0.24 32.86 1.826 1.516<br />

8 2.82 0.903 108.6 72.42 ± 0.35 27.58 1.859 1.440<br />

9 3.00 0.954 116.5 77.70 ± 0.37 22.43 1.890 1.348<br />

10 3.16 1.000 120.8 80.58 ± 0.49 19.42 1.906 1.288<br />

11 3.31 1.041 126.2 84.39 ± 0.01 15.61 1.926 1.193<br />

12 3.46 1.079 132.7 88.51 ± 1.53 11.49 1.946 1.060<br />

All values are represented as mean standard deviation (n=3)<br />

95


Zero order kinetic<br />

data<br />

Formulation Code Regression<br />

coefficient<br />

(r)<br />

Table-19: Kinetic values obtained from in-vitro release profile for microcapsules<br />

First order kinetic<br />

data<br />

Regression<br />

coefficient<br />

(r)<br />

Higuchi Matrix<br />

kinetic data<br />

Regression<br />

coefficient<br />

(r)<br />

Regression<br />

coefficient<br />

(r)<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Peppas kinetic data<br />

Results<br />

Slope ‘n’<br />

FM-1 0.9160 0.073 0.985 0.974 0.200<br />

FM-2 0.930 0.065 0.963 0.945 0.23<br />

FM-3 0.936 0.010 0.973 0.951 0.26<br />

FM-4 0.937 0.017 0.983 0.968 0.26<br />

96


Fig. 17: Zero order plots of Flurbiprofen microcapsules<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

Fig. 18: First order plots of Flurbiprofen microcapsules<br />

<br />

<br />

<br />

97


Fig. 19: Higuchi diffusion plots of Flurbiprofen microcapsules<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

<br />

<br />

<br />

Fig. 20: Peppas exponentional plots of Flurbiprofen microcapsules<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

98


5.6 Evaluation of formulation treated empty capsules<br />

5.6.1 Dimension<br />

Average capsule length<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

Before formaldehyde treatment (untreated cap and body) : 23.5 mm<br />

After formaldehyde treatment (treated body and untreated cap) : 22.5 mm<br />

Average diameter of capsule body<br />

Before formaldehyde treatment : 7.9 mm<br />

After formaldehyde treatment : 7.5 mm<br />

Average length of capsule body<br />

Before formaldehyde treatment : 20.5 mm<br />

After formaldehyde treatment : 19.5 mm<br />

5.6.2 Solubility studies for the treated capsules<br />

When the capsules were subjected to solubility studies in different buffers<br />

for 24 hrs, the following observation were made<br />

a) In all the case of normal capsules, both cap and body dissolved within<br />

fifteen minutes.<br />

b) In the case of formaldehyde treated capsules, only the cap dissolved within<br />

15minutes, while the capsule remained intact for about 24 hours.<br />

99


Quantity test for free formaldehyde<br />

Results<br />

The formaldehyde capsules were tested for the presence of free<br />

formaldehyde. The sample solution was not more intensely colored than the<br />

standard solution inferring that less than 20µg free formaldehyde is present in 25<br />

capsule<br />

5.7 Evaluation of Modified Pulsincap<br />

5.7.1 Weight variation<br />

The filled capsules pass the weight variation test as their weights are<br />

within the specified limits.<br />

5.7.2 Thickness of Cellulose Acetate Phthalate<br />

Table-20: Coating thickness<br />

Formulation Thickness of Coating (mm)<br />

F-1 0.060<br />

*Average of triplicate sets<br />

5.7.3 IR- Study<br />

F-2 0.054<br />

F-3 0.063<br />

F-4 0.067<br />

F-5 0.056<br />

F-6 0.059<br />

F-7 0.057<br />

F-8 0.061<br />

F-9 0.066<br />

From the spectra of pure drug and the combination of drug with polymers,<br />

it was observed that all the characteristics peaks of Flurbiprofen were present in<br />

the combination spectrum, thus indicating compatibility of the drug and polymer.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 100


Fig. 21: I.R. Spectrum of Flurbiprofen + Guargum<br />

Fig. 22: I.R. Spectrum of Flurbiprofen + HPMC<br />

Fig. 23: I.R. Spectrum of Flurbiprofen + Sodium Alginate<br />

Results<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 101


5.7.3 Evaluation of In-vitro drug release of Prepared Pulsatile Capsule Device<br />

Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 21: In-vitro release rate profile of F1 containing 20mg Guar Gum<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 0.098 0.06 ± 0.042 99.93 -1.184 1.999<br />

5 2.23 0.698 9.073 6.05 ± 0.034 93.95 0.781 1.972<br />

6 2.44 0.778 30.31 20.20 ± 0.315 78.80 1.305 1.902<br />

7 2.64 0.845 38.23 25.50 ± 0.130 74.50 1.406 1.872<br />

8 2.82 0.903 51.05 34.04 ± 0.310 60.96 1.531 1.819<br />

9 3.00 0.954 61.40 40.93 ± 0.650 59.06 1.612 1.771<br />

10 3.16 1.000 75.85 50.56 ± 1.767 49.18 1.703 1.697<br />

11 3.31 1.041 81.29 54.19 ± 0.317 45.81 1.730 1.660<br />

12 3.46 1.079 95.40 63.94 ± 0.358 36.09 1.805 1.557<br />

13 3.60 1.113 101.2 67.51 ± 0.239 32.49 1.829 1.511<br />

14 3.74 1.146 107.3 71.50 ± 0.536 28.50 1.854 1.454<br />

15 3.87 1.176 111.9 74.61 ± 0.408 25.39 1.872 1.404<br />

All values are represented as mean standard deviation (n=3)<br />

102


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 22: In-vitro release rate profile of F2 containing 30mg Guar Gum<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 0.000 0.000 100 0.000 2.000<br />

5 2.23 0.698 4.160 2.777 ± 0.216 97.22 0.443 1.987<br />

6 2.44 0.778 32.25 20.82 ± 0.625 79.18 1.318 1.898<br />

7 2.64 0.845 38.38 25.59 ± 0.319 74.41 1.408 1.871<br />

8 2.82 0.903 53.08 35.39 ± 0.092 64.61 1.548 1.810<br />

9 3.00 0.954 58.90 39.27 ± 0.086 60.73 1.594 1.783<br />

10 3.16 1.000 68.15 45.44 ± 0.631 54.56 1.657 1.736<br />

11 3.31 1.041 84.71 56.47 ± 0.092 43.53 1.751 1.638<br />

12 3.46 1.079 89.76 59.84 ± 0.160 40.16 1.776 1.603<br />

13 3.60 1.113 93.41 62.27 ± 0.473 37.73 1.794 1.576<br />

14 3.74 1.146 98.62 65.73 ± 0.408 34.25 1.817 1.534<br />

15 3.87 1.176 104.1 69.43 ± 0.234 30.57 1.841 1.485<br />

All values are represented as mean standard deviation (n=3)<br />

103


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 23: In-vitro release rate profile of F3 containing 40mg Guar Gum<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 0.000 0.000 100 0.000 2.000<br />

5 2.23 0.698 3.330 2.213 ± 0.577 97.78 0.344 1.990<br />

6 2.44 0.778 12.18 8.200 ± 0.080 91.88 0.909 1.963<br />

7 2.64 0.845 23.39 15.59 ± 0.546 84.41 1.192 1.926<br />

8 2.82 0.903 33.88 22.59 ± 0.240 77.41 1.353 1.888<br />

9 3.00 0.954 47.01 31.34 ± 0.086 68.66 1.496 1.836<br />

10 3.16 1.000 56.27 37.51 ± 0.239 62.49 1.574 1.795<br />

11 3.31 1.041 68.70 45.80 ± 0.323 54.20 1.660 1.733<br />

12 3.46 1.079 82.53 55.02 ± 0.155 44.98 1.740 1.653<br />

13 3.60 1.113 85.56 57.04 ± 1.123 42.96 1.756 1.633<br />

14 3.74 1.146 88.52 59.01 ± 0.387 40.99 1.770 1.612<br />

15 3.87 1.176 93.41 62.28 ± 0.479 37.72 1.794 1.576<br />

All values are represented as mean standard deviation (n=3)<br />

104


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 24: In-vitro release rate profile of F4 containing 20mg HPMC<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 3.746 2.497 ± 0.205 97.50 0.397 1.989<br />

5 2.23 0.698 8.990 5.990 ± 0.043 94.01 0.777 1.973<br />

6 2.44 0.778 42.82 28.54 ± 0.768 71.46 1.455 1.854<br />

7 2.64 0.845 50.35 33.57 ± 0.680 66.43 1.525 1.822<br />

8 2.82 0.903 58.44 38.96 ± 0.321 61.04 1.590 1.785<br />

9 3.00 0.954 64.96 43.31 ± 0.735 56.69 1.636 1.753<br />

10 3.16 1.000 71.26 47.51 ± 0.092 52.49 1.676 1.720<br />

11 3.31 1.041 84.71 56.47 ± 0.239 43.53 1.751 1.638<br />

12 3.46 1.079 96.44 64.30 ± 0.629 35.70 1.808 1.552<br />

13 3.60 1.113 99.32 66.10 ± 0.465 33.79 1.820 1.528<br />

14 3.74 1.146 108.9 72.64 ± 0.450 27.36 1.861 1.437<br />

15 3.87 1.176 114.7 76.46 ± 0.802 23.54 1.883 1.371<br />

All values are represented as mean standard deviation (n=3)<br />

105


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 25: In-vitro release rate profile of F5 containing 30mg HPMC<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 0.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 0.000 0.000 100 0.000 2.000<br />

5 2.23 0.698 6.920 4.613 ± 0.115 95.38 0.663 1.979<br />

6 2.44 0.778 41.08 27.38 ± 0.311 72.62 1.437 1.861<br />

7 2.64 0.845 46.55 31.03 ± 0.086 68.97 1.491 1.838<br />

8 2.82 0.903 53.31 35.54 ± 0.239 64.46 1.550 1.890<br />

9 3.00 0.954 65.32 43.42 ± 0.240 56.58 1.637 1.752<br />

10 3.16 1.000 74.68 49.79 ± 0.086 50.21 1.697 1.700<br />

11 3.31 1.041 84.95 56.09 ± 0.532 43.91 1.748 1.642<br />

12 3.46 1.079 94.73 63.08 ± 0.370 36.90 1.799 1.567<br />

13 3.60 1.113 97.74 64.97 ± 0.310 30.03 1.812 1.544<br />

14 3.74 1.146 103.4 68.96 ± 0.086 31.04 1.832 1.491<br />

15 3.87 1.176 107.5 71.71 ± 0.358 28.29 1.855 1.451<br />

All values are represented as mean standard deviation (n=3)<br />

106


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 26: In-vitro release rate profile of F6 containing 40mg HPMC<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 2.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 0.000 0.000 100 0.000 2.000<br />

5 2.23 0.698 0.720 0.483 ± 0.063 99.51 -0.315 1.997<br />

6 2.44 0.778 7.950 5.300 ± 0.088 94.70 0.724 1.976<br />

7 2.64 0.845 27.58 18.39 ± 0.239 81.66 1.264 1.911<br />

8 2.82 0.903 43.52 29.01 ± 0.086 70.99 1.462 1.851<br />

9 3.00 0.954 50.66 33.78 ± 0.327 66.22 1.528 1.820<br />

10 3.16 1.000 56.96 37.97 ± 0.323 62.03 1.579 1.792<br />

11 3.31 1.041 64.42 42.95 ± 0.387 57.05 1.632 1.756<br />

12 3.46 1.079 75.38 50.25 ± 0.319 49.75 1.701 1.696<br />

13 3.60 1.113 81.76 54.45 ± 0.392 45.55 1.735 1.658<br />

14 3.74 1.146 85.56 57.04 ± 0.536 42.96 1.756 1.633<br />

15 3.87 1.176 89.68 59.79 ± 0.363 40.21 1.776 1.604<br />

All values are represented as mean standard deviation (n=3)<br />

107


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 27: In-vitro release rate profile of F7 containing 20mg Sodium Alginate<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 2.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 6.13 4.127 ± 0.361 95.875 0.615 1.981<br />

5 2.23 0.698 13.95 9.303 ± 0.057 90.697 0.968 1.957<br />

6 2.44 0.778 44.37 29.58 ± 0.239 70.42 1.470 1.847<br />

7 2.64 0.845 51.91 34.61 ± 0.392 65.39 1.539 1.815<br />

8 2.82 0.903 59.92 39.68 ± 0.092 60.32 1.598 1.780<br />

9 3.00 0.954 70.10 46.73 ± 0.086 53.27 1.669 1.726<br />

10 3.16 1.000 79.65 53.10 ± 0.327 46.90 1.725 1.671<br />

11 3.31 1.041 90.15 60.10 ± 0.086 39.90 1.778 1.600<br />

12 3.46 1.079 99.16 66.11 ± 0.626 33.98 1.820 1.530<br />

13 3.60 1.113 109.7 73.16 ± 0.729 26.84 1.864 1.428<br />

14 3.74 1.146 118.8 79.22 ± 0.450 20.78 1.898 1.317<br />

15 3.87 1.176 124.4 82.95 ± 0.239 17.05 1.918 1.231<br />

All values are represented as mean standard deviation (n=3)<br />

108


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 28: In-vitro release rate profile of F8 containing 30mg Sodium Alginate<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 2.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 1.874 1.247 ± 0.105 98.75 0.095 1.994<br />

5 2.23 0.698 9.130 6.087 ± 0.011 93.91 0.784 1.972<br />

6 2.44 0.778 35.20 23.47 ± 0.542 76.53 1.370 1.883<br />

7 2.64 0.845 45.55 30.36 ± 0.171 69.64 1.482 1.842<br />

8 2.82 0.903 54.47 36.32 ± 0.184 63.68 1.560 1.804<br />

9 3.00 0.954 63.45 42.33 ± 0.179 57.60 1.626 1.760<br />

10 3.16 1.000 83.70 55.80 ± 0.406 44.20 1.746 1.645<br />

11 3.31 1.041 94.81 63.21 ± 0.392 36.79 1.800 1.565<br />

12 3.46 1.079 96.68 64.45 ± 0.315 35.55 1.809 1.550<br />

13 3.60 1.113 99.32 66.21 ± 0.315 33.79 1.809 1.528<br />

14 3.74 1.146 102.8 68.60 ± 0.502 31.40 1.836 1.496<br />

15 3.87 1.176 105.3 70.25 ± 0.155 29.75 1.846 1.473<br />

All values are represented as mean standard deviation (n=3)<br />

109


Time (T)<br />

Hrs<br />

Square root<br />

of Time<br />

Table 29: In-vitro release rate profile of F9 containing 40mg Sodium Alginate<br />

Log Time<br />

Cum. Drug<br />

release (mg)<br />

Cum.% Drug<br />

release ± SD<br />

Cum.% Drug<br />

retained<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Log Cum. %<br />

drug released<br />

Results<br />

Log Cum. %<br />

Drug retained<br />

0 0.00 0.000 0.000 0.000 0.000 0.000 2.000<br />

1 1.00 0.000 0.000 0.000 100 0.000 2.000<br />

2 1.41 0.301 0.000 0.000 100 0.000 2.000<br />

3 1.73 0.477 0.000 0.000 100 0.000 2.000<br />

4 2.00 0.602 5.777 3.600 ± 0.398 96.44 0.556 1.984<br />

5 2.23 0.698 6.760 4.436 ± 0.057 95.56 0.646 1.980<br />

6 2.44 0.778 36.99 24.66 ± 0.023 75.34 1.391 1.877<br />

7 2.64 0.845 41.34 27.56 ± 0.473 72.44 1.440 1.859<br />

8 2.82 0.903 47.25 31.50 ± 0.392 68.50 1.498 1.835<br />

9 3.00 0.954 60.61 40.41 ± 0.155 59.59 1.606 1.775<br />

10 3.16 1.000 74.84 49.89 ± 0.265 50.11 1.698 1.699<br />

11 3.31 1.041 83.39 55.59 ± 0.239 44.41 1.744 1.647<br />

12 3.46 1.079 98.00 65.33 ± 0.701 34.64 1.815 1.539<br />

13 3.60 1.113 104.9 70.00 ± 0.645 30.00 1.845 1.477<br />

14 3.74 1.146 112.8 75.33 ± 0.551 24.76 1.876 1.392<br />

15 3.87 1.176 116.0 77.36 ± 1.166 22.64 1.888 1.354<br />

All values are represented as mean standard deviation (n=3)<br />

110


Table 30: Kinetic values obtained from in-vitro release profile for microcapsules<br />

Zero order kinetic<br />

data<br />

First order kinetic<br />

data<br />

Higuchi Matrix<br />

kinetic data<br />

Peppas kinetic data<br />

Formulation Code Regression Regression Regression Regression<br />

coefficient coefficient coefficient coefficient Slope ‘n’<br />

(r)<br />

(r)<br />

(r)<br />

(r)<br />

F1 0.955 0.950 0.917 0.717 2.500<br />

F2 0.947 0.958 0.913 0.887 2.644<br />

F3 0.930 0.929 0.875 0.920 2.609<br />

F4 0.958 0.957 0.930 0.915 2.504<br />

F5 0.950 0.945 0.923 0.880 2.622<br />

F6 0.929 0.943 0.930 0.821 2.724<br />

F7 0.967 0.943 0.937 0.926 2.459<br />

F8 0.943 0.954 0.918 0.899 2.593<br />

F9 0.957 0.938 0.910 0.971 2.500<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA<br />

Results<br />

111


Results<br />

Fig. 24: Zero order plots of formulation containing Guar Gum as hydrogel<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

plug<br />

<br />

<br />

<br />

Fig. 25: First order plots of formulation containing Guar Gum as hydrogel<br />

<br />

<br />

<br />

<br />

<br />

<br />

plug<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 112


Results<br />

Fig. 26: Higuchi diffusion plots of formulation containing Guar Gum as<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

hydrogel plug<br />

<br />

<br />

<br />

Fig. 27: Peppas exponentional plots of formulation containing Guar Gum as<br />

<br />

<br />

<br />

<br />

<br />

<br />

hydrogel plug<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 113


Results<br />

Fig. 28: Zero order plots of formulation containing HPMC as hydrogel plug<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

Fig. 29: First order plots of formulation containing HPMC as hydrogel plug<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 114


Results<br />

Fig. 30: Higuchi diffusion plots of formulation containing HPMC as hydrogel<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

plug<br />

<br />

<br />

<br />

Fig. 31: Peppas exponentional plots of formulation containing HPMC as<br />

<br />

<br />

<br />

<br />

<br />

<br />

hydrogel plug<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 115


Results<br />

Fig. 32: Zero order plots of formulation containing Sodium Alginate as<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

hydrogel plug<br />

<br />

<br />

<br />

Fig. 33: First order plots of formulation containing Sodium Alginate as<br />

<br />

<br />

<br />

<br />

<br />

<br />

hydrogel plug<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 116


Results<br />

Fig. 34: Higuchi diffusion plots of formulation containing Sodium Alginate as<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

<br />

hydrogel plug<br />

<br />

<br />

<br />

Fig. 35: Peppas exponentional plots of formulation containing Sodium<br />

<br />

<br />

<br />

<br />

<br />

<br />

Alginate as hydrogel plug<br />

<br />

<br />

<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 117


CHAPTER-6<br />

<br />

Discussion<br />

In the present study, an attempt was made to develop and evaluate<br />

pulsatile drug delivery system containing eudragit microcapsules with<br />

subsequently lower dose of Flurbiprofen for colon specific delivery and for better<br />

treatment of early morning symptoms in rheumatoid arthritis and prevent<br />

unwanted systemic side effects.<br />

Calibration curve for the estimation of Flurbiprofen was constructed in<br />

Ethanol, 1.2 pH 7.4 pH and 6.8 pH buffer at 247 nm the method obeyed Beer’s<br />

Lambert law in the range of 2-16 mcg/ml.<br />

Microcapsules of Flurbiprofen using Eudragit L-100/S-100 as a polymer<br />

by emulsion solvent evaporation method as shown in Table-7. In this method, the<br />

organic phase was slowly poured in liquid paraffin and the emulsion was<br />

stabilized by Span- 80. The organic phase evaporated and finally spherical,<br />

smooth–walled, rigid and discrete microcapsules were formed.<br />

Particle size<br />

The mean particle size of the microcapsules significantly increased with<br />

increase in polymer concentration and was ranged in between 163.68 to 251.84<br />

m. The reason must be, the viscosity of medium which increase as the polymer<br />

concentration increase resulting in enhanced interfacial tension. Shearing<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 118


Discussion<br />

efficiency is also diminished at higher viscosities. This may be resulting in the<br />

formation of large particles. (as shown in Table-13)<br />

Flow properties:<br />

The value of between 18 – 22° indicates reasonable flow and all the<br />

batches were found to fit with respect of flow ability. (as shown in table-13)<br />

Percentage yield:<br />

Percentage yield of the formulation was carried out and was found to be<br />

within the range between 85.85 to 90.24 %. (as shown in Table-14)<br />

Drug Content:<br />

Drug content of the formulation was carried out and was found to be<br />

within the range between 75.9 to 95.59 %. (as shown Table-14)<br />

Scanning Electron Microscopy:<br />

Scanning electron microscopy was performed to characterize the surface<br />

of the formed microcapsules. The outer surface of FM-3 formulation was smooth<br />

and dense, while the internal surface was porous. The shell of microcapsules also<br />

showed some porous structure it may be caused by evaporation of solvent<br />

entrapped within the shell of microcapsules after forming smooth and dense<br />

layer.(as shown in Fig-12)<br />

IR Studies<br />

Drug-polymer interaction study was carried out for pure drug, eudragit L-<br />

100 and S-100 and FM-3.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 119


Discussion<br />

Flurbiprofen was used in this study which contains carboxylic acid<br />

function responsible for a broad hump 3200 cm -1 .The aromatic C-H peaks was<br />

observed at 3076, 3063, 3032 cm -1 . The broad carboxylic acid absorption is<br />

noticed at 1701 cm -1 . These data are in concurrent with the structure of the drug<br />

molecular. .(as shown in Fig-13)<br />

Polymer eudragit L-100 and S-100 gave the broad carboxylic acid peak<br />

3500 cm -1 and C=0 absorption at 1724 cm -1 . These are the characteristics of the<br />

polymer under study. .(as shown in Fig-14,15)<br />

The formulation FM-3 which was subjected to IR. The formulated product<br />

is showed identical spectrum with respect to the spectrum of the pure drug and<br />

polymers, indicating there is no chemical interaction between the drug molecule<br />

and polymers.(as shown in Fig-16)<br />

In-vitro release studies:<br />

The In-vitro release studies of Flurbiprofen from prepared microcapsules<br />

were carried in pH 6.8 buffer as a diffusion medium for a period of 12 hrs. The<br />

drug release from the formulations decreased with increase in the amount of<br />

polymer added in each formulation. The release showed a biphasic release with an<br />

initial burst effect. In the first 30 min. drug release was 29.29%, 27.43%, 26.96%<br />

and 24.70% for FM-1 to FM-4 respectively. The mechanism for the burst release<br />

can be attributed to the drug loaded on the microcapsule or imperfect entrapment<br />

of drug.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 120


Discussion<br />

The overall cumulative % release for FM-1, FM-2, FM-3 and FM-4 were<br />

found to be 95.58%, 91.96%, 88.87%, and 88.51% at the end of 12 th hrs. ( as<br />

shown in Table-15 to 18 )<br />

The ‘r’ values for zero order kinetics of FM-1, FM-2, FM-3 and FM-4<br />

were 0.916, 0.930, 0.936 and 0.937 respectively. The ‘r’ values for first order of<br />

FM-1, FM-2, FM-3 and FM-4 were 0.073, 0.065, 0.010 and 0.017 respectively.<br />

The ‘r’ values indicate the drug release follows zero order.<br />

To ascertain the drug release mechanism, the in-vitro data were also<br />

subjected to Higuchi diffusion. The ‘r’ values of Higuchi diffusion was 0.985,<br />

0.963, 0.973 and 0.983 for formulation FM-1, FM-2, FM-3 and FM-4<br />

respectively. It suggests that the Higuchi diffusion plots of all the formulations<br />

were fairly linear because ‘r’ values near about 1 in all the cases. So it confirms<br />

the drug release by Higuchi diffusion mechanism.<br />

The formulations were subjected to peppas plots by taking log cum % drug<br />

released versus log time. The plots are found fairly linear and slope value was<br />

calculated (n value) which was in ranges of 0.20 to 0.26 indicating the drug was<br />

released by Fickian diffusion mechanism. (as shown in Table-19)<br />

Formaldehyde treatment of hard gelatine capsules:<br />

The bodies of hard gelatine capsules were made insoluble by<br />

formaldehyde treatment. This was done by exposing the bodies of the capsules to<br />

vapours of formaldehyde; the caps were not exposed leaving them water-soluble.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 121


Discussion<br />

The capsules were tested for physical and chemical changes caused by exposure to<br />

vapours of formaldehyde.<br />

Dimensions:<br />

On formaldehyde treatment, the ‘00’ size capsule bodies showed a<br />

significant decrease in length and diameter.<br />

Solubility studies:<br />

When the capsules were subjected to studies in different buffers, the<br />

untreated caps disintegrated within 10 mins in all the media whereas the treated<br />

bodies remained intact for about 24 hrs.<br />

Qualitative test for free formaldehyde:<br />

Limit test for the presence of residual formaldehyde was carried. The<br />

sample solution was less intensively coloured when compared with standard<br />

inferring that less than 20 g/ml of free formaldehyde is present in 25 capsules<br />

bodies) as per the I.P.<br />

Formulation of modified pulsincap:<br />

Microcapsules equivalent to 150 mg of Flurbiprofen were filled into the<br />

treated bodies and plugged with different polymers like guar gum, sodium<br />

alginate, HPMC at different concentrations. The filled capsules were completely<br />

coated with 5% CAP solution. These pulsatile drug delivery systems were further<br />

evaluated for :( as shown in Table-8)<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 122


Thickness:<br />

Discussion<br />

The thickness of the CAP coating was measured by using screw gauge.<br />

The values ranged from 0.05-0.067 mm.(as shown in Table-20)<br />

IR studies:<br />

Drug-polymer interaction study was carried out for physical mixture of<br />

pure drug and polymers (i.e. Flurbiprofen + Guar Gum, Flurbiprofen + HPMC and<br />

Flurbiprofen + Sodium Alginate). From the results, it was observed that all the<br />

characteristic peaks of Flurbiprofen were present in the combination spectrum,<br />

thus indicating compatibility of the drug and polymer in pulsatile device. (as<br />

shown in Fig-21 to 23)<br />

In-vitro release studies:<br />

In-vitro drug release profiles of pulsatile device were found to have very<br />

good sustaining efficacy. During dissolution studies, it was observed that, the<br />

enteric coat of the cellulose acetate phthalate was intact for 2 hours in pH 1.2, but<br />

dissolved in intestinal pH, leaving the soluble cap of capsule, which also dissolved<br />

in pH 7.4 phosphate buffer and then the exposed polymer plug which absorbed the<br />

surrounding fluid, swelled and released the drug through the swollen matrix.<br />

After complete wetting of the plug, it formed a soft mass, which was then easily<br />

ejected out of the capsule body; releasing the eudragit microcapsules into<br />

simulated colonic fluid (pH 6.8 phosphate buffer).<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 123


Discussion<br />

With all the formulations, there was absolutely no drug release in pH 1.2,<br />

thus indicating the efficiency of 5% CAP for enteric coating. Very slight release<br />

was observed in pH 7.4 phosphate buffer.<br />

a) Formulations with guar gum as hydrogel plug:<br />

With formulations F1 (20mg), F2 (30mg) and F3 (40mg) at the end of 5 th<br />

hrs there was 6.050%, 2.777% and 2.212% drug was released respectively and at<br />

the end of 15 th hrs 74.61%, 69.43% and 62.28% drug release was found in F1, F2<br />

and F3 respectively. (as shown in Table-21to 23)<br />

b) Formulation with HPMC as hydrogel plug:<br />

With formulation F4 (20mg), F5 (30mg) and F6 (40mg) at the end of 5 th<br />

hrs 5.990%, 4.613% and 0.483% drug was released respectively and at the end of<br />

15 th hrs 76.46%, 71.71% and 59.79% drug released respectively. (as shown in 24<br />

to 26)<br />

c) Formulations with sodium alginate as hydrogel plug:<br />

With formulations F7 (20 mg), F8 (30 mg) and F9 (40 mg), at the end of<br />

5 th hrs around 9.303%, 6.087% and 4.436% drug released respectively and at the<br />

end of 15 th hrs 82.95%, 70.25% and 77.36% drug was released respectively.(as<br />

shown in Table-27 to 29)<br />

From all the above observations, it was found that the order of sustaining<br />

capacity of polymer is HPMC > Guar gum > Sodium alginate.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 124


Discussion<br />

The ‘r’ values for zero order kinetics of F-1, F-2, F-3, F-4, F-5, F-6, F-7,<br />

F-7, F-8 and F-9 were 0.955, 0.947, 0.930, 0.958, 0.950, 0.929, 0.967, 0.967,<br />

0.943 and 0.957 respectively. The ‘r’ values of the first order kinetics of F-1, F-2,<br />

F-3, F-4, F-5, F-6, F-7, F-7, F-8 and F-9 were 0.950, 0.958, 0.929, 0.957, 0.945,<br />

0.943, 0.943, 0.954, and 0.938 respectively. The ‘r’ value indicates drug release<br />

follows mixed order kinetics.<br />

To ascertain the drug release mechanism, the in-vitro data were also<br />

subjected to Higuchi diffusion. The ‘r’ values of Higuchi diffusion was 0.917,<br />

0.913, 0.875, 0930, 0.923 0.930, 0.937, 0918 and 0.910for formulation F-1 to F-9,<br />

respectively. It suggests that the Higuchi diffusion plots of all the formulations<br />

were fairly linear because ‘r’ values near about 1 in all the cases. So it confirms<br />

the drug release by Higuchi diffusion mechanism.<br />

The formulations were subjected to peppas plots by taking log cum % drug<br />

released versus log time. The plots are found fairly linear and the ‘r’ values are<br />

near to 1 and also slop value was calculated (n value) which was in ranges of 2.45<br />

to 2.72, indicating the drug was released by Super Case II transport diffusion<br />

mechanism.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 125


CHAPTER-7<br />

<br />

Conclusion<br />

The aim of this study was to explore the feasibility of time and pH<br />

dependent colon specific, pulsatile drug delivery system of Flurbiprofen to treat<br />

the rheumatoid arthritis. A satisfactory attempt was made to develop<br />

microcapsules by using pH dependent polymers (eudragit L/S100) and further the<br />

time dependent, pulsatile device was designed for the microcapsules and<br />

evaluated.<br />

The data obtained from the study of “Development and evaluation of<br />

pulsatile drug delivery system of Flurbiprofen” reveals following conclusion:<br />

Eudragit L-100 and S-100 in the ratio 1:2 are suitable for preparation of<br />

microcapsules for colonic targeting.<br />

The mean particle size of microspheres was in the range of 163.68 –<br />

251.84 m depending upon the type of polymer used. The particle size<br />

increased significantly as the amount of polymer increased.<br />

The flow properties of all the prepared microspheres were good as<br />

indicated by low angle of repose ( < 40º).The good flow properties<br />

suggested that the microspheres produced were non-aggregated.<br />

The entrapment efficiency was good in all the cases. This suggested that<br />

optimized parameters were used in the method of preparations.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 126


Conclusion<br />

In-vitro drug release of microcapsules showed biphasic release pattern for<br />

all microcapsules with initial burst release effect, which may be attributed<br />

to the drug loaded onto the surface of the particles.<br />

On the basis of, particle size, drug content, Scanning Electron Microscopy,<br />

IR-study, in-vitro release studies and its kinetic data, FM-3 was selected as<br />

an optimized formulation for designing pulsatile device.<br />

The solubility studies of empty gelatine capsule bodies, which where<br />

crosslinked with formaldehyde treatment, revealed that they are intact for<br />

24 hrs, and hence suitable for colon targeting.<br />

From all obtained results, it was found that the order of sustaining capacity<br />

of polymer is HPMC > Guar gum > Sodium alginate.<br />

Hence, finally it was concluded that the prepared pulsatile drug delivery<br />

system can be considered as one of the promising formulation technique<br />

for preparing colon specific drug delivery systems and hence in<br />

chronotherapeutic management of arthritis.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 127


CHAPTER-8<br />

<br />

Summary<br />

Over the past two decades there has been a growing appreciation on the<br />

importance of circadian rhythms on GIT physiology and on disease states,<br />

together with the realization of the significance of time-of-day of the<br />

administration on resultant pharmacodynamics and pharmacokinetics parameters.<br />

The significance of these day-light variations has not been over looked from the<br />

drug-delivery perspective and pharmaceutical scientists have displayed<br />

considerable ingenuity in the development of time delayed drug delivery system<br />

to address emerging chronotherapeutic formulation.<br />

The colon is a site where both local and systemic delivery of drug can<br />

take place. Treatment could be made more effective if it were possible for drug to<br />

be targeted directly on the colon. Colon-specific system could also be used in<br />

diseases that have diurnal rhythms. In the present study, an attempt was made to<br />

design and characterize pulsatile, colon specific drug delivery system in order to<br />

target the drug to the colon, and intentionally delaying the drug absorption from<br />

therapeutic point of view in the treatment of rheumatoid arthritis, where peak<br />

symptoms are observed in the early morning.<br />

Eudragit L/S 100 in ratio 1:2 was employed to prepare microcapsule of<br />

Flurbiprofen by 0/0 emulsion solvent evaporation method. Four formulations<br />

(FM-1 to FM-4) were prepared by varying the ratio of the drug and polymer.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 128


Summary<br />

These formulations were subjected to various evaluation parameters like particle<br />

size, flow properties, percentage yield, drug content, scanning electron<br />

microscopy, IR-study and in-vitro drug release studies using pH 6.8 phosphate<br />

buffer as dissolution medium. Based on the results obtain, the FM-3 was<br />

considered as the optimum formulation to design time and pH dependent, pulsatile<br />

drug delivery system.<br />

In the next step, the capsule bodies were made insoluble by<br />

formaldehyde treatment and these were subjected to various physical and<br />

chemical test such as dimension measurement, solubility studies and qualitative<br />

for free formaldehyde.<br />

The microcapsules equivalent to 150 mg of drug was filled in to the<br />

formaldehyde treated capsule bodies and plugged with different polymers, at<br />

different concentration (20, 30 and 40 mg). The polymers used as hydrogel plugs<br />

were Guar gum, HPMC and Sodium alginate. The joint of the capsule body and<br />

cape was sealed with small amount of 5 % ethycellulose ethanolic solution. Then<br />

these filled capsules were completely coated with 5 % cellulose acetate phthalate<br />

solution. These pulsatile formulations were subjected to various tests such as<br />

thickness of CAP coating, weight variation and in vitro release studies.<br />

From the in vitro release studies of pulsatile device, it was observed that<br />

with all the formulations, there was absolutely no drug release in simulated gastric<br />

fluid (acidic pH 1.2) for 2 hrs. Negligible amount of drug release was observed in<br />

simulated intestinal fluid (pH 7.4 phosphate buffer), where the dissol ution were<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 129


Summary<br />

carried out for 3 hrs. In remaining hours dissolution was continued in simulated<br />

colonic medium (pH 6.8 phosphate buffers) and here the drug release was found<br />

to be 76.61, 69.43, 62.28 from F-1, F-2, F-3 respectively. 74.46, 71.71, 59.79 from<br />

F-4, F-5, F-6 respectively and 82.95, 70.25, 77.36 from F-7, F-8, F-9 respectively<br />

at the end of 15 hrs.<br />

All the polymers employed in study are suitable for colon targeting. The<br />

order of sustaining capacity of the polymer was HPMC > guar gum > sodium<br />

alginate. The obtained results showed the capability of the system in dealing drug<br />

release for programmable period and time and possibility of exploiting such delay<br />

to attain colon targeting.<br />

.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 130


Bibliography<br />

1. Gothaskar, AV, Joshi AM, Joshi, NH, 2004. Pulsatile drug delivery<br />

system—a review. Drug Del. Technol. 4, http://www.drugdeliverytech.<br />

com/id article=250.<br />

2. Bi-Botti CY. Chronopharmaceutics: Gimmick or clinically relevant<br />

approach to drug delivery—a review. J Control Rel 2004; 98(3):337–<br />

353.<br />

3. Bjorn, L. 1996. The clinical relevance of chronopharmacology in<br />

therapeutics. Pharmacological Res.1996; 33:107–115.<br />

4. Suresh S, Pathak S. Chronopharmaceutics: Emerging role of bio-<br />

rhythms in optimizing drug therapy. Indian J Pharm Sci 2005;<br />

67(2):135-140.<br />

5. Sharma S, Pawar SA, Low density multiparticulate system for pulsatile<br />

release of meloxicam. Int J Pharm 2006; 313: 150-158.<br />

6. Fan TY, Wei SL, Yan WW Chen DB, Li J. An investigation of pulsatile<br />

release tablets with ethycellulose and eudragit-L as film coating<br />

materials and cross-linked polyvinyl pyrolidone in the core tablets. J<br />

Control Rel 2001; 77:245-251.<br />

7. Krishainah YSR, Satyanarayan S. Colon-specific drug delivery system.<br />

In: Jain NK, editor. Advances in controlled and novel drug delivery. 1 st<br />

Ed. New Delhi: CBS publishers and distributors; 2001:89-119.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 131


Bibliography<br />

8. Bhavana V, Khopade AJ, Jain VVD, Jain NK. Oral pulsatile drug<br />

delivery. Eastern pharmacist 1996 Aug; 39 (464): 21-6.<br />

9. Smolensky MH, Peppas AN. Chronobiology drug delivery and<br />

Chronopharmaceutics. Adv Drug Delivery 2007; 59:825-851.<br />

10. Hrushesky WJM, 1994. Timing is everything. The Sci, 32–37.<br />

11. Roger Walker and Cate Whittlsea. Clinical pharmacy and Therapeutics.<br />

Elsevier Health Sci 4 th Ed. 2008:759-776.<br />

12. Harsh Mohan. Textbook of Pathology. Jaypee Brothers, Medical<br />

Publishers Ltd. New Delhi, 4 th Ed. 2003:832-838.<br />

13. http://www.arthritiestreatmentandrelief.com<br />

14. Libo Y, James SC, Joseph AF. Colon specific drug delivery: new<br />

approaches and in-vitro/in-vivo evaluation—review. Int J Pharm 2002;<br />

235:1–15.<br />

15. Shivkumar HG, Promod KTM, Kashappa GD. Pulsatile drug delivery<br />

systems .Indian J Pharma Edu 2003 July-Sep : 37(3) : 125:8.<br />

16. Sarasija S, Stutie P. Chronotherapeutics: emerging role of biorhythms in<br />

optimizing drug therapy. Indian J Phrma Sci 2000; 67:135–140.<br />

17. Wiwattanapatapee R, Luelak Lomlim, Krisanee Saramunee. Dendrimers<br />

conjugates for colonic delivery of 5-aminosalicylic Acid. J Control Rel<br />

2003; (88): 1–9<br />

18. Julie B, Howard, NES, John ME, Gillian P, Fran MB. The tolerability of<br />

multiple oral doses of Pulsincap ® capsules in healthy volunteers. J<br />

Control Rel.1996; 38: 151–158.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 132


Bibliography<br />

19. Sachin Survase, Neeraj Kumar. Pulsatile drug delivery: Current scenario<br />

crips 2007; 8:33<br />

20. Krishnaiah YSR, Reddy PBR, Satyanarayana V, Karthikeyan RS.<br />

Studies on the development of oral colon targeted drug delivery systems<br />

for metronidazole in the treatment of amoebiasis. Int J Pharm 2002;<br />

236: 43-55.<br />

21. Chien YW. Controlled and Modulated Release Drug Delivery System in<br />

Swarbrik J, Boylan JC Eds. Encyclopedia of Pharmaceutical<br />

Technology, Marcel Dekker, New York, 1990:281-313.<br />

22. Sangalli ME, Maroni A, Zema L, Busetti C, Giordano F, Gazzaniga A.<br />

In-vitro and in-vivo evaluation of an oral system for time and / or site-<br />

specific drug delivery. J Control Rel 2001; 73:103-110.<br />

23. Gupta SK, Atkinson L, Theeuwes F, Wong P, Gilbert PJ, Longstreth J.<br />

Pharmacokinetics of verapamil from an osmotic system with delayed<br />

onset. Eur J Pharm and Biopharma1996; 42 (1):74-81<br />

24. Ross AC, Macrae RJ, Walther M, Stevens HNE. Chronopharmaceutical<br />

drug delivery from a pulsatile capsule device based on programmable<br />

erosion. J Pharm Pharmacol. 2000; 52:903-909.<br />

25. Sweetman SC. Martindale The complete drug reference, 33 rd Edn.<br />

Pharmaceutical Press: Great Britain; 2002:41-42.<br />

26. Listair CR, Ross JM, Mathias W, Howard NES. Chronopharmaceutical<br />

drug delivery from a pulsatile capsule device based on programmable<br />

erosion. J Pharm. Pharmacol 2002; 52:903–909.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 133


Bibliography<br />

27. Samanta MK, Suresh NV, Suresh B. Development of Pulsincap Drug<br />

Delivery of salbutamol sulphate for drug tareting. Indian. Pharma Sci<br />

2000; 62(2):102-7.<br />

28. Young-II J, Tomoya O, Zhaopeng Hu. Evaluation of an intestinal<br />

pressure-controlled colon delivery capsules prepared by dipping method.<br />

J Control Rel 2001; 71:75-82.<br />

29. Seshasayan A, Sreenivasa RB, Prasanna R, Ramana Murthy KV. Studies<br />

on release of Rifampicin from Modified Pulsincap Technique. Indian J<br />

Pharma Sci 2001:337-9.<br />

30. Sangalli ME, Maroni A, Zema L, Busetti C, et al. 2001. In vitro and in<br />

vivo evaluation of an oral system for time and/or site-specific drug<br />

delivery. J Control Rel 2001; 73:103–110.<br />

31. Zahirul Khan MI, Zeljko P, Nevenka K. A pH-dependent colon targeted<br />

oral drug delivery system using methacrylic acid copolymers. I.<br />

Manipulation of drug release using Eudragit ® L100-55 and Eudragit ®<br />

S100 combinations. J Control Rel 1999; 58:215–222.<br />

32. Howard NES, Clive GW, Peter GW, Massoud B, Julei SB, Alan CP.<br />

Evaluation of Pulsincap TM to provide regional delivery of dofetilide to<br />

the human GI tract. Int J Pharm2002; 236:27–34.<br />

33. Libo Y, James SC, Joseph AF. Colon specific drug delivery: new<br />

approaches and in vitro/in vivo evaluation—review. Int J Pharm 2002;<br />

235:1–15.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 134


Bibliography<br />

34. Ying-huan Li, Jia-bi Zhu. Modulation of combined release behaviours<br />

from a novel “tablets-in-capsules system”. J Control Rel 2004; 50:111-<br />

22.<br />

35. Tomohiro Takaya, Kiyoshi N, Motoki M, Ikuo Ogita, Norko N, Ryo-<br />

ichi Y, et al. Importance of dissolution process on systemic availability<br />

of drugs delivered by colon delivery system. J Control Rel 1998;<br />

50:111-22.<br />

36. Takashi Ishibashi, Kengo I, Hiroaki Kubo, Masakazu M, Hiroyuki Y.<br />

Evalution of colonic absorbability of drugs in dogs using a novel colon-<br />

targeted delivery capsules(CTDC) J Control Rel 1999; 59:361-76.<br />

37. Jonathan CDS, Alistair CR, Walter K, Richard WB, Ross JM, Howard<br />

NES, et al. Investigating the coating-dependent release mechanism of a<br />

pulsatile capsule using NMR microscopy. J Control Rel 2003; 92:341-7.<br />

38. Matiholimath VS, Dandagi PM, Jain SS , Gadad AP, Kulkarni AR.<br />

Time and pH dependent colon specific, pulsatile delivery o theophylline<br />

for nocturnal asthma. Int J Pharm 2007; 328:49-56.<br />

39. Abraham S, Srinath MS. Development of modified pulsincap drug<br />

delivery system of metronidazole for drug targeting Indian J Pharm Sci<br />

2007; 69(1):18-23.<br />

40. Srisagul Sungthongjeen, Satit Puttipipatkhachorn, Ornlaksana<br />

Paeratakul, Andrei Dashevsky, Roland Bodmeier. Development of<br />

pulsatile release tablets with swelling and rupturable layers. J Control<br />

Rel 2004:147–159.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 135


Bibliography<br />

41. Krishnaiah YSR, Veer Raju P, Dinesh Kumar B, Bhaskar P,<br />

Satyanarayana V. Development of colon targeted drug delivery systems<br />

for Mebendazole. J Control Rel 2001; 77: 87–95<br />

42. Satyanarayana S, Rama Prasad YV, Krishnaiah YSR, Trends in colonic<br />

drug delivery- a review. Indian Drugs 1996; 33(1):1-10.<br />

43. Rama Prasad YV, Krishnaiah YSR, Satyanarayana S. In-vitro evaluation<br />

of guar gum as a carrier for colon-specific drug delivery. J Control Rel<br />

1998; 51:281-87.<br />

44. Krishnaiah YSR, Seetha DA. Guar gum as a carrier for colon specific<br />

delivery; influence of metronidazole tinidazole on in-vitro release of<br />

albendazole from guar gum matrix tablets. And J Pharm. Sci 2001; 4(3):<br />

235-43.<br />

45. Leopold CS, Friend DR. In-vitro study for the assessment of poly (L -<br />

aspartic acid) as a drug carrier for colon-specific drug delivery. Int J<br />

Pharm 1995; 126:139-45.<br />

46. Rubinstein A,Radai R. In vitro and in vivo analysis of colon specificity<br />

of calcium pectinate formulations. Eur J Pharm Biopharm 1995; 41:291-<br />

295.<br />

47. Turkoglu M, Ugurlu T. In vitro evaluation of pectin-HPMC compression<br />

coated 5-aminosalicylic acid tablets for colonic delivery. Eur J Pharm<br />

Biopharm 2002; 53:65-73.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 136


Bibliography<br />

48. Rama Prasad YV, Krishnaiah YSR, Satyanarayana S. In vitro evaluation<br />

of guar gum as a carrier for colon-specific drug delivery. J Control Rel<br />

1998; 51:281-287.<br />

49. Krishnaiah YSR, Satyanaryana S, Rama Prasad YV. Studies of guar<br />

gum compression-coated 5-aminosalicylic acid tablets for colon-specific<br />

drug delivery. Drug Dev Ind Pharm 1999; 25:651-657.<br />

50. Krishnaiah YSR, Bhaskar Reddy PR, Satyanarayana V, Karthikeyan RS.<br />

Studies on the development of oral colon targeted drug delivery systems<br />

for metronidazole in the treatment of amoebiasis. Int J Pharm 2002;<br />

236:43-55.<br />

51. Shinha VR, Kumaria R. Polysaccharide matrix for microbially triggered<br />

drug delivery to the colon. Drug Dev Ind Pharm 2004; 30(2):143-150.<br />

52. Lorenzo-Lamosa ML, Remunan-Lopez C, Vila-Jato JL, Alonson MJ.<br />

Design of microencapsulated chitosan microspheres for colonic drug<br />

delivery. J Control Rel 1998; 52:109-118.<br />

53. Brondsted H, Andersen C, Hovgaard L. Crosslinked dextran – a new<br />

capsule material for colon targeting of drug. J Control Rel 1998; 53:7-<br />

13.<br />

54. Vervoort L, Van den Mooter G, Augustijns P, Kinget R, Inulin<br />

hydrogels. I.Dynamic and equilibrium swelling properties. Int J Pharm<br />

1998; 172:127-135.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 137


Bibliography<br />

55. Vervoort L, Rambant P, Van den Mooter G, Augustijns P, Kinget R.<br />

Inulin hydrogels. II. In vitro degradation study. Int J Pharm 1998;<br />

172:137-145.<br />

56. Rubinstein A, Nakar D, Sintov A. Chondroitin sulphate: a potential<br />

biodegradable carrier for colon-specific drug delivery. Int J Pharm 1992;<br />

84:141-150.<br />

57. Milojevic S, Newton JM, Cummings JH, Gibson GR, Botham RL, Ring<br />

SG, Stockham M, Allwood MC Amylose as a coating for drug delivery<br />

to the colon: preparation and in vitro evaluation using 5-aminosalicylic<br />

acid pellets. J Control Rel 1996; 38:75-84.<br />

58. Shantha KL, Ravichandran P, Rao KP. Azo polymeric hydrogels for<br />

colon targeted drug delivery. Biomaterids 1995; 16: 1313-1318.<br />

59. Takashi Ishibashi, Harumi Hatano, Masao Kobayashi, Masakazu<br />

Mizobe, Hiroyuki Yoshino. Design and evaluation of a new capsule-<br />

type dosage form for colon-targeted delivery of drugs. Intl J Pharm 168<br />

(1998) 31–40.<br />

60. Lalwani A, Santani DD. Pulsatile drug system. A-review Indian J Pharm<br />

Sci 2007; 69(4):487-497.<br />

61. Morta R, Jose L, Vila J. Design of new multiparticulate system for<br />

potential site-specific and controlled drug delivery to the colonic region.<br />

J Control Rel 1998; 55: 67–77.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 138


Bibliography<br />

62. Lorenzo-Lamosa ML, Remun˜a´n-Lo´pez C, Vila-Jato JL, Alonso MJ.<br />

Design of microencapsulated chitosan microspheres for colonic drug<br />

delivery. J Control Rel 1998; 52: 109–118.<br />

63. Shivakumar HN, Sarasija S, Venkataram S. Design and evaluation of a<br />

multiparticulate system for chronotherapeutic delivery of Diclofenac<br />

sodium. Indian J Pharm Sci 2002; 64(2): 133-137.<br />

64. Kristmundsdottir T, Gudmundsson OS, Ingvarsdottir K. Release of<br />

diltiazem from eudragit microparticles prepared by spray-drying. Int J<br />

Pharm 1996; 137: 159-65.<br />

65. Hideki Ichikawa, Yoshinob F, Christianah, Moji, Adeyeye. Design of<br />

prolonged release microcapsules containing diclofenac sodium for oral<br />

suspension and their preparation by the wuster process. Int J Pharm<br />

1997; 156:39-48.<br />

66. Giovani FP, Giulia B, Piera DM, Sante M. Microcapsulation of<br />

semisolide ketoprofen/polymer microspheres. Int J Pharma 2002; 242:<br />

175-78.<br />

67. Alf Lamprecht, Hiromitsu Yamamoto, Hirofumi Takeuchi, Yoshiaki<br />

Kawashima. Microsphere design for the colonic delivery of 5-<br />

fluorouracil. J Control Rel 2003; 90: 313–322.<br />

68. Havgaard L, Brondsted H. Dextran hydrogels for colon-specific drug<br />

delivery. J Control Rel 1995; 36: 159-166.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 139


Bibliography<br />

69. Mine Orlu, Erdal Cevher, Ahmet Araman. Design and evaluation of<br />

colon specific drug delivery system containing flurbiprofen<br />

microsponges. Int J Pharm 2006; 318: 103–117.<br />

70. http://www.drugbankcom./flurbiprofen.<br />

71. http://www.sciencelab.com/flurbiprofen.<br />

72. http://www.roehm.com<br />

73. Government of India Ministry of Health & Family Welfare. Indian<br />

Pharmacopoeia. Delhi: Controller of Publications; 1996.p: 750, 151.<br />

74. Ainley W, Paul JW. Handbook of pharmaceutical excipients:<br />

monograph. 2 ed Ed. . London: The pharmaceutical Press; 2000: P.51-2,<br />

128, 138-9, 257, 113, 119.<br />

75. http://www.tradeindia.com<br />

76. http://www.huasu.en.chemnet.com<br />

77. http://www.adarshguargum.com<br />

78. http://www.chemicalbook.com<br />

79. http://www.ronasgroup.com<br />

80. Dandagi PM, Matiholimath VS, Gadad AP, Kulkarni AR, Jain SS. pH<br />

Sensitive mebeverin microspheres for colon delivery Indian J Phrm Sci<br />

2009; 71 (4): 264-268.<br />

81. Mehat KA. Formulation of enterosoluble microparticles for an acid<br />

labile Protein. J Pharm Pharmaceut Sci (www.ualberta.ca/~csps) 2002;<br />

5(3):234-244.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 140


Bibliography<br />

82. D Nagasamy Venkatesh, Reddy AK, Samanta MK, Suresh B.<br />

Development and In Vitro Evaluation of Colonic Drug Systems for<br />

Tegaserod Maleate. Asian J Pharm 2009; Jan – Mar: 50-53.<br />

83. Manavalan ramasamy. physical pharmaceutics. 2nd ed. india :vignesh<br />

publisher;2004.<br />

84. Patel A, Ray S, Thakur RM. In vitro evaluation and optimization of<br />

controlled release floating drug delivery system of metformin<br />

hydrochloride. DARU 2006; 14(2): 57-64.<br />

85. Kothawade KB, Gattani SG, Surana SJ and Amrutkar JR. Colonic<br />

Delivery of Aceclofenac Using combination of pH and Time Dependent<br />

Polymers. Indian Drugs Nov 2009; 46 (11): 67-70.<br />

86. Saravanan M, Bhaskar K, Srinivasa Rao G, Dhanaraju MD. Ibuprofen<br />

loaded ethylcellulose / polystyrene microsphers an approch to get<br />

prolonged drug release with reduced burst effect and low ethycellulose<br />

content J Microencapsulation 2003; 20 (3): 289-302.<br />

87. Nizar Al-Zoubi, Alkhatib HS, Yasser Bustanji, Khaled Aiedeh, Starros<br />

Malamataris. Sustained-release of buspirone HCL by co spray-drying<br />

with aqueous polymeric dispersion. Eur J Pharma and Biopharm 2008<br />

;(69): 735-742.<br />

88. Seshasayan A, Sreenivasa RB, Prasanna R., Ramana Murthy KV.<br />

Studies on Release of Rifampicin from modified pulsincap technique.<br />

Indian J Pharm Sci 2001:337–339.<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 141


Annexure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 142


Annexure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 143


Annexure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 144


Annexure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 145


Annexure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 146


ERRATA<br />

Annexure<br />

DEPT. <strong>OF</strong> PHARMACEUTICS, LUQMAN COLLEGE <strong>OF</strong> PHARMACY, GULBARGA 147

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!