Вы находитесь на странице: 1из 206

STABILITY INDICATING SIMULTANEOUS

DETERMINATION OF NEW
MUSCLE RELAXANT DRUGS BY
CHROMATOGRAPHIC METHODS

A thesis submitted in partial fulfillment of the


requirements
for award of the degree of

Doctor of Philosophy

Research Guide:

Research Scholar:

Dr. P. U. PATEL

AMIN MAULIKKUMAR R.

M. Pharm., Ph.D.

M. Pharm
Reg. No. : PH/007/057/08

S. K. Patel College of Pharmaceutical Education and Research,


Ganpat University, Ganpat Vidhyanagar-384012, Gujarat,
NOVEMBER-2012

Certificate
This is to certify that, the thesis entitled Stability indicating
simultaneous determination of new muscle relaxant drugs by
chromatographic methods Submitted by Amin Maulikkumar
Rameshchandra of Kalol Institute of Pharmacy, Kalol is the bonafide
work completed under my supervision and guidance for the award of
Degree of Doctor of Philosophy in the Faculty of Pharmacy, Ganpat
Vidhyanagar. The experimental work included in the thesis was carried
out at Department of Pharmaceutical chemistry, Kalol Institute of
Pharmacy, Kalol and Shree S. K. Patel College of Pharmaceutical
Education and Research, under my supervision and the work is up to
my satisfaction.
Research Guide:

Dr. P. U. PATEL
M. Pharm., Ph.D.
Professor & Head, Department of Pharmaceutical Quality Assurance,
Shree S. K. Patel College of Pharmaceutical Education and Research,
Ganpat University, Ganpat Vidhyanagar-384012
Forwarded through:
Dr. R. K. PATEL
M. Pharm., Ph.D.
I/C Principal,
Shree S. K. Patel College of Pharmaceutical Education and Research,
Ganpat University, Ganpat Vidhyanagar-384012
Date:
Place: Ganpat Vidhyanagar

THESIS APPROVAL SHEET


The Ph. D. Thesis entitled Stability indicating simultaneous
determination of new muscle relaxant drugs by chromatographic
methods by Amin Maulikkumar Rameshchandra has been
approved for the award of the Degree of Doctor of Philosophy
under the Faculty of Pharmacy, Ganpat University, Ganpat
Vidhyanagar, Dist: Mehsana (Gujarat).

External Examiner:

Research Guide:

Dr. P. U. PATEL
M. Pharm., Ph.D.

Date:
Place: Ganpat Vidhyanagar

Certificate
This is to certify that the suggestion given by doctoral
committee during pre submission seminar held on 5 th May
2012,

vide

letter

of

Ganpat

University

no.

89/GNU/Ph.D./600/2012 dated 28th May, 2012 are duly


incorporate in the thesis.

Research Guide:

Dr. P. U. PATEL
M. Pharm., Ph.D.

Professor & Head,


Department of Pharmaceutical Quality Assurance,
Shree S. K. Patel College of Pharmaceutical Education and Research,
Ganpat University, Ganpat Vidhyangar-384012
Date:
Place: Ganpat Vidhyanagar

DECLARATION
I hereby declare that the topic entitled Stability

indicating

simultaneous determination of new muscle relaxant drugs by


chromatographic methods

which is submitted herewith to Ganpat

University for the award of Doctor of Philosophy in the Faculty of


Pharmacy is the result of work carried out by me at Kalol Institute of
Pharmacy, Kalol and Shree S. K. Patel College of Pharmaceutical
Education and Research, under guidance of Dr.P.U.Patel, Professor &
Head, Department of Pharmaceutical Quality Assurance, Shree S. K.
Patel College of Pharmaceutical Education and Research, Ganpat
University. I further declare that results of this work have not been
previously submitted for any degree or fellowship.

Date:
Place: Ganpat Vidhyangar

Amin Maulikkumar R.
M.Pharm

GANPAT UNIVERSITY
DECLARATION BY THE AUTHOR OF THE THESIS
I, Mr. Maulikkumar Rameshchandra Amin, Reg. No. PH/007/057/08 registered as a
research scholar of Ph.D. programe in the Department of Pharmacy, Ganpat University do
hereby submit my thesis, entitled: Stability indicating simultaneous determination of
new muscle relaxant drugs by chromatographic methods (herein referred to as my
thesis) in printed as well as in electronic forms for holding in the library of records of the
University.
I hereby declare that:

1. The electronic version of my thesis submitted herewith on CDROM is in PDF format.


2. My thesis is my original work of which the copyright vests in me and my thesis do
not infringe or violate the rights of anyone else.
3. The contents of the electronic version of my thesis submitted herewith are the same as
those submitted as final hard copy of my thesis after my viva voce and adjudication of
my thesis.
4.

I agree to abide by the terms and conditions of the Ganpat University Policy on
Intellectual Property (hereinafter Policy) currently in effect, as approved by the
competent authority of the university.

5.

I agree to allow the university to make available the abstract of my thesis to any user
in both hard copies (printed) and electronic forms.

6. For the Universitys own, non-commercial, academic use I grant to the University the
non-exclusive license to make limited copies of my thesis in whole or in part and to
loan such copies at the Universitys discretion to academic persons and bodies
approved from time to time by the University for non-commercial academic use. All
usage under this clause will be governed by the relevant fair use provisions in the
Policy and by the Indian Copyright Act in force at the time of submission of the
thesis.
7. I agree to allow the University to place such copies of the electronic version of my
thesis on the private intranet maintained by the University for its own academic
community.
8. I agree to allow the University to publish such copies of the electronic version of my
thesis on a public access website of the internet.

9. If in the opinion of the University my thesis contains patentable or copyrightable


material and if the University decides to proceed with the process of securing
copyrights and/or patents, I authorize the University to do so. I also undertake not to
disclose any of the patentable intellectual properties before being permitted by the
University to do so, or for a period of one year from the date of final thesis
examination, whichever is earlier.
10. In accordance with the Intellectual Property Policy of the University, I accept that
any commercializable intellectual property contained in my thesis is the joint property
of me, my co-workers, my supervisors and the Institute. I authorize the University to
proceed with protection of the intellectual property rights in accordance with
prevailing laws. I agree to abide by the provisions of the University Intellectual
Property Right Policy to facilitate protection of the intellectual property contained in
my thesis.
11. If I intend to file a patent based on my thesis when the University does not wish so, I
shall notify my intention to the University. In such case, my thesis should be marked
as patentable intellectual property and access to my thesis is restricted. No part of my
thesis should be disclosed by the University to any person(s) without my written
authorization for one year after my information to the University to protect the IP on
my own, within 2 years after the date of submission of the thesis or the period
necessary for sealing the patent, whichever is earliest.

Name of Research student

Name of Guide

AMIN MAULIKKUMAR R.

Dr. P. U. PATEL

M.Pharm.

Signature of Research student


Date:
Place: Ganpat Vidhyanagar

M.Pharm., Ph.D.

Signature of Guide

ACKNOWLEDGEMENT
Thesis is first step of research methodology, for every student during the course of
Ph.D. Today, at the acme of my thesis, with heartiness, I gratefully remember my research
guide, parents and my colleagues and all those hands that have contributed directly or
indirectly; as one flower make no garland. This presentation would not have taken shape
without their wholehearted encouragement and live involvement.
Teacher is a guide, philosopher and friend which I could experience in my respected
guide Dr. Paresh U. Patel, M. Pharm. Ph.D., Professor, Department of Pharmaceutical Quality
Assurance of Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat
Vidhyanagar. I wish to express my sincere gratitude for his constant guidance, supervision,
unceasing encouragement which paved the way for the successful completion of this
research work. His attitude towards work, his optimistic thinking, and ideas of work and
experimental has instilled me more confidence than before. It is a pleasure and privilege for
me to acknowledge gratefully the interest and attention so generously lavished by him.
It is with great pleasure and profound gratitude of reverence that I express my
esteemed Dr. Madhabhai M. Patel, Former Principal, Kalol Institute of Pharmacy, Kalol for
his valuable guidance during the course of my research work.
I warmly extend my acknowledgement to Dr. Atulbhai Patel, Managing director of
Umiya mata sanchalit samaj seva education trust for providing facilities during this course of
investigation.
I express my special thanks to Dr. R. K. Patel, I/C Principal of Shree S. K. Patel College
of Pharmaceutical Education and Research, Ganpat University, Ganpat Vidhyanagar for the
appreciable suggestions.
I would like to express my sincere thanks to honorable Shri Anilbhai T. Patel,
President, Ganpat University and respected Prof. L. N. Patel, Vice Chancellor, Ganpat
University for permitting me to pursue this work for Ph.D. degree.
Words are an inadequate medium to express my deep sense of gratitude to
Dr. B. N. Suhagia, Dean, Faculty of Pharmacy, Dharmsinh Desai University, Nadiad for the
helpful suggestions whenever required.

It gives immense pleasure to thank Dr. Deepa Patel, Mr. Ravi Shah, Mrs. Advaita
Patel and Mr. Kandarp Patel for their valuable support, co-operation and proper guidance
during this work.
I acknowledge the support of my colleagues Dr. Bhupendra Chauhan, Dr. Rajnikant
Patel, Dr. Samir Patel, Mr. Nileshbhai Patel, Ms. Jagruti Patel, Ms. Aditi Bariya, Mr. Rajesh
Keralia, Mr Chirag Patel and Mr. Kaushik Kanada for their kind help and encouragement.
I would like to sincerely thank Dr. L. J. Patel, Professor & Head, Department of
Pharmaceutical Chemistry of Shree S. K. Patel College of Pharmaceutical Education and
Research, Ganpat Vidhyanagar for his valuable suggestion.
I express my heartfelt gratitude to Ms. Mittal Patel, a librarian, Kalol Institute of
Pharmacy, Kalol for providing excellent library facility. I am thankful to Mr. Bhavesh Patel
(Store Keeper, Kalol Institute of Pharmacy, Kalol) who provided chemicals, during my work
in laboratory when I required. I owe a special thanks to Mr. Vijaybhai Raval for helped me in
maximum utilization of computer center.
I am thankful to my college Clerk Mr. Prayag Thakore and Mr. Surubha Vaghela as
well as non-teaching staff Sanjay, Jagdish, Chirag, Tushar, Alpesh, Raju, Suresh for providing
me moral support.
Research never possible without materials so I am heartly grateful to Zydus Cadila
Ltd, Ahmedabad for providing me the gift samples. My special thanks to Mr. Jitendra
Verma, Deputy Manager, Analytical Department, Zydus Research Centre, Ahmedabad.
I would fail in duty if I do not express my overriding debt to my Father Shri
Rameshchndra Amin, Mother Smt. Hemlattaben Amin, Brother Dr. Paragkumar Amin,
Bhabhi Mrs. Alaknanda Amin and nephew Preet who contributed to this research project in
countless ways. All I know is that without their care and faith in me it was impossible for me
to reach at this stage of my life.
Words can never express love of my wife Mrs. Jinali Amin for her constant
emotional support during hardships of this project.

November-2012

Mr.Maulikkumar R. Amin

INDEX
PARTICULARS OF CHAPTER
Content

Chapter
1

Introduction

Page
No.
1-46

1.1

Muscle relaxants

1-4

1.2

Drug profiles

4-9

1.3

Chromatography

9-25

1.4

Development of new analytical method

25-34

1.6

Stability indicating analytical method

34-41

1.7

References

42-46

Review of literature

47-75

Aim of the present work

Chemicals, glasswares and instruments

76
77-80

4.1

Chemicals

77

4.2

Active Pharmaceutical Ingredients

77

4.3

Glasswares

78

4.4

Instruments

78

4.5

Other requirements

79

4.6

References

80

Stability indicating method development and validation for

81-128

simultaneous estimation of Diclofenac potassium,


Chlorzoxazone and Paracetamol
5.1

Stability indicating HPLC method development and validation

81-103

for simultaneous estimation of Diclofenac potassium,


Chlorzoxazone and Paracetamol
5.2

Stability indicating HPTLC method for simultaneous estimation

104-126

of diclofenac potassium, Chlorzoxazone and Paracetamol


5.3

Statistical comparison between official and proposed methods

127

5.4

References

128

Stability indicating method development and

validation for

129-173

simultaneous estimation of Diclofenac potassium,


Paracetamol and Methocarbamol
6.1

Stability indicating HPLC method development for simultaneous

129-150

estimation of Diclofenac potassium, Paracetamol and


Methocarbamol
6.2

Stability indicating HPLC method for simultaneous estimation

151-171

of Diclofenac potassium, Paracetamol and Methocarbamol


6.3

Statistical comparison between official and proposed methods

172

6.4

References

173

Summary

174-175

Publications

176

List of Tables
Table
No.
1.3.1

Caption

Page
No.
22

Controlling sample retention by changing solvent strength

1.6.1

Various degradation conditions described for the stress testing

37

2.1.1

Official methods for paracetamol

47

2.1.2

Official methods for paracetamol in combination

50

2.1.3

Official methods for diclofenac potassium in combination

50

2.1.4

Official methods for chlorzoxazone in combination

51

2.1.5

Official methods for methocarbamol

52

2.2.1

Reported methods for paracetamol

53

2.2.2

Reported methods for diclofenac potassium

60

2.2.3

Reported methods for chlorzoxazone

64

2.2.4

Reported methods for methocarbamol

66

4.2.1

List of active pharmaceutical ingredients

78

5.1.1a

Results of optimization of mobile phase

89

5.1.1b

Linearity and range data for DIC, CHL and PCM by HPLC

91

5.1.2

Recovery study of DIC, CHL and PCM by HPLC

92

5.1.3

Intra-day & Inter-day precision of DIC, CHL and PCM by HPLC

92

5.1.4

Robustness data for DIC, CHL and PCM by HPLC

93

5.1.5

System suitability parameters for DIC, CHL and PCM by HPLC

93

5.1.6

Forced degradation study of marketed product by HPLC

102

5.2.1

Linearity and range data for DIC, CHL and PCM by HPTLC

114

5.2.2

Recovery study of DIC, CHL and PCM by HPTLC

115

5.2.3

Results of Intra-day & Inter-day precision DIC, CHL and PCM by

115

HPTLC
5.2.4

Robustness data for DIC, CHL and PCM by HPTLC

116

5.2.5

Result of Forced degradation study of drug products

125

5.3.1

Statistical comparison for DIC, CHL and PCM between proposed

127

methods

6.1.1a

Results of optimization of mobile phase

136

6.1.1b

Linearity and range data for DIC, PCM and MET by HPLC

139

6.1.2

Recovery study for DIC, PCM and MET by HPLC

139

6.1.3

Intra-day & Inter-day precision for DIC, PCM and MET by HPLC

140

6.1.4

Robustness data for DIC, PCM and MET by HPLC

140

6.1.5

System suitability parameters for DIC, PCM and MET by HPLC

141

6.1.6

Forced degradation study of DIC, PCM and MET by HPLC

149

6.2.1

Linearity and range data for DIC, PCM and MET by HPTLC

160

6.2.2

Recovery study for DIC, PCM and MET by HPTLC

160

6.2.3

Intra-day & Inter-day precision for DIC, PCM and MET by HPTLC

161

6.2.4

Robustness data for DIC, PCM and MET by HPTLC

161

6.2.5

Forced degradation study of marketed product by HPTLC

170

6.3.1

Statistical comparison for DIC, PCM and MET between proposed

172

methods

List of Figures
Figure No.

Caption

Page
No.
3

1.1.1

Site of action for muscle relaxants

1.3.3.1

Calculation of resolution for two adjacent bands 1 and 2

20

1.3.3.2

Measuring the relative valley height for two overlapping bands

21

1.3.3.3

Solvent selectivity triangle for composition of mobile phase

23

1.6.1

Flow chart for performing stress studies for photolytic degradation

38

1.6.2

Flow chart for performing stress studies for hydrolytic degradation

39

under acid and alkali conditions


1.6.3

Flow chart for performing stress studies for hydrolytic degradation

40

under neutral condition (in water)


1.6.4

41

5.1.1

Flow chart for performing stress studies for degradation under


oxidative
Conditions
Overlay UV spectra of DIC, CHL and PCM

5.1.2a

Blank Chromatogram of DIC, CHL and PCM

89

5.1.2b

Chromatogram of DIC, CHL and PCM by HPLC

90

5.1.3

Calibration curve of DIC by HPLC

90

5.1.4

Calibration curve of CHL by HPLC

91

5.1.5

Calibration curve of PCM by HPLC

91

5.1.6a

Chromatogram of acidic degradation of DIC by HPLC

94

5.1.6b

Chromatogram of acidic degradation of CHL by HPLC

95

5.1.6c

Chromatogram of acidic degradation of PCM by HPLC

95

5.1.7a

Chromatogram of alkaline degradation of DIC by HPLC

95

5.1.7b

Chromatogram of alkaline degradation of CHL by HPLC

96

5.1.7c

Chromatogram of alkaline degradation of PCM by HPLC

96

5.1.8a

Chromatogram of oxidative degradation of DIC by HPLC

96

5.1.8b

Chromatogram of oxidative degradation of CHL by HPLC

97

5.1.8c

Chromatogram of oxidative degradation of PCM by HPLC

97

83

5.1.9a

Chromatogram of thermal degradation of DIC by HPLC

97

5.1.9b

Chromatogram of thermal degradation of CHL by HPLC

98

5.1.9c

Chromatogram of thermal degradation of PCM by HPLC

98

5.1.10a

Chromatogram of neutral degradation of DIC by HPLC

98

5.1.10b

Chromatogram of neutral degradation of CHL by HPLC

99

5.1.10c

Chromatogram of neutral degradation of PCM by HPLC

99

5.1.11

Chromatogram of acidic degradation of marketed product by

99

HPLC
5.1.12

Chromatogram of alkaline degradation of marketed product by

100

HPLC
5.1.13

Chromatogram of oxidative degradation of marketed product by

100

HPLC
5.1.14

Chromatogram of thermal degradation of marketed product by

100

HPLC
5.1.15

Chromatogram of neutral degradation of marketed product by

101

HPLC
5.2.1

Overlay UV spectra of DIC, CHL and PCM

105

5.2.2

Chromatogram of CHL, PCM and DIC

112

5.2.3

Calibration curve for DIC by HPTLC

113

5.2.4

Calibration curve for CHL by HPTLC

113

5.2.5

Calibration curve for PCM by HPTLC

114

5.2.6a

Chromatogram of acidic degradation of DIC by HPTLC

117

5.2.6b

Chromatogram of acidic degradation of CHL by HPTLC

117

5.2.6c

Chromatogram of acidic degradation of PCM by HPTLC

118

5.2.7a

Chromatogram of alkaline degradation of DIC by HPTLC

118

5.2.7b

Chromatogram of alkaline degradation of CHL by HPTLC

118

5.2.7c

Chromatogram of alkaline degradation of PCM by HPTLC

119

5.2.8a

Chromatogram of oxidative degradation of DIC by HPTLC

119

5.2.8b

Chromatogram of oxidative degradation of CHL by HPTLC

119

5.2.8c

Chromatogram of oxidative degradation of PCM by HPTLC

120

5.2.9a

Chromatogram of thermal degradation of DIC by HPTLC

120

5.2.9b

Chromatogram of thermal degradation of CHL by HPTLC

120

5.2.9c

Chromatogram of thermal degradation of PCM by HPTLC

121

5.2.10a

Chromatogram of neutral degradation of DIC by HPTLC

121

5.2.10b

Chromatogram of neutral degradation of CHL by HPTLC

121

5.2.10c

Chromatogram of neutral degradation of PCM by HPTLC

122

5.2.11

Chromatogram of acidic degradation of marketed product by

122

HPTLC
5.2.12

Chromatogram of alkaline degradation of marketed product by

123

HPTLC
5.2.13

Chromatogram of oxidative degradation of marketed product by

123

HPTLC
5.2.14

Chromatogram of thermal degradation of marketed product by

123

HPTLC
5.2.15

Chromatogram of neutral degradation of marketed product by

124

HPTLC
6.1.1

Overlay UV spectra of DIC, PCM and MET

131

6.1.2a

Blank Chromatogram by HPLC

137

6.1.2b

Chromatogram of DIC, PCM and MET by HPLC

137

6.1.3

Calibration curve of DIC by HPLC

138

6.1.4

Calibration curve of PCM by HPLC

138

6.1.5

Calibration curve of MET by HPLC

139

6.1.6a

Chromatogram of acidic degradation of DIC by HPLC

142

6.1.6b

Chromatogram of acidic degradation of PCM by HPLC

142

6.1.6c

Chromatogram of acidic degradation of MET by HPLC

142

6.1.7a

Chromatogram of alkaline degradation of DIC by HPLC

143

6.1.7b

Chromatogram of alkaline degradation of PCM by HPLC

143

6.1.7c

Chromatogram of alkaline degradation of MET by HPLC

143

6.1.8a

Chromatogram of oxidative degradation of DIC by HPLC

144

6.1.8b

Chromatogram of oxidative degradation of PCM by HPLC

144

6.1.8c

Chromatogram of oxidative degradation of MET by HPLC

144

6.1.9a

Chromatogram of thermal degradation of DIC by HPLC

145

6.1.9b

Chromatogram of thermal degradation of PCM by HPLC

145

6.1.9c

Chromatogram of thermal degradation of MET by HPLC

145

6.1.10a

Chromatogram of neutral degradation of DIC by HPLC

146

6.1.10b

Chromatogram of neutral degradation of PCM by HPLC

146

6.1.10c

Chromatogram of neutral degradation of MET by HPLC

146

6.1.11

147

6.2.1

Chromatogram of acidic degradation of marketed product by


HPLC
Chromatogram of alkaline degradation of marketed product by
HPLC
Chromatogram of oxidative degradation of marketed product by
HPLC
Chromatogram of thermal degradation of marketed product by
HPLC
Chromatogram of neutral degradation of marketed product by
HPLC
Overlay UV spectra of DIC, PCM and MET

6.2.2

Chromatogram of DIC, PCM and MET by HPTLC

158

6.2.3

Calibration curve for DIC by HPTLC

159

6.2.4

Calibration curve of PCM by HPTLC

159

6.2.5

Calibration curve of MET by HPTLC

160

6.2.6a

Chromatogram of acidic degradation of DIC by HPTLC

162

6.2.6b

Chromatogram of acidic degradation of PCM by HPTLC

162

6.2.6c

Chromatogram of acidic degradation of MET by HPTLC

163

6.2.7a

Chromatogram of alkaline degradation of DIC by HPTLC

163

6.2.7b

Chromatogram of alkaline degradation of PCM by HPTLC

164

6.2.7c

Chromatogram of alkaline degradation of MET by HPTLC

164

6.2.8a

Chromatogram of oxidative degradation of DIC by HPTLC

164

6.2.8b

Chromatogram of oxidative degradation of PCM by HPTLC

165

6.2.8c

Chromatogram of oxidative degradation of MET by HPTLC

165

6.2.9a

Chromatogram of thermal degradation of DIC by HPTLC

165

6.2.9b

Chromatogram of thermal degradation of PCM by HPTLC

166

6.1.12
6.1.13
6.1.14
6.1.15

147
147
148
148
152

6.2.9c

Chromatogram of thermal degradation of MET by HPTLC

166

6.2.10a

Chromatogram of neutral degradation of DIC by HPTLC

166

6.2.10b

Chromatogram of neutral degradation of PCM by HPTLC

167

6.2.10c

Chromatogram of neutral degradation of MET by HPTLC

167

6.2.11

Chromatogram of acidic degradation of marketed product by

167

HPTLC
6.2.12

Chromatogram of alkaline degradation of marketed product by

168

HPTLC
6.2.13

Chromatogram of oxidative degradation of marketed product by

168

HPTLC
6.2.14

Chromatogram of thermal degradation of marketed product by

169

HPTLC

6.2.15

Chromatogram of neutral degradation of marketed product by


HPTLC

169

ABBREVIATION
GENERAL ABBREVIATION
IP- Indian Pharmacopeia
BP- British Pharmacopoeia
USP- United State Pharmacopeia
FDA- Food and Drug Administration
DIC- Diclofenac Potassium
CHL- Chlorzoxazone
MET- Methocarbamol
PCM- Paracetamol
SD- Standard Deviation
RSD- Relative Standard Deviation
l- Micro Liter
ml- Mili Liter
Ach- Acetylcholine
NaOH- Sodium hydroxide
HCl- Hydrochloric acid
H2O2- Hydrogen peroxide
Max- Maxima
HPLC- High Performance Liquid Chromatography
HPTLC- High Performance Thin Layer Liquid Chromatography
LC- Liquid Chromatography
Rf- Retention factor
tR- Retention time
AR- Analytical reagent
Min- Minute
hr- Hour
WHO- World Health Organization
Vs- Versus
H2O- Water

ODS- Octa Decyl Silane


ppm- Parts per million
AUC- Area Under Curve
LOD- Limit of Detection
LOQ- Limit of Quantification
UV- Ultra Violet
COA- Certificate of Analysis
Conc- Concentration
i.d.- Internal Diameter
Edn.- Edition
ICH- International Conference of Harmonization
KH2PO4- Mono potassium phosphate
GC- Gas Chromatography
ACN- Acetonitrile
PDA- Photo Diode Array

JOURNAL ABBREVIATION
Asian J. Research Chemistry- Asian Journal of Research Chemistry
Anal. Chem- Analytical Chemistry
Ann. Intern. Med.- Annals of Internal Meidicine
Arch Intern Med.- Archives of Internal Medicine
Curr Med Res Opin- Journal of Current Medical Research and Opinion
Drug Dev. Ind. Phrm.- Drug Development and Industrial Pharmacy
E Journal of Chemistry- European Journal of Chemistry
Eur J Pharmacol- European Journal of Pharmacology
Journal of AOAC Int.- Journal of AOAC International
J. of Chromatogr. B: Biomed. Sci. Appl.- Journal of Chromatography B:
Biomedical Sciences and Applications
J Biomed Sci and Res.- Journal of Biomedical Science and Research
Journal of Anal Bioanal Techniques- Journal of Analytical and Bioanalytical
Techniques
J. Chromatogr. B- Journal of Chromatography B

J Clin Pharmacol- Journal of Clinical Pharmacology


J. Pharm. Biomed. Anal.- Journal of Pharmaceutical and Biomedical Analysis
Clin Pharmacokinet.- Journal of Clinical Pharmacokinetics
J. Pharm. Biomed. Anal.- Journal of Biomedical Analysis
N Engl J Med.- The New England Journal of Medicine
Turk J Chemistry- Turkish Journal of Chemistry

SYMBOL ABBREVIATION
- Micro
mg- Milligram
- Lambda
- Beta
- Alpha
&- And
%- Percentage
ng- Nano gram
M- Molar
R2- Coefficient of variance
g/mol- Gram per mole
Rs- Resolution
L- Microlitre
C -Degree celsius
mL- Mili liter
Tf- Tailing factor
Tp- Theoretical plate
v/v/v- volume/volume/volume
w/w- weight by weight
mM- Mili molar
- Standard deviation
DL- Detection limit
QL- Quantification limit

Chapter 1

Introduction

1. INTRODUCTION
1.1 Muscle relaxants
Skeletal muscle relaxants are a heterogeneous group of medications that are
commonly used to treat two different types of underlying conditions: spasticity from
upper motor neuron syndromes and muscular pain or spasms from peripheral
musculoskeletal conditions. Skeletal muscle relaxants are drugs that act peripherally
at neuromuscular junction/muscle fibre itself or centrally in the cerebrospinal axis to
reduce muscle tone and paralysis. The neuromuscular blocking agents are used
primarily in conjugation with general anesthetics to provide muscle relaxation for
surgery, while centrally acting muscle relaxants are used mainly for painful muscle
spasms and neurological conditions. Although these drugs have been classified into
one class, the Food and Drug Administration (FDA) has approved only baclofen,
dantrolene, and tizanidine in this class for the treatment of spasticity; tizanidine and
the remainder of the skeletal muscle relaxant class are approved for treatment of
musculoskeletal conditions. Spasticity is a clinical condition that is a motor disorder
characterized by increase in tonic stretch reflexes (muscle tone) with exaggerated
tendon jerks, resulting from hyper excitability of the stretch reflex, as one component
of the upper motor neuron syndrome.Spasticity from the upper motor neuron
syndrome can result from a variety of conditions that affect the brain or the spinal
cord such as: multiple sclerosis, spinal cord injury, traumatic brain injury, cerebral
palsy, and post-stroke syndrome. In many patients with these chronic conditions,
spasticity can be disabling and painful with a marked effect on their functional ability
and quality of life.
A muscle relaxant is a drug which affects skeletal muscle function and decreases the
muscle tone. It may be used to alleviate symptoms such as muscle spasm and pain,
and hyperreflexia. The term "muscle relaxant" is used to refer to two major
therapeutic groups: neuromuscular blockers and spasmolytics. Neuromuscular
blockers act by interfering with transmission at the neuromuscular end plate and have
no CNS activity. They are often used during surgical procedures and in intensive care
and emergency medicine to cause paralysis[1-2].
Spasmolytics, also known as "centrally-acting" muscle relaxants, are used to alleviate
musculoskeletal pain and spasms and to reduce spasticity in a variety of neurological

Ganpat University

Chapter 1

Introduction

conditions. While both neuromuscular blockers and spasmolytics are often grouped
together as muscle relaxants the term is commonly used to refer to spasmolytics only.
Muscles can be divided into two classes, the voluntary or skeletal muscles and the
involuntary or smooth muscles. The heart muscle, the myocardium, is a unique type
of muscle that does not fit into either category. Skeletal muscles are those that are
involved in movement of arms, legs and under voluntary control. Smooth muscles are
those that are not under conscious control. The muscles in the digestive organs are
smooth muscles.
Usually it is the skeletal or striated muscles that will require therapy for painful spasm
or will need to be relaxed to allow the surgeon to gain access to the abdomen easily.
Muscle spasm may be associated with a trauma or may be brought on by multiple
sclerosis, cerebral palsy, stroke, or an injury to the spinal cord. Severe cold, an
interruption of blood supply to a muscle, or overexertion of the muscle also can lead
to spasms. A muscle spasm actually is an increase in muscle tone brought on by an
abnormality in motor control by the spinal nerves.
Skeletal muscles are controlled by large nerves in the spinal cord. The nerve cell or
neuron is part of the spinal cord, but its projections, the axon and the many dendrites
course outward to connect to muscle cells. The nerve axon is a sensory device that
senses the muscle cells current condition. The dendrites are motor fibers that deliver
the instructions to change its state to the muscle fiber. The area at which the muscle
and nerve connect is called the neuromuscular junction. It is here that the end releases
a chemical called a neurotransmitter that crosses the microscopic space between the
nerve and muscle and causes the desired response. Five such neurotransmitters have
been described: acetylcholine, serotonin, norepinephrine, glycine, and gammaamminobutyric acid or GABA.
Neuromuscular blocking agent (tubocurarine, pancuronium, vancuronium) cause
inhibition of muscle twitches but there was no effect if the blood supply of the hind
limb was occluded. The only possible site of action could be the neuromuscular
junction. The competitive blockers have affinity for the nicotinic cholinergic receptors
at muscle end-plate[3].
Common musculoskeletal conditions causing tenderness and muscle spasms include
fibromyalgia, tension headaches, myofascial pain syndrome, and mechanical low back
or neck pain. In these conditions, muscle spasm is related to local factors involving
the affected muscle groups. There is no increased tone or reflex. These conditions
Ganpat University

Chapter 1

Introduction

are usually acute and occur more commonly than spasticity in clinical practice. They
can cause significant disability and pain in some patients. Skeletal muscle relaxants
are one of several classes of medications such as anti-inflammatory drugs and pain
relievers that are used to treat these conditions.

Figure 1.1.1 site of action for muscle relaxants


Chlorzoxazone is a centrally acting muscle relaxant used to treat muscle spasm and
the resulting pain or discomfort. It acts on the spinal cord by depressing reflexes.
Aceclofenac is a phenylacetic acid derivative with analgesic and anti-iflammatory
activity and an improved gastrointestinal tolerance compared with the other NSAIDs,
such as diclofenac.
Paracetamol is a widely-used analgesic and antipyretic, unlike aspirin, it is not a very
effective anti-inflammatory agent. It is well tolerated, lacks many of the side-effects
of aspirin, and is available over-the-counter, so it is commonly used for the relief of
fever, headaches, and other minor aches and pains. Paracetamol is also useful in the
management of more severe pain, where it allows lower dosages of additional nonsteroidal anti-inflammatory drugs (NSAIDs) to be used, thereby minimizing overall
side-effects. It is also used in combination with opioid analgesics[4].

Ganpat University

Chapter 1

Introduction

Classification of muscle relaxant drugs:


1. Peripherally acting muscle relaxants
Mechanism of action:
Competitive block: - The site of action of both competitive and depolarizing blockers
is the end plate of skeletal muscle fibres. The competitive blockers have affinity for
nicotinic cholinergic receptors at muscle end plate, but have no intrinsic activity.
Competitive blockers also block prejunction nicotinic receptors located on motor
nerve endings. Since activation of these receptors by Ach normally facilitates
mobilization of additional quanta of Ach from the axon to the motor nerve endings,
their blockage contributes to depression of neuromuscular transmission.
Skeletal muscles:2. Centrally acting muscle relaxant drugs:
These drugs reduce skeletal muscle tone by selective action in the cerebrospinal axis,
without altering consciousness. They selectively depress spinal and supraspinal
polysynaptic reflexes involved in the regulation of muscle tone without significantly
affecting monosynaptically mediated stretch reflex. Polysynaptic pathways in the
ascending reticular formation which are involved in maintenance of wakefulness are
also depressed.

1.2 Drug Profiles


1.2.1 Chlorzoxazone[5-7]
Systematic (IUPAC) name: 5-Chlorobenzoxazol-2-ol
Synonyms: Chlorobenzoxazolinone
Structure:
O
OH
Cl

Molecular Formula: C7H4 Cl NO2


Mol. mass: 169.565 g/mole
Description: Colourless or white crystalline powder
Melting point: 190 to 194C
Solubility:- Sparingly soluble in water, soluble in acetone, methanol, and
isopropanol; freely soluble in aqueous solutions of alkali hydroxides and ammonia.
Dissociation constant (pKa): 8.0
Ganpat University

Chapter 1

Introduction

Partition Coefficient: 1.6


Half life: 1 hr
UV detection: 280nm
Category: neuromuscular blocker
Mechanism of action[8]:
Chlorzoxazone a centrally acting acting agent for painful musculoskeletal condition it
primarily act at the level of the spinal cord and subcortical area of brain. It inhibits
degranulation of mast cells, subsequently preventing the release of histamine and
slow-reacting substance of anaphylaxis, mediators of type I allergic reactions. It
reduces release of inflammatory leukotrienes. It acts by inhibiting calcium and
potassium influx which would lead to neuronal inhibition and muscle relaxation. It
acts primarily at the level of the spinal cord and subcortical areas of brain where it
inhibits multisynaptic reflex arcs involved in producing and maintaining skeletal
muscle spasm.
Pharmacokinetics[9]:
It is absorbed orally and undergoes first pass metabolism and is excreted by kidney.
t1/2 is 2-3 hrs. It is indicated in spasticity due to neurological disorders and in painful
muscle spasms of spinal origin.
Indications:
For acute and chronic treatment of signs and symptoms of osteoarthritis and
rheumatoid arthritis.
Contraindications: hypersensitivity, liver disease, porphyria, lactation
Adverse effects: headache, gastric irritation, nausea
Special precautions: reduce dose is necessary, avoid alcohol as it has an additive
effect and increase the depression.
1.2.2 Paracetamol[10-12]
Systematic (IUPAC) name: N-(4-Hydroxyphenyl)acetamide
Synonyms: Acetaminophen; N-Acetylpaminophenol
Structure:
HO
O

N
H

Ganpat University

CH3

Chapter 1

Introduction

Molecular Formula: C8H9NO2


Mol. mass: 151.2 gm/mole
Description: White crystals or crystalline powder
Melting point: 169 to 170 C
Solubility:- Very slightly soluble in cold water, considerably more soluble in hot
water; soluble in ethanol, methanol, dimethylformamide, ethylene dichloride, acetone,
and ethyl acetate; very slightly soluble in chloroform; slightly soluble in ether;
practically insoluble in petroleum ether, pentane, and benzene
Dissociation constant (pKa): 9.5
Partition Coefficient: 0.5
Log P: 4.21
Half life: 1-3 hrs.
UV detection: 245nm
Category: analgesics
Mechanism of action:
Paracetamol has analgesic and antipyretic action with weak anti inflammatory
activity. These effects are related to inhibition of prostaglandin synthesis.
Pharmacokinetics[13]:
Paracetamol is rapidly absorbed on oral administration. Peak plasma levels are
achieved within to 1 hour. It is metabolized in the liver and metabolites are
excreted in urine as conjugation products of glucuronic and sulfuric acid. The ability
of infants liver for glucuronidation of paracetamol is poor and this may result in
enhanced toxicity of the drug in neonates.
Indications:
Dosage: Paracetamol is used as analgesic and antipyretic. The total daily dose not
exceed than 2.5gm in adults. It can be used in a liquid dosage form in children.
Contraindications:
Adverse effects[14]: Paracetamol may cause fever, neutropenia, thrombocytopenia,
and skin reactions. Larger doses produce extensive damage to the liver and may cause
death due to liver failure. The drug may produce anemia as a result of haemolysis.

Ganpat University

Chapter 1

Introduction

1.2.3 Diclofenac potassium[15-18]:Systematic (IUPAC) name: 2-[(2,6-dichlorophenyl)amino]benzeneacetic acid


Synonyms: Diclophenac
Structure:
Cl
O K+
NH
O
Cl

Molecular Formula: C14H10Cl2KNO2


Mol. mass: 296.2 gm/mole
Description: Yellowish to white crystals or crystalline powder
Melting point: 156 to 158 C
Solubility: Soluble in water and methanol
Dissociation constant (pKa): 4.2
Partition Coefficient: 4.5
Log P: 4.21
Half life: 1-2 hrs.
UV detection: 254nm
Category: Antipyretic-analgesics agent
Mechanism of action[19]:
Diclofenac potassium tablets are a non-steroidal anti-inflammatory drug (NSAID) that
exhibits anti-inflammatory, analgesic, and antipyretic activities in animal models. It
inhibits both leukocyte migration and the enzyme cylooxygenase (COX-1 and COX2) which leads to peripheral inhibition of prostaglandin synthesis. As prostaglandins
sensitize pain receptors, inhibition of their synthesis is responsible for the analgesic
effects of diclofenac. Antipyretic effects may be due to action on the hypothalamus,
resulting in peripheral dilation, increased cutaneous blood flow, and subsequent heat
dissipation.
Pharmacokinetics:
It is well absorbed orally and metabolized and excreted both in urine and bile. The
plasma t1/2 is 5 hrs. Though, it has good tissue penetrability and concentration is
synovial fluid is maintained for 3 times longer period than in plasma, excreting
extended therapeutic action in joints.
Ganpat University

Chapter 1

Introduction

Indications[20]: It is used mainly in rheumatoid arthritis, severe osteoarthritis and in


ankylosing spondylitis.
Contraindications:
It is contraindicated in patients with peptic ulcer, hypersensitivity of diclofenac or
other NSAIDs. Also, patients having impaired cardiac, renal, hepatic function, and
asthma.
Adverse effects:
It produces mild epigastric pain, nausea, dizziness, and rashes. Gastric ulcer and
bleeding are less common.
Reversible elevation of serum aminotransferases has been reprted more commonly;
kidney damage is rare.
1.2.4 Methocarbamol[21-23]
Systematic (IUPAC) name: 3-(2-methoxyphenoxy)-l,2-propanediol-1-carbamate
Synonyms: Guaifenesin carbamate
Structure:
OH
H2N

O
O

Molecular Formula: C11H15NO5


Mol. mass: 241.2gm/mole
Description: Yellowish to white crystalline powder
Melting point: 94 to 96 C
Solubility: Soluble in water and methanol
Dissociation constant (pKa):
Partition Coefficient: Log P: 0.63
Half life: 1-2 hrs.
UV detection: 275nm
Category: Muscle relaxant
Mechanism of action[24]:
The mechanism of action of methocarbamol in humans has not been established, but
may be due to central nervous system depression. It has no direct action on the
contractile mechanism of striated muscle, the motor end plate or the nerve fiber11-13
Ganpat University

Chapter 1

Introduction

Pharmacokinetics:
Indications:
For use as an adjunct to rest, physical therapy, and other measures for the relief of
discomforts associated with acute, painful musculoskeletal conditions. It is also given
along with surgical procedure to reduce pain.
Contraindications:
Coma or pre-coma states, brain damage, myasthenia gravis, renal impairment,
epilepsy, pregnancy and lactation
Adverse effects:
It causes drowsiness, confusion, gastric irritation, and sedation. It also produces
muscular weakness, sedation, malaise, light headache, and sometimes diarrhoea.

1.3. Chromatography
It is a special separation process for complex mixture and very similar component
with great precision. Chromatography can purify basically any soluble or volatile
substance if the right adsorbent material, carrier fluid and operating conditions are
employed. A mixture of various components enters a chromatography process and
different components are flushed through the system at different rates. These
differential rates of migration as the mixture moves over adsorptive materials provide
separation. Repeated sorption/desorption acts that take place during the movement of
sample over the stationary phase. The smaller the affinity a molecule has for
stationary phase, the shorter time spent in a column.
HPLC is one of the classes of liquid chromatography according to the nature of the
stationary and mobile phase. It has gained importance in analytical chemistry due to
its high resolution capacity, sensitivity and specificity. The separation in HPLC is
done by partitioning between a mobile phase and stationary column material and
depends on the solubilities of solute. Mobile phase is pumped at a high pressure
through a packed column with fine particles of silica or chemically modified silica,
etc. The sample is injected and the solute after separation, enter the detector, the data
from which can be quantified. Special technique used for separation in HPLC is
isocratic and gradient elution, where elution strength of elute is increased during a run
by changing polarity, pH or ionic strength. HPLC can be used to resolve and
determine the active drug and small amount of impurity in active drug.
Ganpat University

Chapter 1

Introduction

Pharmaceutical compounds and preparation often cause chemical or physical


instabilities. The instabilities may produce toxic reaction products, reduced activity of
compound or produce unusable product. Stability testing is therefore carried out to
ensure that deterioration does not exceed an acceptable level in order to ensure safety
of patient and maintain activity of product. Developing stability indicating method is
necessary to carry out any stability study. Stress testing of drugs was performed
according to the International Conference on Harmonization (ICH) guideline in order
to validate method. Stress testing showed that drug underwent acid, alkaline and
oxidative degradation, on other hand, it showed stability towards photo & thermal
degradation.
1.3.1 History of Chromatography
Prior to the 1970s, few reliable chromatographic methods were commercially
available to the laboratory scientist. During 1970's, most chemical separations were
carried out using a variety of techniques including open-column chromatography,
paper

chromatography,

and

thin-layer

chromatography.

However,

these

chromatographic techniques were inadequate for quantification of compounds and


resolution between similar compounds. During this time, pressure liquid
chromatography began to be used to decrease flow through time, thus reducing
purification times of compounds being isolated by column chromatography. However,
flow rates were inconsistent, and the question of whether it was better to have
constant flow rate or constant pressure was debated. High-pressure liquid
chromatography was developed in the mid-1970 and quickly improved with the
development of column packing materials and the additional convenience of on-line
detectors. In the late 1970's, new methods including reverse phase liquid
chromatography allowed for improved separation between very similar compounds.
By the 1980's HPLC was commonly used for the separation of chemical compounds.
New techniques improved separation, identification, purification and quantification
far above the previous techniques. Computers and automation added to the
convenience of HPLC. Improvements in this type of columns and thus reproducibility
were made as such terms as micro-column, affinity columns, and Fast HPLC began to
immerge. The past decade has seen a vast undertaking in the development of the
micro-columns, and other specialized columns. The dimensions of the typical HPLC
column are varying in length with an internal diameter between 3-5 mm. The usual
diameter of micro-columns, or capillary columns, ranges from 3 m to 200 m. Fast
Ganpat University

10

Chapter 1

Introduction

HPLC utilizes a column that is shorter than the typical column, with a length of about
3 mm long, and they are packed with smaller particles.
Currently, one has the option of considering over different types of columns for the
separation of compounds, as well as a variety of detectors to interface with the HPLC
in order to get optimal analysis of the compound. We hope this review will provide a
reference, which all levels of HPLC users will be able to find quick answers to their
HPLC problems. Although HPLC is widely considered to be a technique mainly for
biotechnological, biomedical and biochemical research as well as for the
pharmaceutical industry, these fields currently comprise only about 50% of HPLC
users. Currently HPLC is used by a variety of fields including cosmetics, energy, food
and environmental industries[25].
Chromatography encompasses a diverse and important group of methods that permits
the scientist to separate closely related components of complex mixtures, many of
these separations are impossible by other means. In all chromatographic separations,
the sample is dissolved in a mobile phase, which may be a gas, liquid or a
supercritical fluid. This mobile phase is the forced through an immiscible stationary
phase, which is fixed in a column or on a solid surface. The two phase or chosen so
that the components of the sample distribute themselves between mobile and
stationary phase to varying degrees. In contrast, components that are weakly held by
stationary phase travel rapidly. As a consequence of these differences in mobility,
sample components separated into discrete bands that can be analyzed qualitatively
and or quantitatively. Each component has a characteristic time of passage through
the system, called a "retention time." Chromatographic separation is achieved when
the retention time of the analyte differs from that of other components in the sample.
A chromatograph takes a chemical mixture carried by liquid or gas and separates it
into its component parts as a result of differential distributions of the solutes as they
flow around or over a stationary liquid or solid phase. Various techniques for the
separation of complex mixtures rely on the differential affinities of substances for a
gas or liquid mobile medium and for a stationary absorbing medium through which
they pass; such as paper, gelatin, alumina or silica. A chromatogram is the visual
output of the chromatograph. Different peaks or patterns on the chromatograph
correspond to different components of the separated mixture. Analytical
chromatography is used to determine the identity and concentration of molecules in a
mixture. Preparative chromatography is used to purify larger quantities of a molecular
Ganpat University

11

Chapter 1

Introduction

species. Most of the following refers to analytical chromatography. This is a method


used to divide or separate mixtures[26-27].
1.3.2 Types of chromatography
It is a technique by which the components in a sample, carried by the liquid or
gaseous phase, are resolved by sorption-desorption steps on the stationary phase. A
more fundamental classification of chromatographic methods is one based upon the
type of mobile and stationary phases and the kinds of equilibrium involved in the
transfer of the solutes between phases. Classification of column chromatography
techniques is as follows.
1.3.2.1 High Performance Liquid Chromatography (HPLC)
HPLC chromatographic separation is based on interaction and differential partition of
the sample between the mobile liquid phase and the stationary phase. The commonly
used chromatographic methods can be roughly divided into the following groups, not
necessarily in order of importance:
1. Chiral
2. Ion--exchange
3. Ion--pair/affinity
4. Normal phase
5. Reversed phase
6. Size exclusion
1) Chiral Chromatography
Separation of the enantiomers can be achieved on chiral stationary phases by
formation of diastereomers via derivatizing agents or mobile phase additives on
achiral stationary phases. When used as an impurity test method, the sensitivity is
enhanced if the enantiomeric impurity elutes before the enantiomeric drug.
2) Ion-exchange Chromatography
Separation is based on the charge-bearing functional groups, anion exchange for
sample negative ion (X), or cation exchange - for sample positive ion (X), gradient
elution by pH is common.
3) Ion-pair/Affinity Chromatography
Separation is based on a chemical interaction specific to the target species. The more
popular reversed phase mode uses a buffer and an added counter-ion of opposite
charge to the sample with separation being influenced by pH, ionic strength,
temperature, concentration of and type of organic co-solvent(s). Affinity
Ganpat University

12

Chapter 1

Introduction

chromatography, common for macromolecules, employs a ligand (biologically active


molecule bonded covalently to the solid matrix), which interacts with its homologous
antigen (analyte) as a reversible complex that can be eluted by changing buffer
conditions.
4) Normal Phase Chromatography
Normal phase chromatography is a chromatographic technique that uses organic
solvents for the mobile phase and a polar stationary phase. Here, the less polar
components elute faster than the more polar components.
5) Reversed Phase Chromatography
The test method most commonly used in reversed phase HPLC method. UV detection
is the most common detection technique. Reversed phase chromatography, a bonded
phase chromatographic technique, uses water as the base solvent. Separation based on
solvent strength and selectivity also may be affected by column temperature and pH.
In general, the more polar components elute faster than the less polar components.
UV detection can be used with all chromatographic techniques. The concern for this
type of detector is the loss of sensitivity with lamp aging and varying sensitivity at the
low level depending on design and/or manufacturer. A point to note is that
observations on the HPLC chromatograms, by UV detection in combination with
reversed-phase HPLC, may not be a true indication of the facts for the following
reasons:
Compounds much more polar than the compound of interest may be masked
(elute together) in the solvent front or void volume.
Compounds very less polar than the analyte may elute either late during the
chromatographic run or retained in the column.
Compounds with lower UV extinction coefficients or different wavelength
maxima may not be detectable at the low level relative to the visibility of the
analyte since only one wavelength is normally monitored.
6) Size Exclusion Chromatography
It is also known as gel permeation or filtration, separation is based on the molecular
size or hydrodynamic volume of the components. Molecules that are too large for the
pores of the porous packing material on the column elute first, small molecules that
enter the pores elute last and the elution rates of the rest depend on their relative
sizes[28-31].

Ganpat University

13

Chapter 1

Introduction

1.3.2.2 Gas Chromatography (GC)


Gas chromatography is based on the volatilized sample transported by the carrier gas
as the moving phase through the stationary phase of the column where separation
takes place by the sorption/desorption process. Samples for gas chromatographic
analysis are normally low molecular weight compounds that are volatile and stable at
high temperature. In this respect, residual solvents in drug substances and drug
products are suitable for gas chromatographic analysis. Chemical derivatives can also
be formed to achieve volatility and thermal stability.
Common detectors are flame ionization (FID) for carbon-containing compounds,
electron capture (ECD) for halogenated compounds, flame photometric (FPD) for
compounds containing sulphur or phosphorous and nitrogen-phosphorous (NPD) for
compounds containing nitrogen or phosphorous. Chiral separation also can be
achieved by gas chromatography. The capillary column that provides improved
separation by resolution and analysis speed is rapidly replacing the packed column.
The location of the analyte on the gas chromatogram is described by retention time
(R), which is similar to HPLC.
1.3.2.3 Thin-Layer Chromatography (TLC)
Thin-layer chromatography is the simplest of the more common chromatographic
techniques. Separation is based on migration of the sample spotted on a coated
(stationary phase) plate with one edge dipped in a mixture of solvents (mobile phase).
The whole system is contained in an enclosed tank Detection techniques include
fluorescence, UV and sprays (universal and specific) for compounds that are not
naturally colored. The location of the analyte on the TLC plate is described by the R
value which is the ratio of the migration distance of the compound of interest to the
mobile phase front Of the three techniques, gas, liquid and thin-layer, TLC is the most
universal test method as all components are present on the plate and with appropriate
detection techniques, all components can be observed. However, it normally is not as
accurate or sensitive as HPLC. TLC has a higher analytical variation than HPLC.
Planar Chromatography as opposed to column chromatography (e.g. GC, HPLC)
utilizes a flat (planar) stationary phase for separation. In Thin-Layer Chromatography
(TLC) this stationary phase is supported by a glass plate or a foil (plastic or
aluminum). Again unlike column separations, the TLC plate constitutes an open
system, which passes through the individual steps of the TLC analysis in an off-line
mode.The relative independence of sample application, chromatogram development,
Ganpat University

14

Chapter 1

Introduction

detection, etc. in time and location makes possible the parallel analysis of many
samples on the same plate. The most advanced form of instrumental TLC is
commonly called high performance thin-layer chromatography (HPTLC), but the term
does not simply imply instrumental TLC on special high performance layers. HPTLC
is an entire concept that includes a widely standardized methodology based on
scientific facts as well as the use of validated methods for qualitative and quantitative
analysis. Sophisticated instruments, controlled by an integrated software platform
ensure to the highest possible degree the usefulness, reliability, and reproducibility of
generated data. HPTLC is therefore the term for a method that meets all quality
requirements of todays analytical labs even in a fully regulated environment.
Initial costs for an HPTLC system as well as maintenance, and cost per sample still
remain comparatively low and all advantages derived from the planar separation
principle are certainly maintained. The possibility of visual evaluation of separated
samples on the plate is one of the most valuable aspects of TLC. It reaches a
completely new dimension in HPTLC through the use of modern techniques for
generating and evaluating digital images.
Features of HPTLC:
The advantages of this off-line arrangement as compared with an on-line process,
such as column high-performance liquid chromatography (HPLC), have been outlined
and include the following:
Technically, it is simple to learn and operate.
Several analysts work simultaneously on the system.
Lower analysis time and less cost per analysis.
Low maintenance cost.
Visual detection possible-as it is an open system.
Availability of a great range of stationary phases with unique selectivity for
mixture

components.

Chromatographic

layer

(sorbent)

requires

no

regeneration as TLC/HPTLC plates are disposable.


Ability to choose solvents for the mobile phase is not restricted by low UV
transparency or the need for ultra-high purity. Corrosive and UV-absorbing
mobile phases can be employed.
No prior treatment for solvents like filtration and degassing.

Ganpat University

15

Chapter 1

Introduction

There is no possibility of interference from previous analysis as fresh


stationary and mobile phases are used for each analysis. No carry over, hence
no contamination.
Repetition of densitometric evaluation of the same sample can be achieved
under different conditions without repeating the chromatography to optimize
quantification, since all sample fractions are stored on the TLC/HPTLC plate.
Samples rarely require cleanup.
High sample throughput since several samples can be chromatographed
simultaneously.
Lower expenditure of solvent purchase and disposal since the required amount
of mobile phase per sample is small. In addition, it minimizes exposure risks
of toxic organic effluents and reduces possibilities of environment pollution.
Accuracy and precision of quantification is high because samples and
standards are chromatographed and measured under the identical experimental
conditions on a single TLC/HPTLC plate.
Sensitivity limits of analysis are typically at nanogram (ng) to pictogram (pg)
levels.
Use of different universal and selective detection methods
HPTLC is a modern adaptation of TLC with better and advanced separation efficiency
and detection limits.
HPTLC methodology:
Set the analytical objective first that may be quantification or qualitative identification
or separation of two components/multicomponent mixtures or optimization of
analysis time before starting HPTLC. Method for analyzing drugs in multicomponent
dosage forms by HPTLC demands primary knowledge about the nature of the sample,
namely, structure, polarity, volatility, stability, and the solubility parameter. Method
development involves considerable trial and error procedures. The most difficult
problem usually is where to start, with what kind of mobile phase.
Selection of stationary phase is quite easy, that is, to start with silica gel which is
reasonable and nearly suits all kind of drugs. Mobile phase optimization is carried out
by using three level techniques. First level involves use of neat solvents and then by
finding some such solvents which can have average separation power for the desired
drugs. Second level involves decreasing or increasing solvent strength using hexane
or water for respective purposes. Third level involves trying of mixtures instead of
Ganpat University

16

Chapter 1

Introduction

neat solvents from the selected solvents of first and second level which can further be
optimized by the use of modifier like acids or bases. Analytes are detected using
fluorescence mode or absorbance mode. But, if the analytes are not detected perfectly
than it needs change of stationary phase or mobile phase or need the help of pre or
post chromatographic derivatization. Optimization can be started only after a
reasonable chromatogram which can be done by slight change in mobile-phase
composition. This leads to a reasonable chromatogram which has all the desired peaks
in symmetry and well separated. Procedure for HPTLC method development is
outlined as follow.
Stationary phase:
HPTLC can be regarded as the most advanced form of modern TLC. It uses HPTLC
plates featuring small particles with a narrow size distribution. As a result,
homogenous layers with a smooth surface can be obtained. HPTLC uses smaller
plates (10x10 or 10x20 cm) with significantly decreased development distance
(typically 6 cm) and analysis time (7-20 min). HPTLC plates provide improved
resolution, higher detection sensitivity, and improved in situ quantification and are
used for industrial pharmaceutical densitometric quantitative analysis.
Mobile phase:
The selection of mobile phase is based on adsorbent material used as stationary phase
and physical and chemical properties of analyte.
General mobile-phase systems that are used based on their diverse selectivity
properties are diethyl ether, methylene chloride, and chloroform combined
individually or together with hexane as the strength-adjusting solvent for normalphase TLC and methanol, acetonitrile, and tetrahydrofuran mixed with water for
strength adjustment in reversed-phase TLC.
Accurate volumetric measurements of the components of the mobile phase must be
performed separately and precisely in adequate volumetric glassware and shaken to
ensure proper mixing of the content.
Volumes smaller than 1 ml are measured with a suitable micropipette. Volumes up to
20 ml are measured with a graduated volumetric pipette of suitable size. Volumes
larger than 20 ml are measured with a graduated cylinder of appropriate size. To
minimize volume errors, developing solvents are prepared in a volume that is
sufficient for one working day.

Ganpat University

17

Chapter 1

Introduction

Layer prewashing:
Plates are generally handled only at the upper edge to avoid contamination. Usually
plates are used without pretreatment unless chromatography produces impurity fronts
due to contamination of the plate. For reproducibility studies and quantitative
analysis, layers are often prewashed using 20 ml methanol (generally, methanol is
used as a prewashing solvent; however, a mixture of methanol and ethyl acetate or
even mobile phase of the method may also be used) per trough in a 20x10 cm twintrough chamber (TTC). Up to two 20x10 cm or four 10x10 cm plates can be
developed back-to-back in each trough of the TTC.
Preparation of plate:
Precoated layers: TLC plates can be made in any lab with suitable apparatus.
However such layers do not adhere well to the glass support. Precoated plates that use
small quantities of very high molecular weight polymer as binder overcomes most
limitations of a home-made layer. Precoated layers are reasonably abrasion resistant,
very uniform in layer thickness, reproducible, preactivated, and ready to use. They are
available with glass or aluminum or polyester support. Aluminum foil plates are less
expensive to buy, cheaper, can be cut, and therefore easy to carry around or transport
or mail. Glass plates are the best for highest quality of results. Most often, layers
containing a fluorescent indicator F 254 are used. This enables the visualization of
samples in a UV cabinet very simply, instantly, and in a nondestructive
manner.Commonly used size of plates in TLC is 20x20 cm and in HPTLC 20x10 cm
or 10x10 cm is widespread.
Sample application:
In Thin-Layer Chromatography manual sample application with capillaries is usually
performed for simple analyses. Sample volumes of 0.5 to 5 L can be applied as spots
onto conventional layers without intermediate drying. HPTLC layers take up to 1 L
per spot. More demanding qualitative, quantitative, and preparative analyses or
separations are made possible only by instruments for band wise application of
samples using the spray-on technique. Particularly HPTLC takes full advantage of the
gain in separation power and reproducibility available by precise positioning and
volume dosage.
Spray-on technique:
With LINOMAT, an ideal instrument for sample application for instrumental and
preparative

Thin-Layer

Ganpat University

Chromatography

samples

are

sprayed

onto

the
18

Chapter 1

Introduction

chromatographic layers in the form of narrow bands. This technique allows larger
volumes to be applied than by contact transfer (spotting). During the spraying the
solvent of the sample evaporates almost entirely concentrating the sample into narrow
band of selectable length. Starting zones sprayed on as narrow bands ensure the
highest resolution attainable with any given thin-layer chromatographic system. For
qualitative and quantitative HPTLC as well as for demanding preparative separation
spray-on application as bands is a necessity.
Key features:
Operation in standalone mode or under WINCATS.
Sample application as narrow bands using the spray-on technique.
Application of solutions onto any planar medium.
Semi-automatic operation, only changing of sample (cleaning, filling and
replacing the syringe) is performed manually.
Automatic sample application is a key factor for productivity of the HPTLC
laboratory. The requirements for an instrument serving this purpose, i.e. precision,
robustness during routine use and convenient handling are fully met by the Automatic
TLC Sampler 4. The LINOMAT offers fully automatic sample application for
qualitative and quantitative analyses as well as for preparative separations. it is suited
for routine use and high sample throughput in mass analysis. Samples are either
applied as spots through contact transfer (0.1-5 L) or as bands or rectangles (0.5 to
>50L) using the spray-on technique. Starting zones sprayed on as narrow bands offer
the best separation attainable with a given chromatographic system. Application in the
form of rectangles allows precise application of large volumes without damaging the
layer. Prior to chromatography, these rectangles are focused into narrow bands with a
solvent of high elution strength.
Development of chromatogram:
Thin-layer chromatography differs from all other chromatographic techniques in the
fact that in addition to stationary and mobile phases, a gas phase is present. This gas
phase can significantly influence the result of the separation.
Processes in the Developing Chamber
The classical way of developing a chromatogram is to place the plate in a chamber,
which contains a sufficient amount of developing solvent.
The lower end of the plate should be immersed several millimeters. Driven by
capillary action the developing solvent moves up the layer until the desired running
Ganpat University

19

Chapter 1

Introduction

distance is reached and chromatography is stopped. The following considerations


primarily concern silica gel as stationary phase and developments, which can be
described as adsorption chromatography[32-33].
1.3.3 Basics of separation
Most chromatographers have some idea of how a change in experimental conditions
will affect an HPLC chromatogram. In reversed phase separations an increase
organic phase in the mobile phase will shorten run time but usually lead to increased
band overlap. Decrease in flow rate increase run time but usually improves separation.
Sometimes changing the column will improve separation. This awareness of how
conditions affect the chromatogram is a combination of training and experience.
1.3.3.1 Resolution: General consideration
Chromatographers measure the quality of separations by resolution (Rs) of adjacent
bands. More is the Rs better is separation.

Figure 1.3.3.1: Calculation of resolution for two adjacent bands 1 and 2


Rs=2(t2-t1)
w1+w2
Here t1 and t2 are the retention times of the first and second bands and W 1 and W2 are
their baseline bandwidth. The resolution of two adjacent band with Rs=1 illustrated in
(Fig.1.3.3.1) Resolution Rs is equal to the distance between the peak centers divided
by the average bandwidth. To increase resolution, either the two bands must be
moved further apart, or bandwidth must be reduced.
1.3.3.1.1 Measurement of resolution
Resolution can be estimated or measured in three different ways:
1.3.3.1.1.1 Calculation based on equation
Equation 1.3.1 can be used for the measurement of resolution whenever the bands are
well separated, so that retention times and bandwidths can be determined reliably. The
manual determination of baseline bandwidth W involves the construction of tangents
Ganpat University

20

Chapter 1

Introduction

to each side of the band, and the measurement of the distance between the
intersections of these tangents with the baseline. Calculation of Rs using this method
may not be reliable when Rs is less than one.
1.3.3.1.1.2. Comparison with standard resolution curves
A comparison of two adjacent bands with standard resolution curves can also be used
to determine value of Rs. This approach does not require any calculations, is quite
convenient, and is applicable to overlapping band. Ideal representation of two
overlapping bands can be calculated as a function of relative band size (height or area)
and resolution. Actual overlapping bands can be compared with the ideal curves to
match real and ideal as closely as possible. It does not matter whether the larger
band elutes first or last; just mentally transpose the peaks. Once a match has been
achieved, the Rs value for closest match is then the resolution of the real band pair.
1.3.3.1.1.3 Calculation based on the valley between the two bands
Third way of estimating Rs, based on the height of the valley between two adjacent
bands, can be used for 0.8<Rs <1.5. The procedure provides more precise value of Rs
but require slightly more effort than the standard resolution curve approach. This
valley height hv is expressed as a percentage of the height of the smaller of the two
bands. Assumes band area calculated from perpendicular drop through the valley
divides the area of the two bands for integration. (Fig. 1.3.3.2)

Figure 1.3.3.2: Measuring the relative valley height for two overlapping bands
1.3.3.2 Minimum resolution
A common objective in HPLC separation is to separate all bands of interest with some
minimum resolution for accurate Quantitation of sample components. Baseline
resolution occurs when the detector trace for the first band returns to the baseline
before the next band begins to leave the column. With baseline resolution of all bands,
the HPLC data system is able to draw an accurate baseline under each band, thereby
increasing the accuracy of band area or peak-height measurements.

Ganpat University

21

Chapter 1

Introduction

It is convenient to define the critical band pair in each chromatogram obtained during
method development. The critical pair is that band pair with the smallest value of Rs.
In method development the separation conditions are changed systematically to
improve separation of the critical band pair. This process continues until acceptable
resolution of the entire sample is obtained.
1.3.3.3 Effect of solvent strength
Sample retention can be controlled by varying the solvent strength of the mobile
phase. A strong solvent decreases retention and weak solvent increases retention.
Table 2.1 summarizes the primary mean for varying solvent strength with different
HPLC method[34,35].
Table 1.3.1 Controlling Sample Retention by Changing Solvent Strength
HPLC method

How solvent strength is usually varied

Reversed Phase

Water (A) plus organic solvent (B) (eg. Water-ACN); increase in


%B decreases K

Normal phase

Nonpolar organic solvent (A) plus polar organic solvent (B) (e.g.
hexane-propanol); increases in % B decreases K.

Ion Pair

Same as reverse phase.

Ion exchange

Buffered aqueous solution plus added salt (e.g. 5mM sodium


acetate plus 50 mM NaC1); increases in

(C) ionic strength

(NaCl concentration) decreases K


1.3.3.4 Effect of selectivity
The next step in method development (after adjusting % B for 0.5<K<20) is achange
of conditions that will vary band spacing or selectivity (), changes in can be
created by a change in the mobile phase, a change in the type of column packing, or a
change in temperature. Usually, it is best to start with changes in the mobile phases.
1.3.3.4.1 Change in the mobile phase
1.3.3.4.1.1 Solvent -type selectivity
A change in organic solvent is a powerful way to change band spacing for both
reversed and normal phase HPLC. Usually, it is the stronger solvent component (B
solvent) that will be changed for this purpose. There are many solvents to choose
from, which complicates the selection of preferred solvents for this purpose. The
solvent selectivity triangle in figure-1.3.3.3 is a useful guide for choosing among
different solvents for the purpose of a large change in band spacing solvents are
attracted to sample molecules in the mobile phase by a combination of dipole and
Ganpat University

22

Chapter 1

Introduction

hydrogen bonding interactions. As a result, solvent is expected to depend on the


dipole moment, acidity, and basicity of the solvent molecule.

Figure 1.3.3.3: Solvent selectivity triangle for composition of mobile phase


1.3.3.4.1.2 Optimizing solvent-Type Selectivity
A change of the strong solvent (B-solvent) often result in large changes in band
spacing, such that bands that were formerly overlapped are now resolved and bands
that were formerly resolved are now overlapped. As a result, a mixture of the two
strong solvents often provides intermediate band spacing and acceptable resolution. In
first experiment designed to adjust solvent strength and the range of K value. It is
advisable to start with a relatively strong mobile phase. The sample is weakly retained
and leaves the column quickly with poor resolution of the sample.
1.3.2.4.1.3 Selectivity for ionic compound
For ionic samples that contain ionized or ionizable components further changes in
mobile phase are possible as a means of varying selectivity: change of pH, use of ion
pairing reagents or amine additives, change of buffer or buffer concentration.
1.3.2.4.1.4 Selective Complexation
In rare cases it may be possible to add a complexing agent to the mobile phase that
interacts selectively with one or more sample components. If complexing agent is
used, the equilibrium between the sample compound and complexing agent must be
rapidly reversible; otherwise, broad bands and poor chromatography are likely to
result.
1.3.2.4.2 Changes in the column
The nature of the column packing can have a major effect on band spacing. In most
cases it is not practical to combine different packing into a single column, although
columns of different type have been connected in series. Therefore, a change in the
Ganpat University

23

Chapter 1

Introduction

column necessarily involves an abrupt change in selectivity, as opposed to the


continuous change in selectivity that is possible by changing mobile phase
composition. This limits the ability of the column to fine-tune band spacing for
samples that contain a relatively large number of components. For this reason a
change in the column usually should be combined with changes in the mobile phase to
optimize band spacing.
Most HPLC column packing is made by bonding an organic layer on to the internal
surface of porous silica particles.
The resulting column packing can exhibit differences in selectivity as results of a
number of factors are as follows:I. Chemical nature or functionality of the bonded phase (e.g. C18, Phenyl or
cyano)
II. Amount of bonded phase permit surface of the silica particle.
III. Way in which the bonded phase is attached to the silica surface (e.g.
monofunctional Vs polyfunctional)
IV. Nature of the silica surface, which caries among different silica sources.
1.3.2.4.3 Changes in temperature
An increase in column temperature by 1oC will usually decrease retention (K) by 1 to
2%. A change in K can also result in change in , so temperature is a potentially
useful parameter for changing band spacing and improving resolution. One advantage
of using temperature for optimizing selectivity is convenience. No change in the
column is required, nor is it necessary to make up a new mobile phase; however, for a
large increase in temperature it may be necessary to reduce % B to maintain
0.5 < K < 20.
Temperature has not been widely used for controlling band spacing, because of
certain consideration:
1.

HPLC equipment is often not equipped with a column thermostat.

2.

HPLC columns are not stable at higher temperature particularly for a


mobile phase pH below 3 or above 6.

3.

Solvent viscosity and vapour pressure depend strongly on temperature,


which restricts the practical range in which temperature can be varied.

4.

it has been assumed that a change in temperature is usually less effective


for changing value of .

Ganpat University

24

Chapter 1

Introduction

These considerations have been undergoing a reexamination and it is expected that in


the future temperature will be used increasingly for the purpose of controlling bond
spacing and facilitating HPLC method development[36-41].

1.4 Development of new analytical method


A regulatory analytical procedure is used to evaluate a defined characteristic of the
drug substance or drug product. An alternative analytical procedure is proposed by
the applicant for use other than regulatory analytical procedure. A stability-indicating
assay is a validated quantitative analytical procedure that can detect the changes with
time in the pertinent properties of the drug substance and drug product. A stability
indicating assay accurately measures the active ingredients, without interference from
degradation products, process impurities, excipients, or other potential impurities [42].
Stability testing forms an important part of the process of drug product development.
The purpose of stability testing is to provide evidence on how the quality of a drug
substance or drug product varies with time under the influence of a variety of
environmental factors such as temperature, humidity and light, and enables
recommendation of storage conditions, retest periods and shelf lives to be established.
The two main aspects of drug product that play an important role in shelf life
determination are assay of active drug, and degradants generated, during the stability
study. The assay of drug product in stability test sample needs to be determined using
stability indicating method, as recommended by the International Conference on
Harmonization(ICH) guidelines[43] and USP 26[44].
The modern methods of choice for quantitative analysis are HPLC, GLC, and
HPTLC, which are highly sophisticated. Chromatographic methods are commonly
used in regulatory laboratories for the qualitative and quantitative analysis of drug
substance, drug products, raw materials and biological samples throughout all phases
of drug development, from research to quality control. High performance liquid
chromatography (HPLC) is the fastest growing analytical technique for the analysis of
drugs. Its simplicity, high specificity and wide range of sensitivity make it ideal for
the analysis of many drugs in both dosage forms and biological fluids. The rapid
growth of HPLC has been facilitated by the development of reliable, moderately
priced instrumentation and efficient columns[45]. High performance thin layer
chromatography (HPTLC) is a classical separative technique that has employed wide
Ganpat University

25

Chapter 1

Introduction

spread popularity in the analysis of complex mixtures of natural origin. Now a days
HPTLC is becoming a routine analytical technique due to its advantages of low
operating cost, high sample put, and need for minimum sample clean-up. The major
advantage of HPTLC is that several samples can be run simultaneously using small
quantity of mobile phase unlike HPLC, thus lowering analysis time and cost per
analysis[46].
1.5 Validation of analytical methods[47-53]
As defined by the USP, method validation provides an assurance of reliability during
normal use, and is sometime referred to as the proces of providing documented
evidence that the method does what it is intended to do The objective of validation of
an analytical method is to demonstrate that the procedure, when correctly applied,
produces results that are fit for purpose. To be fit for the intended purpose, the method
must meet certain validation characteristics. Typical validation characteristics, which
should be considered, are: selectivity, specificity, linearity, range, accuracy, precision,
limit of detection, limit of quantification, ruggedness, robustness and system
suitability testing.
1.5.1 Specificity
Specificity is the ability to assess unequevalently the analyte in the presence of
components, which may be expected to be present. Typically it might be include
impurities, degradants, etc. Specificity investigation should be conducted during the
validation of identification tests, the determination of impurities and the assay. The
procedures used to demonstrate Specificity will depend on the intended objective of
the analytical procedure. It is not always possible to demonstrate that an analytical
procedure as specific for a particular analyte. In this case a combination of two or
more analytical procedures is recommended to achieve the necessary level of
discrimination.
Suitable identification test should be able to discriminate between compounds of
closely related structures which are likely to be present. The discrimination of a
procedure may be confirmed by obtaining positive results (perhaps by comparison
with a known reference material) from samples containing the analyte, coupled with
negative results from sample which does not contain the analyte. The choice of such
potentially interfering materials should be based on sound scientific Judgment with a
consideration of the interferences that could occur.

Ganpat University

26

Chapter 1

Introduction

Assay and impurity test(s): For chromatographic procedure, representative


chromatograms should be used to demonstrate specificity and individual components
should be labeled. Critical separation in chromatography should be investigated at an
appropriately level for critical separation Specificity can be demonstrated by the
resolution of t he two components which elute closest to each other. In case where a
non-specific assay is used, other supporting analytical procedure should be used to
demonstrate overall Specificity. For example, where a titration is adopted to assay the
drug substance for release, the combination of the assay and a suitable test for
impurities can be used. The approach is similar for both assay and impurity tests:
Impurities are available: For the assay, this should involve demonstration of the
discrimination of the analyte in the presence of impurities and/or excipients;
practically, this can be done by spiking pure substance (drug substance or drug
product) with appropriate levels of impurities and /or excipients and demonstrating
that the assay result is unaffected by the presence of these materials (by comparison
with the assay result obtained on unspiked samples). For the impurity test the
discrimination may be established by spiking drug substance or drug product with
appropriate levels of impurities and demonstrating the separation of these impurities
individually and/or from other components in the sample matrix. For the impurity test
the discrimination may be established by spiking drug substance or drug product with
appropriate levels of impurities and demonstrating the separation of these impurities
individually and/or from other components in the sample matrix. Impurities are not
available: If impurity or degradation product, standards are unavailable, specificity
may be demonstrated by comparing the test. Results of samples containing impurities
or degradation products to a second well-characterized procedure e.g. pharmacopoeial
appropriate, this should include samples stored under relevant stress conditions: light,
heat, humidity, acid/base hydrolysis and oxidation.
For the assay, the two results should be compared. For the impurity tests the impurity
profiles should be compared. Peak purity tests may be useful to show that the analyte
chromatographic peak is not attributable to more than one component (e.g. diode
array, mass spectrometry).

Ganpat University

27

Chapter 1

Introduction

1.5.2 Accuracy
The accuracy of an analytical procedure expresses the closeness of agreement
between the value which is accepted either as a conventional true value or an accepted
reference value and the value found. This is sometimes termed trueness. Accuracy
should be established across the specified range of the analytical procedure. Drug
substance: Several methods of determining accuracy are available: a) Application of
an analytical procedure to an analyte of known purity (e.g. reference material) b)
Comparison of the results of the proposed analytical procedure with those of a second
well-characterized procedure, the accuracy of which state and/or defined; c) Accuracy
may be inferred once precision, linearity and specificity have been established.
Drug product: Several methods for determining accuracy are available: a) Application
of the analytical procedure to synthetic mixture of the drug products components to
which known quantities of the drug substance to be analyzed have been added. b) In
cases where it is impossible to obtain samples of all drug product components, it may
be acceptable either to add known quantities of the drug product or to compare the
result obtained from a second, well characterized procedure, the accuracy of which is
stated and/or defined. c) Accuracy may be inferred once precision, linearity and
specificity have been established. Impurities (Quantitation): Accuracy should be
assessed on samples (drug substance/drug product) spiked with known amounts of
impurities. In cases where it is impossible to obtain samples of certain impurities
and/or degradation products, it is considered acceptable to compare results obtained
by an independent procedure. The response factor of the drug substance can be used.
It should be clear how the individual or total impurities are to be determined e.g.
weight/weight or area percent, in all cases with respect to the major analyte.
Recommended data: Accuracy should be assessed using a minimum of 9
determinations over a minimum of 3 concentration levels covering the specified range
(e.g.3 concentrations/3 replicates each of the total analytical procedure). Accuracy
should be reported as percent recovery by the assay of known added amount of
analyte in the sample or as the difference between the mean and the accepted true
value together with the confidence intervals.
1.5.3 Precision
The precision of an analytical procedure expresses the closeness of agreement (degree
of scatter) between a series of measurements obtained from multiple sampling of the
same homogeneous sample under the prescribed conditions. Precision may be
Ganpat University

28

Chapter 1

Introduction

considered at three levels: repeatability, intermediate precision and reproducibility.


Precision should be investigated using homogeneous, authentic samples. However, if
it is not possible to obtain a homogeneous sample it may be investigated using
artificially prepared samples or a sample solution. The precision of an analytical
procedure is usually expressed as the variance, standard deviation or coefficient of
variation of a series of measurements. Repeatability: Repeatability expresses the
precision under the same operating conditions over a short interval of time.
Repeatability is also termed intra-assay precision. Repeatability should be assessed
using: a) A minimum of 9 determinations covering the specified range for the
procedure

(e.g.3concentrations/3replicates

each)

or

b)

minimum

of

determinations at 100% of the test concentration.


Intermediate

precision:

Intermediate

precision

expresses

within-laboratories

variations: different, days, different analyst, different equipment, etc. the extent to
which intermediate precision should be established depends on the circumstances
under which the procedure is intended to be used. The applicant should establish the
effects of random events on the precision of the analytical procedure. Typical
variations to be studied include days, analysts, equipment, etc. it is not considered
necessary to study these effects individually. The use of an experimental design
(matrix) is encouraged.
Reproducibility: is assessed by means of an inter laboratory trial. Reproducibility
should be considered in case of the standardization of an analytical procedure, for
instance, for inclusion of procedures in pharmacopoeias, these data are not part of the
marketing authorization dossier. Reproducibility expresses the precision between
laboratories

(collaborative

studies,

usually

applied

to

standardization

of

methodology). Validation of tests for assay and for quantitative determination of


impurities includes an investigation of precision. Recommended data: The standard
deviation relative standard deviation (coefficient of variation) and confidence interval
should be reported for each type of precision investigated.
1.5.4 Detection limit
The detection limit of an individual analytical procedure is the lowest amount of
analyte in a sample which can be detected but not necessarily quantitated as an exact
value. Several approaches for determining the detection limit are possible depending
on whether the procedure is a non-instrumental or instrumental, approaches other than
those listed below may e acceptable.
Ganpat University

29

Chapter 1

Introduction

Based on visual evaluation: Visual evaluation may be used for non instrumental
methods but may also be used with instrumental methods. The detection limit is
determined by the analysis of samples with known concentrations of analyte and by
establishing the minimum level at which analyte can be reliably detected.
Based on signal-to-noise: This approach can only be applied to analytical procedures
which exhibit baseline noise. Determination of the signal-to-noise ratio is performed
by comparing measured signals from samples with known low concentrations of
analyte with those of blank samples and establishing the minimum concentration at
which the analyte can be reliable detected. A signal-to-noise ratio between 3 or 2:1 or
generally considered acceptable for estimating the detection limit.
Based on the standard deviation of the response and the slope: The detection limit
(DL) may be expressed as: DL =3.3 / S
Where = the standard deviation of the response
S = the slope of the calibration curve The slope S may be estimated from the
calibration curve of the analyte. The estimate of may be carried out in a variety of
ways, for example; (A) Based on the standard deviation of the blank: Measurement of
the magnitude of analytical background response is performed by analyzing an
appropriate number of blank samples and calculating the standard deviation of these
responses. (B) Based on the calibration curve: A specific calibration curve should be
studied using samples containing an analyte in the range of DL. The residual standard
deviation of a regression line or the standard deviation of y-intercepts of regression
lines may be used as the standard deviation.
Recommended data: The detection limit and the method used for determining the
detection limit should be presented. If DL is determined based on visual evaluation or
based on signal to noise ratio, the presentation of the relevant chromatograms i s
considered acceptable for justification. In cases where an estimated value for the
detection limit is obtained by calculation or extrapolation, this estimate may
subsequently be the independent analysis of a suitable number of samples known to
be near or prepared at the detection limit.
1.5.5 Quantitation limit
The quantitation limit of an individual analytical procedure is the lowest amount of
analyte in a sample which can be quantitatively determined with suitable precision
and accuracy. The quantitation limit is a parameter of quantitative assays for low
levels of compound in sample matrices, and is used particularly for the determination
Ganpat University

30

Chapter 1

Introduction

of impurities and/or degradation products. Several approaches for determining the


quantitation limit are possible, depending on whether the procedure is a noninstrumental or instrumental. Approaches other than those listed below may be
acceptable.
Based on visual evaluation: Visual evaluation may be used for non-instrumental
methods but may also be used with instrumental methods. The quantitation limit is
generally determined by the analysis of samples with known concentrations of analyte
and by establishing the minimum level at which the analyte can be quantified with
acceptable accuracy and precision.
Based on signal-to-noise approach: This approach can only be applied to analytical
procedures that exhibit baseline noise. Determination of the signal-to-noise ratio is
performed by comparing measured signals from samples with known low
concentrations of analyte with th ose of blank samples and by establishing the
minimum concentration at which the analyte can be reliably quantified. A typical
signal-to-noise ratio is 10:1
Based on the standard deviation of the response and the slope: The quantitation
limit (QL) may be expressed as: DL =10 /S
Where = the standard deviation of the response
S = slope of the calibration curve
The slope S may be estimated from the calibration curve of the analyte. The estimate
of may be carried out in a variety of ways for example:
Based on Standard Deviation of the Blank: Measurement of the magnitude of
analytical background response is performed by analyzing an appropriate number of
blank samples and calculating the standard deviation of these responses.
Based on the calibration curve: A specific calibration curve should be studied using
samples, containing an analyte in the range of QL. The residual standard deviation of
a regression line or the standard deviation of y-intercepts of regression lines may be
used as the standard deviation.
Recommended data: The quantitation limit and the method used for determining the
quantitation limit should be presented. The limit should be subsequently validated by
the analysis of a suitable number of samples known to be near of prepared at the
quantitation limit.

Ganpat University

31

Chapter 1

Introduction

1.5.6 Linearity
The linearity of an analytical procedure is its ability (within a given range) to obtain
test results which are directly proportional to concentration (amount) of analyte in the
sample. A linear relationship should be evaluated across the range of the analytical
procedure. It may be demonstrated directly on the drug substance (by dilution of a
standard stock solution) and/or separate weighing of synthetic mixtures of the drug
product components, using the proposed procedure. The latter aspect can be studied
during investigation of the range. Linearity should be evaluated by visual inspection
of a plot of signals as a function of analyte concentration or content. If there is a linear
relationship, test results should be evaluated by appropriate statistical methods, for
example, by calculation of a regression lime by the method of least squares. In some
cases to obtain linearity between assays and sample concentration, the data may need
to be subjected to a mathematic al transformation prior to the regression analysis.
Data from the regression lie itself may be helpful to provide mathematical estimates
of the degree of linearity. The correlation coefficient, y-intercept, slope of the
regression line and residual sum of square should be submitted. A plot of the data
should be included. In addition, an analysis of the deviation of the actual data points
from the regression line may also be helpful for evaluating linearity. Some analytical
procedures, such as immunoassays, do not demonstrate linearity after any
transformation. In this case, the analytical response should be described by an
appropriate function of the concentration (amount) of an analyte in a sample. For the
establishment of linearity, a minimum of 5 concentrations is recommended.
1.5.7 Range
The range of an analytical procedure is the interval between the upper and lower
concentration (amounts) of analyte in the sample (including these concentrations) for
which it has been demonstrated that the analytical procedure has a suitable level of
precision, accuracy and linearity. The specified range is normally derived from
linearity studies and depends on the intended application of the procedure. It is
established by confirming that the analytical procedure provides an acceptable degree
of linearity, accuracy and precision when applied to samples containing amounts of
analyte within or at the extremes of the specified range of the analytical procedure.
The following minimum specified range should be considered. a) For the assay of a
drug substance or a finished (drug) product; normally from 80 to 120 percent of the
test concentration. b) For content uniformity, covering minimum of 70 to 130 percent
Ganpat University

32

Chapter 1

Introduction

of the test concentration unless a wider more appropriate range. Based on the nature
of the dosage form (e.g. metered dose inhalers), is justified. C) For dissolution
testing:+20% over the specified range.
For the determination of an impurity: from the reporting level of an impurity to 120%
of the specification. If assay and purity are performed together as one test and only a
100% standard is used, linearity should cover the range from the reporting level of the
impurities to 120% of the assay specification.
1.5.8 Robustness
The robustness of an analytical procedure is a measure of its capacity to remain
unaffected by small, but deliberate variation in method parameters and provides an
indication of its reliability during normal usage. The evaluation of robustness should
be considered during the development phase and depends on the type of procedure
under study. It should show the reliability of an analysis with respect to deliberate
variation in method parameters. If measurements are susceptible to variations in
analytical condition the analytical conditions should be suitably controlled or a
precautionary statement should be included in the procedure. One consequence of the
evaluation of robustness should be that a series of system suitability parameters (e.g.
resolution test) is established to ensure that the validity of the analytical procedure is
maintained whenever used. Examples of typical variations are:
a) Stability of analytical solutions. b)Extraction time. In the case of liquid
chromatography, examples of typical variation are: a) Influence of variations of pH in
a mobile phase. b) Influence of variations in mobile phase composition. c) Different
columns (different lots and/or suppliers). d) Temperature. e) Flow rate.
1.5.9 Ruggedness
Method ruggedness is defined as the reproducibility of results when the method is
performed under actual condition. This includes different analysis laboratories,
instruments source of reagents, chemicals, solvents and so on. The strategies for
determining method ruggedness will very delivery on the type and complexity of the
method and the time available for validation. Often, the real ruggedness of a method
can only be determined over time by experience in different laboratories.
1.5.10 System suitability testing
System suitability testing is an integral part of many analytical procedures. The tests
are based on the concept that the equipment, electronics, analytical operations and
samples to be analyzed constitute an integral system that can be evaluated as such.
Ganpat University

33

Chapter 1

Introduction

System suitability test parameters to be established for a particular procedure depend


on the type of procedure being validated. The relationship between validation and
other phase of the method evaluation/ utilization process is often unclear. The
hierarchical concept of method utilization wherein the stage of method development
validation and utilization are distinct and sequential represents a situation which is
practically undesirable and potentially inefficient. After the method has been
conceived and operationally developed its performance is validated with respect to
several different performance parameters. Successful completion of the validation
results in a method which can reliable be used to characterize real samples. The
objective of the analytical procedure should be clearly understood since this will
govern the validation characteristics, which need to be evaluated. International
conference on harmonization recently published its own list of important validation
characterization for various procedures.

1.6 Stability indicating method


1.6.1 Introduction
The stability indicating assay is a method that is employed for the analysis of stability
samples in pharmaceutical industry. With the advent of International Conference on
Harmonisation (ICH) guidelines, the requirement of establishment of stability
indicating assay method (SIAM) has become more clearly mandated. The guidelines
explicitly require conduct of forced decomposition studies under a variety of
conditions, like pH, light, oxidation, dry heat, etc. and separation of drug form
degradation products. The method is expected to allow analysis of individual
degradation products[54]. For the development of a sound scientific protocol for the
stability studies, an understanding of the conditions under which a drug degrades as
well as the mechanism of the breakdown is needed. This is established through a
series of stress studies designed to elucidate the intrinsic stability of the new molecule
by establishing its degradation pathway[55]. A stress stability study is often referred to
as a pre-formulation study or characterization study. The result of these studies are
the basis for developing appropriate dosage forms, formulations and manufacturing
processes, selecting appropriate packaging and storage conditions and the analytical
methods to be used in stability studies. The information derived from stress testing
can be used to establish the methodology employed, the parameters followed and
specifications for long-term testing under accelerated and normal storage condition
Ganpat University

34

Chapter 1

Introduction

conditions[56]. Stress studies are different from accelerated studies because the former
is carried under more sever conditions. Stress testing is testing under extreme
conditions, used to characterize only the drug substance. Accelerated testing applies
both to the drug substance and drug product and involves testing under conditions
more severe than normal, which serve to generate data useful in predicting what might
happen during storage under normal conditions[57].
1.6.2 Practical conduct of stress testing
The ICH guideline Q1A suggests the following conditions to be employed: (i) 10C
increments above the accelerated temperatures (e.g. 50C, 60C, etc.), (ii) humidity
where appropriate (e.g.75% or greater), (iii) hydrolysis across a wide range of pH
values, (iv) oxidation and (v) photolysis. However, the guideline provides no details
on how hydrolytic, photolytic and oxidative studies have to be actually performed. In
other words, the practical aspects concerning the conduct of stress testing are
addressed neither by the regulatory guidelines nor by any other document, leaving the
performance of these studies to the prudence of the applicant. On the other hand, the
information is available in literature but in staggered way, with suggested approaches
differing a lot from one another. Few approaches towards the practical conduct of
stress studies are reported in literature[58]. A comprehensive document providing
guidance on the practical conduct and issues related to stress testing under variety of
ICH prescribed conditions has been published. This report from the authors proposes
a classification scheme and offers decision trees to help in the selection of the right
type of stress condition in a minimum number of attempts. This guidance document
on the conduct of stress tests to determine inherent stability of drugs will be followed
in the current study From the guidance on the conduct of stress tests to determine
inherent stability of drugs, the following observations were made clearly: the
condition used to study decomposition in acid revealed that 0.1N hydrochloric acid
was most commonly used. A few reports indicate the use of 1N HCl and even higher
strength and the use of sulfuric acid in varying strengths. Large variations were also
seen in the reaction (temperature) conditions and periods of study. The temperatures
varied between 40C and 110C. The reaction time varied, for example, drugs being
kept at 100C or at boiling conditions, for periods ranging from a few minutes to as
long as 2 months. The extent of decompositions also varied. For example, a 35% loss
of retinoic acid was observed on refluxing in 0.1N HCl for just 5min, whereas no drug
decomposition was reported after refluxing nabilone in 0.1N acid for a week.It is
Ganpat University

35

Chapter 1

Introduction

observed that NaOH is most often used for the hydrolysis of drugs in alkaline
conditions, at strengths of 0.1N and 1N, with the occasional use of potassium
hydroxide. As with acidic degradation, great variation is observed in the time and
temperature of alkali exposure. Depending on their inherent stability, some drugs (for
example, nabilone) show no degradation even after refluxing in 0.1N NaOH for a
week, whereas, others (such as trifluoperazine) undergo complete degradation in 0.1N
alkali for 24 h at 30C[59]. At neutral pH, no significant degradation was obtained
when the temperature was 37C for celiprolol, and refluxing conditions were used for
sertraline. The testing is generally done in water. The slow rate of decomposition in
neutral conditions is understandable because reactions at neutral pH are non-catalytic
and hence long periods at exaggerated temperatures may be required to obtain
sufficient quantities of degradation products. The most commonly used oxidizing
agent, hydrogen peroxide, is used in varying strengths between 1% and 30%. Some
drugs (ranitidine HCl and cimetidine HCl) degrade when exposed to 3 %H2O2 for
short periods at room temperature (RT)[59]. In other cases, exposure to high
concentration of H2O2, even at extreme conditions, does not cause any significant
degradation (for example sertraline HCl). This could happen since non-oxidizable
drugs are not expected to show any change-even in the presence of high concentration
of oxidizing agents. Photolytic studies are done on drugs in either solid form or
solution, in water or in acidic and alkaline solutions and also on drugs dissolved in
either methanol or acetonitrile. Mostly drugs are exposed to short /long wavelength
UV or fluorescent light of varying illumination (approximately 4300-17000 lux). The
Various degradation conditions described for the stressed testing are shown in Table
1.6.1.

Ganpat University

36

Chapter 1

Introduction

Table1.6.1 Various degradation conditions described for the stress testing


Decompositi

Recently used

Temperatu

on condition

most

re

Reaction time

Extent of
degradation

frequently
Acidic

Neutral

HCl
(0.1N/1N/5N)
H2SO4
NaOH
(0.1N/1N5N)
Water

Oxidation

H2O2(1%-3%)

Alkaline

Photolytic

UV/Fluorescent
light (4300 to
17000 lux)
RT= room temperature

40C-110C

Few min-2
months

Negligiblecomplete

80C/Reflux

5 min-21days

85C/Reflux

Very long
periods
Few hoursweek
Few hoursmonths

InsignificantExtensive
No significant
degradation
InsignificantExtensive
Nil-Complete

RTRefluxing
RT

Flow charts or decision trees (Figures 1.6.1-1.6.4) for investigating the different types
of stress conditions for new drug substances are shown which assume that the new
drug is labile in nature to the stress conditions. Depending on the results, the strength
of the reaction condition may be increased or decreased. The change, if required, is
done stepwise and stress conditions are accepted when sufficient decomposition is
obtained. The term sufficient decomposition is taken in the broadest sense, meaning
80%-100% decomposition if the objective is isolation of the degradation products, or
between 20-80% decompositions when the objective is to establish degradation
pathways.

Ganpat University

37

Chapter 1

Introduction

START

1.2 x106 Lux hr


No Degradation
6.0 x106 Lux hr

Total
Degradation

Sufficient
Degradation

Reduce exposure

No Degradation
Sufficient
Degradation

Declare drug to be
practically stable

ACCEPT

Figure 1.6.1 Flow chart for performing stress studies for photolytic degradation

Ganpat University

38

Chapter 1

Introduction

START

No Degradation

0.1N HCl/ NaOH,


8 h, Reflux

Total Degradation

Sufficient Degradation

1 N HCl /NaOH,
12 h, Reflux

0.01HCl/ NaOH,
8 h, at 40c

Sufficient Degradation

Sufficient Degradation

No Degradation

2 N HCl/ NaOH,
24 h, Reflux

Sufficient
Degradation

Total Degradation

ACCEPT

No Degradation

5 N HC l/ NaOH,
24 h, Reflux

Sufficient Degradation

Sufficient
Degradation

1N HCl/ NaOH,
2 h at, 25C

Total Degradation

Carry out studies


under milder
conditions

No Degradation

Declare drug to
be practically
stable
Figure 1.6.2: Flow chart for performing stress studies for hydrolytic degradation
under acid and alkali conditions

Ganpat University

39

Chapter 1

Introduction

START

No Degradation

Water/ 12 h,
Reflux

Total Degradation

Sufficient Degradation

Water/ 1 day,
Reflux

Sufficient Degradation

No Degradation

Water/ 2 day,
Reflux

Sufficient
D degradation

Water/ 8 hr, at
40C

Total Degradation

ACCEPT

No Degradation

Water/ 5 day
Reflux

Sufficient Degradation

Sufficient Degradation

Sufficient
Degradation

Water/ 2 hr, at
25C

Total Degradation

Carry out studies


under milder
conditions

No Degradation

Declare drug to
be practically
stable
Figure 1.6.3: Flow chart for performing stress studies for hydrolytic degradation
under neutral condition (in water)

Ganpat University

40

Chapter 1

Introduction

START

No Degradation

3% H2O2/ 24 h,
RT

3% H2O2/ 6 h,
RT

Sufficient
Degradation

Sufficient
Degradation

No Degradation

10% H2O2/ 24
h, RT

Sufficient
Degradation

1% H2O2/ 3 h,
RT

Total Degradation

ACCEPT

No degradation
Sufficient Degradation

30% H2O2/ 24 h,
RT

Total Degradation

Sufficient
Degradation

1% H2O2/ 30 min,
RT

Total Degradation

Carry out studies


under milder
conditions

Declare drug to
be practically
stable
Figure 1.6.4: Flow chart for performing stress studies for degradation under oxidative
conditions.

Ganpat University

41

Chapter 1

Introduction

1.7 References
1. Tripathi KD, Essentials of Medical Phamacology, 6th Ed., Jaypee Brothers
Medical Publishers, 2008; 339-50.
2. Rang HP, Dale MM, Ritter JM, Flower RJ, Rang and Dales Pharmacology,
6th Ed., Elsevier, 2005: 385-386.
3. Bras H, Jankowska E, Noga B, Skoog B, Comparison of Effects of Various
Types of NA and 5-HT Agonists on Transmission from Group II Muscle
Afferents in the Cat, European Journal of Neuroscience, 1990; 2(12): 10291039.
4. Chou R, Qaseem A, Snow V et al., Diagnosis and treatment of low back pain:
a joint clinical practice guideline from the American College of Physicians
and the American Pain Society, Ann. Intern. Med., 2007; 147 (7): 478-91.
5. Frye RF and Stiff J, Analysis of Chlorzoxazone in plasma and urine by high
performance liquid chromatography, J. of Chromatogr. B: Biomed. Sci.
Appl.,1996; 68(2): 291-296
6. Dong DL, Luan Y, Feng TM, Fan CL, Yue P, Sun ZJ, Gu RM, Yang BF,
Chlorzoxazone inhibits contraction of rat thoracic aorta., Eur J Pharmacol.,
2006; 545(2): 161-6.
7. Sweetman SC, Martindale: The Complete Drug Reference, 37th Ed. London:
Pharmaceutical Press, 2011; 1392-93.
8. Wan J, Ernstgard L, Song BJ, Shoaf SE, Chlorzoxazone metabolism is
increased in fasted Sprague-Dawley rats., J Pharm Pharmacol., 2006; 58(1):
51-61
9. Park JY, Kim KA, Park PW, Ha JM, Effect of high-dose aspirin on CYP2E1
activity in healthy subjects measured using Chlorzoxazone as a probe., J Clin
Pharmacol., 2006; 46(1): 109-114.
10. Forrest JA, Clements JA, Prescott LF, Clinical pharmacokinetics of
Paracetamol, Clin Pharmacokinet.,1982; 7: 93-107.
11. ONeil MJ, The Merck Index: An Encyclopedia of chemicals, drugs and
biological, 14th Ed., Merck Research Laboratories, 2006; 9.
12. Sweetman SC, Martindale: The Complete Drug Reference, 37th edition Ed.
London: Pharmaceutical Press, 2011; 76-79

Ganpat University

42

Chapter 1

Introduction

13. Levy G, Comparative pharmacokinetics of aspirin and acetaminophen, Arch


Intern Med. 1981; 141: 279-281.
14. Satoskar

RS,

Bhandarkar

SD,

Ainapure

SS,

Pharmacology

and

th

pharmacotheraputics, 6 Ed., Popular prakashan, 1999; 162-163.


15. Solomon DH, Avorn J, Sturmer T, Glynn RJ, Mogun H, Schneeweiss S,
Cardiovascular outcomes in new users of coxibs and nonsteroidal
antiinflammatory drugs: high-risk subgroups and time course of risk, Arthritis
Rheum., 2006; 54(5): 1378-89.
16. Gan TJ, Diclofenac: an update on its mechanism of action and safety profile.,
Curr Med Res Opin., 2010; 26(7): 1715-31.
17. FitzGerald GA, Patrono C, The coxibs, selective inhibitors of cyclooxygenase2 N Engl J Med. 2000; 345(6): 433-42.
18. Sweetman SC, Martindale: The Complete Drug Reference, 37th edition Ed.
London: Pharmaceutical Press, 2011; 32-3.
19. Satoskar

RS,

Bhandarkar

SD,

Ainapure

SS,

Pharmacology

and

th

pharmacotheraputics, 6 Ed., Popular prakashan, 1999; 164-166.


20. Goodman & Gilmans The Pharmacological Basis of Therapeutics, 11th Ed.,
Mcgraw-hill medical publishing division, 1634-1644.
21. Forist AA, Judy RW, Comparative pharmacokinetics of chlorphenesin,
carbamate and methocarbamol in man, Journal of Pharmaceutical Sciences,
1971; 60: 1686-1688.
22. Weng N, HPLC and CE Methods for Pharmaceutical Analysis, J. Chromatogr.
B Biomed. Appl., 1994; 654: 287-292.
23. ONeil MJ, The Merck Index: An Encyclopedia of chemicals, drugs and
biological, 14th Ed., Merck Research Laboratories, 2006; 1033.
24. Sica DA, Comstock TJ, Davis J, Manning L, Powell R, Melikian A, Wright G,
Pharmacokinetics and protein binding of methocarbamol in renal insufficiency
and normals. Eur J. Clin Pharmacol., 1990; 39(2): 193-94.
25. Roy D, Implementation of Revised Schedule M and SSI, The Indian
Pharmacist, Dec. 2003; 8-11.
26. Brown P, Deanotonis K, Settle F, High Performance Liquid Chromatography,
Handbook of Instrumental Techniques for Analytical Chemistry Prentice Hall,
Inc., A Simon and Schuster Company, New Jersey, 1997; 147-159.
27. United States pharmacopoeia, XXII, 2007; 1740.
Ganpat University

43

Chapter 1

Introduction

28. Mant CT, Hodges RS, High-Performance Liquid Chromatography of Peptides


and Proteins: Separations, Analysis, and Conformation, CRC Press: Boston,
1991; 158-163.
29. McClure WF, Process Analytical Chemistry & Spectroscopic tools, 2nd Ed.,
Wiley Publishers, 1994; 66: 43-54.
30. Brown PR, Weston A, Principle and practices of High Performance Liquid
Chromatography & Capillary Electrophoresis, 1st Ed., Academic Press, 1990;
62: 995-1008.
31. Davidson AG, Beckett AH, Stenlake JB, Chromatography In; Practical
Pharmaceutical Chemistry, 4th Ed., CBS Publishers and Distributors, New
Delhi, 1997; 2: 85-174.
32. Sethi PD, Charegaonkar D, Identification of drugs in Pharmaceutical
Formulations by Thin Layer Chromatography, 2nd Ed., CBS Publishers &
Distributers, 2008; 2-15.
33. Patel RB, Patel MR, Patel BG, Experimental Aspects and Implementation of
HPTLC, New York: Springer, 2011; 41- 54.
34. Willard HH, Merritt LL, Dean JA, Settle FA, HPLC Theory and
Instrumentation, Instrumental Methods of Analysis, 8th Ed., CBS Publishers
and Distributors, New Delhi, 2002; 581-617.
35. Miyabe K, Gniochon G, Brown, PR and Grushka E, Fundamental
Interpretation of the Peak in Linear Reversed Phase Liquid Chromatography,
Advances in Chromatography, Marcel Dekker, Inc., Newyork, 2000; 40: 1107.
36. Snyder LR, Kirkland JJ, Glajch LJ, Getting Started, Practical HPLC Method
Development, 2nd Ed., John Willey and Sons, Inc., Newyork, 1997; 5-1: 351360.
37. Willard HH, Merritt LL, Dean JA, Settle FA, HPLC Theory and
Instrumentation, Instrumental Methods of Analysis, 8th Ed., CBS Publishers
and Distributors, New Delhi, 2002; 1-12.
38. Snyder RI, Kirkland JJ, Glajesh JI, Basics of Separation: Practical HPCL
Method development, 2nd Ed., Published By John Wiley and sons Inc, New
York, 1997; 21-50.

Ganpat University

44

Chapter 1

Introduction

39. Remington, Chromatography; The Science and Practice of Pharmacy Printed


by the Make Printing Company Eston, pennsylvania, 19th Ed. 1995; 1: 537544.
40. Sethi PD, HPLC Quantitative Analysis of Pharmaceutical Formulations, CBS
Publisher and distribors, New Delhi, 2001; 58-67.
41. Backett AH, Stenlke JB, Davidson A, Instrumental Methods in the
development and use of medicines; practical pharmaceutical chemistry, 4th
Ed., CBS Publishers and Distributors New Delhi, 2002; 11: 1-8.
42. Guidance for Industry: Analytical procedure and method validation chemistry,
manufacturing and controls documentation. Centre for drug evaluation and
research, Rockville, Maryland, August, 2004.
43. International Conference on Harmonization: Draft Revised Guidance on Q1A
Stability Testing New drug substances and products, Federal Register, 2000;
65: 246.
44. The United States Pharmacopoeia, 26th Ed., US Pharmacopoeial Convention,
Rockville, MD, 2003; 1151.
45. Munson

JW,

High-performance

liquid

chromatography:

Theory,

instrumentation, and pharmaceutical applications. In; Munson, JW, Eds.


Pharmaceutical analysis modern methos part B, New York, Marcel Dekker,
Inc., 2001; 16-17.
46. Munson JW, Quantitative thin-layer chromatography. In; Munson, J.w., Eds.
Pharmaceutical analysis modern methods part B, New York, Marcel Dekker,
Inc., 2001; 155-156.
47. Albert R, Horwitz W, Validation of analytical procedure, Anal. Chem., 1997;
69: 789.
48. Singh S, Garg S, Understanding of analytical method validation and
characteristics, Pharma Times, 1999; 15-20.
49. Felinger A, Brown P R, Grushka E, Mathematical analysis of multi component
chromatograms, advance in chromatography, Marcel Dekker Inc., New York,
1998; 39: 201-248.
50. Ahuja S, Seypinski S, Handbook of modern pharmaceutical analysis,
academic press, New York, 2001; 346-356: 415-441.
51. The United State Pharmacopoeia, 24th Ed., US Pharmacopoeial Convention,
Rockville, MD, 2000; 1923.
Ganpat University

45

Chapter 1

Introduction

52. ICH, Q2B Validation of Analytical Procedure: Methodology, in: Proceeding


of the International Conference on Harmonization, Geneva, March 1996.
53. ICH, Guidance on Analytical Method Validation, in: Proceedings of
International Convention on Quality for the Pharmaceutical Industry, Toronto,
Canada, September 2002.
54. Bakshi M, Singh S, Development of validated stability-indicating assay
methods-critical review, Journal of Pharmaceutical and Biomedical Analysis,
2002; 28: 1010-1040.
55. Rhodes CT, Introductory overview, Drug Stability: Principle and practices,
J.T.Carstensen, C.T.Rhodes (ed.) Marcel Dekker, New York, 2000: 1-12.
56. Matthews BR, Regulatory aspects of Stability Testing in Europe in: Drug
Stability:Principles and Practices, Carstensen JT and Rhodes C T, 3rd Ed.,
Marcel Dekker, New York, 2000; 107: 580.
57. Hong DD, Shah M, Development and validation of HPLC stability-indicating
assays.in: Drug Stability: Principles and Practices, J.T. Carstensen and C.T.
Rhodes, 3rd Ed., Marcel Dekker, New York, 2000; 107: 329-384.
58. Caviglioli G, Stability Indicating HPLC Assay for Retinoic Acid in Hard
Gelatin Capsules Containing Lactose and as Bulk Drug Substance, Drug
Dev.Ind.Phrm., 1994; 20: 2395-2408.
59. Floery K, Analytical profile of Drug Substances, Academic Press.London.UK,
1979; 8: 217-223.

Ganpat University

46

Chapter 2

Review of Literature

2. REVIEW OF LITERATURE
The drugs are official in IP, BP, and USP. The official methods are based on UV and
HPLC techniques. Some of them require an internal standard, a tedious sample
preparation procedure, costly solvents and maintenance of column temperature etc.
The reported methods are mainly developed keeping in view the requirements of research
mainly in biological fluids using high analytical methods like HPLC-MS, HPTLC, ion
pair chromatography, fluorimetric detection using extraction procedures for sample
preparation. Consequently, many methods are focused on the analysis of the drug in
biological tissues and fluids.
Analytical methods for quantification of muscle relaxant drugs under study are discussed
in this chapter. Table 2.1.1and table 2.2.2 gives a summary of the official methods of
drugs taken under study. Table 2.2.1 gives a review of reported methods drugs as single
and combined formulations.
Several methods have been reported for the analysis of muscle relaxant drugs either in
single drug or mixture of drugs. Analytical methods for muscle relaxant drugs are
summarized in following table.
2.1 OFFICIAL METHODS FOR DRUG UNDER STUDY
Table 2.1.1 Official methods for Paracetamol
Drug
Paracetamol

Method

Description
phase:-

Refere
nce
1
steel

Liquid

Stationary

chromatography

column, 25cm x 4mm long packed with

stainless

octylsilane silica gel


Mobile phase:- Disodium hydrogen
phosphate: methanol (75:25 v/v)
Detection:- 243 nm
Paracetamol

Liquid

Flow rate:- 1.0 mL/min


Stationary phase:- stainless

syrup

chromatography

column, 20cm x 4.6mm long packed with

steel

octadecylsilyl silica gel


Mobile

Ganpat University

phase:-

0.01

Soidumbutane

47

Chapter 2

Review of Literature

sulphonate: methanol (85:15 v/v)


Detection:- 243 nm
Flow rate:- 1.0 mL/min
Paracetamol

Liquid

Stationary

tablets

chromatography

column, 20cm X 4.6mm long packed

method

with octadecylsilane silica gel


Mobile

phase:-

phase:-

butanesuphonate

stainless

0.01
in

85ml

steel

Sodium
water:

methanol and 0.4 volumes of formic acid


Detection:- 243 nm
Paracetamol

Titration

Flow rate:- 1.0 mL/min


Titrant:- 0.1 M Cerium sulphate

Paracetamol

HPLC

Stationary phase:- Nitrile gp bonded

capsule

2
3

with porus silica (3-10m) 3.9mm x


30cm
Mobile phase:- water: methanol (90:10
v/v)
Detection:- 243 nm

Paracetamol

HPLC

oral solution

Flow rate:- 1.0 mL/min


Stationary phase:- Nitrile gp bonded

with porus silica (3-10m) 3.9mm x


30cm
Mobile phase:- water: methanol (90:10
v/v)
Detection:- 243 nm

Paracetamol

UV spectroscopy

Flow rate:- 1.0 mL/min


Wavelength: 243nm

Paracetamol

HPLC

Stationary phase:- Nitrile gp bonded

effervescent

with porus silica (3-10m) 3.9mm x

oral solution

30cm
Mobile phase:- water: methanol (90:10
v/v)

Ganpat University

48

Chapter 2

Review of Literature

Detection:- 243 nm
Flow rate:- 1.0 mL/min
Paracetamol

HPLC

suppository

Stationary phase:- Nitrile gp bonded

with porus silica (3-10m) 3.9mm x


30cm
Mobile phase:- water: methanol (90:10
v/v)
Detection:- 243 nm
Flow rate:- 1.0 mL/min

Paracetamol

HPLC

Stationary phase:- Nitrile gp bonded

oral

with porus silica (3-10m) 3.9mm x

suspension

30cm

Mobile phase:- water: methanol (90:10


v/v)
Detection:- 243 nm
Flow rate:- 1.0 mL/min
Paracetamol

HPLC

tablet

Stationary phase:- Nitrile gp bonded

with porus silica (3-10m) 3.9mm x


30cm
Mobile phase:- water: methanol (90:10
v/v)
Detection:- 243 nm
Flow rate:- 1.0 mL/min

Paracetamol

HPLC

Stationary phase:- Nitrile gp bonded

extended

with porus silica (3-10m) 3.9mm x

release

30cm

Mobile phase:- water: methanol (90:10


v/v)
Detection:- 243 nm
Flow rate:- 1.0 mL/min

Ganpat University

49

Chapter 2

Review of Literature

Table 2.1.2 Official methods for Paracetamol in combination


Drug

Method

Paracetamol

HPLC

and Aspirin

Description

Reference

Stationary phase:- Nitrile gp bonded

with porus silica (3-10m) 3.9mm x


30cm
Mobile

phase:-

Chloroform:

methanol: glacial acetic acid (70:20:1


v/v/v)
Detection:- 280 nm
Flow rate:- 1.0 mL/min
Paracetamol,
Aspirin

HPLC

and

Stationary phase:- Nitrile gp bonded

with porus silica (3-10m) 4.6mm x

Caffeine

10cm
Mobile

phase:-

Chloroform:

methanol: glacial acetic acid (69:28:3


v/v/v)
Detection:- 275 nm
Flow rate:- 1.0 mL/min
Paracetamol

HPLC

and Caffeine

Stationary phase:- Nitrile gp bonded

with porus silica (3-10m) 4.6mm x


10cm
Mobile

phase:-

Chloroform:

methanol: glacial acetic acid (69:28:3


v/v/v)
Detection:- 275 nm
Flow rate:- 1.0 mL/min
Table 2.1.3 Official methods for Diclofenac potassium in combination
Drug
Diclofenac
potassium

Method
Liquid
chromatography

Description
Stationary phase:-

Reference
4

Mobile phase:- Methanol: phosphate


buffer (pH 2.5) (70:30 v/v)

Ganpat University

50

Chapter 2

Review of Literature

Detection:- 254 nm
Diclofenac
potassium
tablets

Flow rate:- 1.0 mL/min


Stationary phase:-

HPLC

Mobile phase:- Methanol: phosphate


buffer (pH 2.5) (70:30 v/v)
Detection:- 254nm

Diclofenac
potassium
delayed
release

Liquid
chromatography

Flow rate:- 1.0 mL/min


Stationary phase:-

Mobile phase:- Methanol: phosphate


buffer (pH 2.5) (70:30 v/v)
Detection:- 254 nm
Flow rate:- 1.0 mL/min

Diclofenac
tablet

Stationary phase:- Plate coated with

TLC

silica gel GF254


Mobile phase:- Toluene: hexane:
formic acid (90:5:5 v/v/v)
Detection:- 254 nm

Diclofenac
sodium
injection

Stationary phase:- Plate coated with

TLC

silica gel GF254 plate


Mobile

phase:-

Chloroform:

acetone: formic acid (90:5:5 v/v/v)


Detection:- 254 nm
Diclofenac
potassium

Potentiometric

Solvent:- Anhyrous acetic acid

Titrant:- 0.1M perchloric acid

Table 2.1.4 Official methods for Chlorzoxazone in combination


Drug

Method

Chlorzoxazone Liquid
tablets
chromatography

Description
Stationary phase:-

Reference
7

Mobile phase:- Water: acetonitrile:


glacial acetic acid (70:30:1 v/v/v)
Detection:- 280 nm
Flow rate:- 1.5 mL/min

Ganpat University

51

Chapter 2

Review of Literature

Table 2.1.5 Official methods for Methocarbamol


Drug

Method

Description

Reference

Methocarbamol UV

Detection:- 274 nm

Methocarbamol UV

Detection:- 274 nm

Methocarbamol Liquid

Stationary phase:-

tablet

Mobile

injections
chromatography

phase:-

buffer(pH 4.5): methanol

Phospate
(75:25

v/v)
Detection:- 280 nm
Flow rate:- 1.5 mL/min

Ganpat University

52

Chapter 2

Review of Literature

2.2 REPORTED METHODS FOR DRUG UNDER STUDY


Table 2.2.1 Reported methods for Paracetamol
Drug
Paracetamol

Title:-

Method and description

Reference
9

Reverse Phase HPLC Method for Determination of


Aceclofenac and Paracetamol in Tablet Dosage Form.
Description
Stationary phase:- ODS C18
Mobile phase:- methanol: water (70:30 v/v)
Flow rate:- 1.0 mL/min
Detection:- 245 nm
Paracetamol

10

Title:Reverse phase HPLC method for determination of


aceclofenac and paracetamol in tablet dosage forms
Description
Stationary phase:- ODS C18
Mobile phase:- methanol: water (70:30 v/v)
Flow rate:- 1.0 mL/min
Detection:- 255 nm

Paracetamol

11

Title:RP-HPLC method for determination of aceclofenac,


chlorzoxazone

and

paracetamol

in

bulk

and

pharmaceutical formulation
Stationary phase:- Reverse Phase C18
Mobile phase:- acetonitrile: water (60:40 v/v)
Flow rate:- 1.0 mL/min
Detection:- 230 nm

Ganpat University

53

Chapter 2

Paracetamol

Review of Literature

12

Title:Validation of a RP-HPLC method for simultaneous


determination of paracetamol, methocarbamol, and
diclofenac potassium in tablets
Description:Stationary phase:- C18 column ODS Hypersil
Mobile phase:- 25mM phosphate buffer: acetonitrile
(65:35 v/v)
Flow rate:- 1.2 mL/min
Detection:- 225 nm

Paracetamol

13

Title:Simultaneous

determination

of

paracetamol,

pseudoephedrine, dextrophan and chlorpheniramine in


human plasma by liquid chromatographytandem mass
spectrometry
Description:Stationary phase:- C18 column ODS
Mobile phase:- acetic acid: methanol (20:80 v/v)
Flow rate:- 0.30 mL/min
Detection:- 275 nm
Paracetamol

Title:-

14

HPLC Method for the Analysis of Paracetamol,


Caffeine and Dipyrone
Description:Stationary phase:- C8 column ( Bondapack)
Mobile phase:- methanol: acetonitrile: isopropyl alcohol
(40:30:30 v/v)
Flow rate:- 1.0 mL/min
Detection:- 215 nm

Ganpat University

54

Chapter 2

Paracetamol

Review of Literature

15

Title:
Spectrophotometric

Methods

for

Simultaneous

Estimation of Dexibuprofen and Paracetamol


Description:
Solvent:- methanol
Wavelength:- 249.5 nm
Paracetamol

16

Title:
Simultaneous

determination

of

paracetamol

and

lornoxicam by RP-HPLC in bulk and tablet formulation


Description:
Stationary phase:- C18 column ODS
Mobile phase:- methanol : water (85:15v/v)
Flow rate:- 1.0 mL/min
Detection:- 259 nm
Paracetamol

17

Title:
Determination

of

paracetamol

and

tramadol

hydrochloride in pharmaceutical mixture using HPLC


and GC-MS
Description:
Stationary phase:- C18 column
Mobile phase:- phosphate buffer: acetonitrile (90:10 v/v)
Detection:- 220 nm
Paracetamol

18

Title:
Sensitive

liquid

chromatographytandem

mass

spectrometry method for the simultaneous determination


of paracetamol and guaifenesin in human plasma
Description:
Stationary phase:- C18 column
Mobile phase:- phosphate buffer: methanol (80:20 v/v)
Detection:- 225 nm

Ganpat University

55

Chapter 2

Paracetamol

Review of Literature

Title:Simultaneous

Determination

of

Paracetamol

and

19

Caffeine in Human Plasma by LCESIMS


Description:
Stationary phase:- C18
Mobile phase:- formic acid: methanol (60:40 v/v)
Detection:- 254 nm
Paracetamol

20

Title:
UV

spectrophotometric

development

for

and

simultaneous

RP-HPLC

method

determination

of

paracetaomol and etodolac in pharmaceutical dosage


form
Description:
Solvent:- methanol
Wavelength:- 226 nm
Paracetamol

Title:

21

Stability indicating method for the determination of


paracetamol in its pharmaceutical preparations by TLC
densitometric method
Description:
Stationary phase:- silica gel 60F254
Mobile phase:- acetic acid: benzene: acetic acid
(1:1:0.05 v/v/v)
Detection:- 250 nm
Range:- 5-20 g/spot

Ganpat University

56

Chapter 2

Paracetamol

Review of Literature

Title:-

22

The quantification of Paracetamol glucuronide and


Paracetamol sulphate in plasma and urine using a single
high-performance liquid chromatography assay
Description:
Wavelength:- 254 nm
Mobile phase:- KH2PO4: isopropanol-TGF (98:1.5:0.1
v/v/v)
Paracetamol

Title:-

23

Liquid chromatographic determination of cetrizine


hydrochloride and paracetamol in human plasma and
pharmaceutical formulations
Description:
Stationary phase:- C18 column
Mobile phase:- acetonitrile: water (55:45 v/v)
Flow rate:- 1.0 mL/min
Detection:- 230 nm
Paracetamol

Title:-

24

Determination of paracetamol and orphenadrine citrate in


dosage formulations and in human plasma
Description:
Stationary phase:- C18 column
Mobile phase:- acetonitrile: water (50:50 v/v)
Flow rate:- 1.0 mL/min
Detection:- 215 nm

Ganpat University

57

Chapter 2

Paracetamol

Review of Literature

25

Title:A

validated

paracetamol

method
and

its

for

the

determination

glucuronide

and

of

sulphate

metabolites in the urine of HIV/AIDS patients using


wavelength-switching UV detection
Description:
Stationary phase:- C18 column
Mobile phase:- 50mM sodium acetate buffer :
acetonitrile (96:4 v/v)
Flow rate:- 1.0 mL/min
Wavelength:- 254 nm
Paracetamol

26

Title:Simultaneous
Tolperisone

estimation

of

hydrochloride

in

Paracetamol
tablet

by

and
UV

spectrophotometric methods
Description:
Solvent:- metanol
Wavelength:- 254 nm
Paracetamol

27

Title:Simultaneous Determination of Paracetamol and


Piroxicam in Tablets by Thin Layer Chromatography
Combined with Densitometry
Description:
Stationary phase:- silica gel GF254
Mobile

phase:-

n-Dichloroethane:

methanol:

triethylamine (10:2.5:1 v/v/v)


Detection:- 288 nm
Range:- 1.625-14.625 g/spot

Ganpat University

58

Chapter 2

Paracetamol

Review of Literature

28

Title:RP-UPLC

method

for

combinational

assay

of

lornoxicam and paracetamol in combined tablet dosage


form
Description
Stationary phase:- UPLC BEH C18
Mobile phase:- acetonitrile:30mM Ammonium acetate
(40:50, v/v)
Flow rate:- 0.5 mL/min
Detection:- 260 nm
Paracetamol

Title:Simultaneous

determination

of

aceclofenac,

29

paracetamol, and chlorzoxazone by RP-HPLC in


pharmaceutical dosage form.
Description
Stationary phase:- Column:- C18 (Zorbax )
Mobile phase:- acetonitrile : buffer (40:60 v/v)
Flow rate:- 1.0 mL/min
Detection:- 220 nm
Paracetamol

Title:-

30

Simultaneous HPTLC Determination of paracetamol


and dexketoprofen trometamol in pharmaceutical dosage
form
Description
Stationary phase:- Silica gel G 60F254 plates
Mobile phase:- toluene: ethyl acetate: acetic acid (6: 4:
0.2 v/v/v)
Detection:- 256 nm
Range:- 25-150 ng/spot

Ganpat University

59

Chapter 2

Review of Literature

Table 2.2.2 Reported methods of Diclofenac potassium


Diclofenac

Title:-

31

Validated stability indicating RP-HPLC method for


simultaneous determination & in vitro dissolution studies of
thiocolchicoside and diclofenac potassium from tablet
dosage forms.
Description
Stationary phase:- C18 column (Zorbax SB CN)
Mobile phase:- 5 mM sodium dihydrogen phosphate:
methanol (60:40 v/v)
Flow rate:- 1.0 mL/min
Detection:- 258 nm
Diclofenac

Title:-

32

Development of UV spectrophotometric methods for


simultaneous estimation of Famotidine and Diclofenac
potassium in combined dosage form using simultaneous
equation method
Description
Solvent:- methanol
Detection:- 286 nm
Diclofenac

Title:-

33

New Stability-Indicating RP-HPLC Method for


Determination of Diclofenac Potassium and Metaxalone
from Their Combined Dosage Form
Description
Stationary phase:- C18 column (Hibar Lichrosphere-100)
Mobile phase:- methanol: water (80:20 v/v)
Flow rate:- 1.0 mL/min
Detection:- 280 nm

Ganpat University

60

Chapter 2

Diclofenac

Review of Literature

34

Title:A

validated

RP-HPLC

method

for

simultaneous

estimation of paracetamol and diclofenac potassium in


pharmaceutical formulation
Description
Stationary phase:- C18 column (Phenomenex LUNA)
Mobile phase:- acetonitrile: sodium dihydrogen ortho
phosphate (70:30 v/v)
Flow rate:- 1.0 mL/min
Detection:- 278 nm
Diclofenac

Title:-

35

Simultaneous estimation of diclofenac and rabeprazole


by high performance liquid chromatographic method in
combined dosage forms
Description
Stationary phase:- C18 column (HiQ SiL)
Mobile phase:- water: methanol: acetonitrile (20:40:40
v/v)
Flow rate:- 1.2 mL/min
Detection:- 284 nm
Diclofenac

Title:-

36

Validated HPLC Method for Simultaneous Quantitation


of
Diclofenac Sodium and Misoprostol in Bulk Drug and
Formulation
Description
Stationary phase:- C18 (Hypersil BDS)
Mobile phase:- acetonitrile: water (85:15 v/v)
Flow rate:- 1.0 mL/min
Detection:- 220 nm

Ganpat University

61

Chapter 2

Diclofenac

Review of Literature

Title:-

37

High Performance Thin Layer Chromatographic method


for determination of diclofenac sodium in pharmaceutical
formulations
Description
Stationary phase:- silica gel 60 F254 aluminium plate
Mobile phase:- toluene : ethyl acetate : glacial acetic acid
(60:40:1 v/v/v)
Detection:- 282 nm
Linearity:- 5-80 g/mL
Diclofenac

Title:-

38

Validated HPTLC method for simultaneous estimation of


diclofenac potassium and metaxalone in bulk drug and
formulation
Description
Stationary phase:- silica gel 60 F254 aluminum plate
Mobile phase:- chloroform: methanol (8:1 v/v)
Detection:- 282 nm
Linearity:- 100-180 ng/mL
Diclofenac

Title:-

39

Development and validation of a bioanalytical method for


direct extraction of diclofenac potassium from spiked
plasma
Description
Stationary phase:- C18 column (Hypersil)
Mobile phase:- acetonitrile: triethylamine (80:20 v/v)
Flow rate:- 1.0 mL/min
Detection:- 276 nm

Ganpat University

62

Chapter 2

Diclofenac

Review of Literature

40

Title:A RP-HPLC method for determination of diclofenac


with rabeprazole in solid dosage form
Description
Stationary phase:- C18 column (Phenomenox)
Mobile phase:- acetonitrile: 50mM ammonium acetate
buffer (60:40 v/v)
Flow rate:- 1.0 mL/min
Detection:- 254 nm

Diclofenac

41

Title:Development and validation of a HPTLC method for


simultaneous

densitometric

analysis

of

diclofenac

potassium and dicyclomine hydrochloride as the bulk


drugs and in the tablet dosage form
Description
Stationary phase:- silica gel 60 F254 plates
Mobile phase:- toluene:methanol:acetic acid (8:2:0.1
v/v/v)
Wavelengh:- 272 nm
Linearity:- 0.2-1.6 ng/spot

Ganpat University

63

Chapter 2

Review of Literature

Table 2.2.3 Reported methods for Chlorzoxazone


Chlorzoxazone

42

Title:Simultaneous estimation of paracetamol, chlorzoxazone and


diclofenac potassium in pharmaceutical formulation by RPHPLC method
Description
Stationary phase:- C18 column
Mobile

phase:-

methanol-0.01M

monobasic

sodium

phosphate (70:30 v/v)


Flow rate:- 1.0 mL/min
Detection:- 254 nm
Chlorzoxazone

43

Title:Simultaneous determination of paracetamol, diclofenac


sodium and chlorzoxazone by HPLC from tablet
Description
Stationary phase:- C18 column (Sodex)
Mobile phase:- methanol: water: triethylamine (55:45:0.1
v/v/v)
Flow rate:- 1.0 mL/min
Detection:- 275 nm

Chlorzoxazone

44

Title:Determination

of

6-hydroxychlorzoxazone

and

chlorzoxazone in porcine microsome samples


Description
Stationary phase:- C18 column (ODS-AQ)
Mobile phase:- phosphoric acid: methanol (60:40 v/v)
Flow rate:- 1.0 mL/min
Detection:- 287 nm

Ganpat University

64

Chapter 2

Chlorzoxazone

Review of Literature

45

Title:Simultaneous estimation of tramadol hydrochloride and


chlorzoxazone by absorbance correction method
Description
Solvent:- methanol
Wavelength:- 225 nm

Chlorzoxazone

46

Title:Determination

of

Cytochrome

P450

2El

Activity

in

Microsomes by Thin-Layer Chromatography Using [2-14C]


Chlorzoxazone
Description
Stationary phase:- silica gel 60 F254 plates
Mobile phase:- acetone: hexane (45:55 v/v)
Flow rate:- 1.0 mL/min
Detection:- 287 nm
Chlorzoxazone

47

Title:The Simultaneous Estimation of aceclofenac, paracetamol and


chlorzozazone in pharmaceutical dosage forms by HPTLC
Description
Stationary phase:- silica gel G60F254
Mobile phase:- toluene: ethyl acetate: glacial acetic acid
(17.5:10:0.5 v/v)
Detection:- 271 nm

Chlorzoxazone

48

Title:Simultaneous determination of paracetamol and chlorzoxazone


using orthogonal functions ratio spectrophotometry
Description
Solvent:- methanol
Wavelength:- 287 nm

Ganpat University

65

Chapter 2

Review of Literature

Table 2.2.4 Reported methods for Methocarbamol


Methocarbamol

49

Title:Simultaneous spectrophotometric estimation of ibuprofen


and methocarbamol in tablet dosage form
Description
Solvent:- methanol
Wavelengh of ibuprofen:- 222 nm
Wavelengh of methocarbamol:- 224 nm

Methocarbamol

50

Title:Simultaneous

determination

of

paracetamol

and

methocarbamol in tablets by ratio spectra derivative


spectrophotometry and LC
Description
Solvent:- methanol
Wavelengh of paracetamol:- 243 nm
Wavelengh of methocarbamol:- 230 nm
Methocarbamol

Title:-

51

Determination of acetaminophen and methocarbamol in


Bilayered tablets using RP-HPLC
Description
Stationary phase:- C18 column (ODS-MG-5)
Mobile phase:- water: methanol: glacial acetic (600:400:15)
Flow rate:- 1.0 mL/min
Detection:- 273 nm
Methocarbamol

Title:-

52

Determination of methocarbamol concentration in human


plasma by high performance liquid chromatography
tandem mass spectrometry
Description
Range:- 150-12000 ng/mL

Ganpat University

66

Chapter 2

Methocarbamol

Review of Literature

53

Title:A

stability-indicating

chromatographic

method

high-performance
for

the

liquid

determination

of

methocarbamol in veterinary preparations


Description
Stationary phase:- C18 column
Mobile phase:- methanol: water: tetrahydrofuran (25:65:10
v/v/v)
Flow rate:- 0.9 mL/min
Detection:- 274 nm
Methocarbamol

Title:-

54

Stability indicating HPLC method for simultaneous


determination of methocarbamol and nimesulide from tablet
matrix
Description
Stationary phase:- supercosil LC-8
Mobile phase:- methanol: water (60:40 v/v)
Flow rate:- 1.2 mL/min
Detection:- 276 nm
Methocarbamol

Title:-

55

Simultaneous HPLC Determination of methocarbamol,


paracetamol and diclofenac Sodium
Description
Stationary phase:- supelcosil LC-8
Mobile phase:- methanol: water: glacial acetic acid
(400:600:05 v/v/v)
Flow rate:- 0.8 mL/min
Detection:- 275 nm

Ganpat University

67

Chapter 2

Methocarbamo
l

Review of Literature

Title:-

56

Simultaneous determination of diclofenac potassium and


methocarbamol in ternary mixture with guaifenesin by
reversed phase liquid chromatography
Description
Stationary phase:- C18 column (Symmetry Waters)
Mobile phase:- phosphate buffer: acetonitrile (20:80 v/v)
Flow rate:- 1.0 mL/min
Detection:- 282 nm

Ganpat University

68

Chapter 2

Review of Literature

References
1. Indian Pharmacopoeia Vol.II, Government of India, Ministry of health &
family welfare, published by the Indian Pharmacopoeial Commission, 2007;
1514-1516.
2. British Pharmacopoeia Vol.II, Department of health, social services & public
safety, London: stationary office, 2007: 1575-1576.
3. United States of Pharmacopoeia NF vol.II, The official compendia of
standards, Asian edition, 2007; 1388-1392.
4. Indian Pharmacopoeia Vol.II, Government of India, Ministry of health &
family welfare, published by the Indian Pharmacopoeial Commission, 2007;
1020-22.
5. United States of Pharmacopoeia NF Vol.I, The official compendia of
standards, Asian edition, 2007; 1921-1924.
6. British Pharmacopoeia Vol.I, Department of health, social services & public
safety, London: stationary office, 2007: 665.
7. United States of Pharmacopoeia NF Vol.II, The official compendia of
standards, Asian edition, 2007; 1741.
8. United States of Pharmacopoeia NF Vol.II, The official compendia of
standards, Asian edition, 2007; 2610.
9. Godse VP, Deodhar MN, Bhosale AV, Sonawane RA, Sakpal PS, Borkar DD,
Bafana YS, Reverse Phase HPLC method for determination of aceclofenac
and paracetamol in Tablet Dosage Form, Asian J. Research Chemistry, 2009;
(2)1: 37-40.
10. Momin MY, Yeole PG, Puranik MP, Wadher SJ, Reverse phase HPLC
method for determination of aceclofenac and paracetamol in tablet dosage
forms, Indian Journal of Pharmaceutical Science, 2006; 68(3): 387-389.
11. Yadav AH, Kothapalli LP, Barhate AN, Pawar

I, Pawar CR, RP-HPLC

method for determination of aceclofenac, chlorzoxazone and paracetamol in


bulk and pharmaceutical formulation, International Journal of Pharma
Research and Development, 2009; 1(10): 1-7.

Ganpat University

69

Chapter 2

Review of Literature

12. Subraminian G, Vasudevan M, Ravishakar S, Suresh B, Validation of a RPHPLC

method

for

simultaneous

determination

of

paracetamol,

methocarbamol, and diclofenac potassium in tablets, Indian Journal of


Pharmaceutical Sciences, 2005; 4(2): 260-263.
13. Hong Gang L, Hong Y, Ruan Z, Jiang B, Simultaneous determination of
Paracetamol, pseudoephedrine, dextrophan and chlorpheniramine in human
plasma by liquid chromatography-tandem mass spectrometry, Journal of
Chromatography B, 2010; 878: 682-688.
14. Levent AM, HPLC Method for the Analysis of Paracetamol, caffeine and
dipyrone, Turk J Chemistry, 2002; 26(1): 521-528.
15. Chitlange S, Soni R, Wankhede S, Kulkarni A, Spectrophotometric Methods
for Simultaneous Estimation of Dexibuprofen and Paracetamol, Asian J.
Research Chem. 2009; 2(1): 30-33.
16. Jagtap AS, Yadav SS ,Rao JS, Simultaneous determination of paracetamol
and lornoxicam by RP-HPLC in bulk and tablet formulation, Pharmacie
Globale, 2011; 9(4): 1-4.
17. Belal T, Awad T, Clark C R, Determination of paracetamol and tramadol
hydrochloride in pharmaceutical mixture using HPLC and GC-MS, Journal of
chromatographic science, 2009; 47(10): 849-854.
18. Chen X, Huang J, Kong Z, Zhong D, Sensitive liquid chromatography-tandem
mass spectrometry method for the simultaneous determination of paracetamol
and guaifenesin in human plasma, Journal of Chromatography B, 2005;
817(2): 263-269.
19. Anna Wang, Jin Sun, Haijun Feng, Shuo Gao and Zhonggui He, Simultaneous
Determination of Paracetamol and Caffeine in Human Plasma by LCESI
MS, Chromatographia, 2010; 67(3): 281-285.
20. Jadi MK, Narayan UL, UV spectrophotometric and RP-HPLC method
development for simultaneous determination of paracetaomol and etodolac in
pharmaceutical dosage form, 2009; 56(1): 169-174.

Ganpat University

70

Chapter 2

Review of Literature

21. Nadia M. Mostafa, Stability indicating method for the determinationof


paracetamol in its pharmaceutical preparations by TLC densitometric method,
Journal of Saudi Chemical Society 2010; 14: 341-344.
22. Jensen LS, Valentine J, Milne RW, Evans AM, The Quantification of
Paracetamol, Paracetamol Glucuronide and Paracetamol Sulphate in Plasma
and Urine Using a Single High-Performance Liquid Chromatography Assay,
J. Pharm. Biomed. Anal.,2004; 34: 585-593
23. Basavaraj S, Nagaralli J, Seetharamappa B, Gowda G, Mahaveer B, Liquid
Chromatographic Determination of Ceterizine and Paracetamol in Human
Plasma and Pharmaceutical Formulations, J. Chromatogr. B., 2003; 798: 4954.
24. Arayne MS, Determination of paracetamol and orphenadrine citrate in dosage
formulations and in human plasma, Journal of Chinese Chemical Society,
2009; 56: 169-174.
25. Girolamo O, Neill M, Wainer IW, A Validated Method for the Determination
of Paracetamol and its Glucuronide and Sulphate Metabolites in the Urine of
HIV/AIDS Patients Using Wavelength-Switching UV Detection, J. Pharm.
Biomed. Anal., 1998; 17: 1191-1197.
26. Patel MG, Parmar RR, Nayak PP, Shah SA, Simultaneous estimation of
Paracetamol

and

Tolperisone

hydrochloride

in

tablet

by

UV

spectrophotometric methods, Journal of Pharmaceutical Science and


Bioscientific Research, 2012; 2(2) 63-67.
27. Atul A, Shirkhedkar, Afsar M. Shaikh and Sanjay J. Surana, Simultaneous
determination of paracetamol and piroxicam in tablets by TLC combined with
densitometry, Shirkhedkar A A, Shaikh A M, Surana S J, Eurasian Journal of
Analytical Chemistry, 2008; 3(2): 258-267.
28. Srinivasu T, Rao BN, Annaoura MM, Chandrashekhar TG, RP-UPLC method
for combinational assay of lornoxicam and paracetamol in combined tablet
dosage form, International Journal of Pharma. Science & Research, 2012;
3(4): 1149-1154.

Ganpat University

71

Chapter 2

Review of Literature

29. Shaikh KA, Devkhile AB, Simultaneous determination of aceclofenac,


paracetamol, and chlorzoxazone by RP-HPLC in pharmaceutical dosage form,
Journal of Chromatographic Sciences, 2008; 46(7): 649-652.
30. Rao JR, Mulla TS, Bharekar VV, Yadav SS, Rajput MP, Simultaneous
HPTLC determination of paracetamol and dexketoprofen trometamol in
pharmaceutical dosage form, Der Pharma Chemica, 2011; 3(3): 32-38.
31. Jadhav SD, Butle SR, Patil SD, Jagdap P K, Validated stability indicating RPHPLC method for simultaneous determination & in vitro dissolution studies of
thiocolchicoside and diclofenac potassium from tablet dosage forms., Arabian
Journal of Chemistry, 2012, Article in Press.
32. Mehta SA, Umarkar AR, Chaple DR, Thote LT, Development of UV
Spectrophotometric Methods for Simultaneous Estimation of Famotidine and
Diclofenac Potassium in Combined Dosage Form Using Simultaneous
Equation Method, Journal of Pharmacy Research, 2011; 4(7), 2045-2046.
33. Panda SS, Patnaik d, Ravikumar B, New Stability-Indicating RP-HPLC
Method for
Determination of Diclofenac Potassium and Metaxalone from Their
Combined Dosage Form, Scientia Pharmaceutica, 2012, Article in Press.
34. Gowramma B, Rajan S, Muralidharan S, Meyyanathan S, and Suresh B, A
validated RP-HPLC method for simultaneous estimation of paracetamol and
diclofenac potassium in pharmaceutical formulation, International Journal of
Chemtech Research, 2010; 2(1), 676-680.
35. Choudhary B, Goyal A, Khokra S , Kaushik B, Simultaneous estimation of
diclofenac and rabeprazole by high performance liquid chromatographic
method in combined dosage forms, International Journal of Pharmaceutical
Science and drug research, 2009; 1(1): 43-45.
36. Dhaneshwar SR, Bhusari VK, Validated HPLC method for simultaneous
quantitation of

diclofenac sodium and misoprostol in bulk drug and

formulation, Pelagia Research Library Der Chemica Sinica, 2010; 1(2): 110118.

Ganpat University

72

Chapter 2

Review of Literature

37. Thongchai W, Liawruangrath B, Thongpoon C, MachanHigh T, High


Performance Thin Layer Chromatographic method for determination of
diclofenac sodium in pharmaceutical formulations, Chiang Mai J. Sci. 2006;
33(1): 123-128.
38. Rajput MP, Bharekar VV, Yadav SS, Mulla TS, Rao JR,Validated HPTLC
method for simultaneous estimation of diclofenac potassium and metaxalone
in bulk drug and formulation, Pharmacie Globale International Journal of
Comprehensive Pharmacy, 2011; 12 (04): 1-4.
39. Sarfraz A, Sarfraz M, Ahmad M, Development and validation of a
bioanalytical method for direct extraction of diclofenac potassium from spiked
plasma, Tropical Journal of Pharmaceutical Research, 2011; 10(5): 663-69.
40. Arunadevi S, Subramania M, Suresh B, A RP-HPLC method for
determination of diclofenac with rabeprazole in solid dosage form, Pharma
science monitor, 2011; 2(2): 171-178.
41. Potawale SE, Nanda RK, Bhagwat VV, Hamane SC, Deshmukh S,
Puttamsetti S, Development and validation of a HPTLC method for
simultaneous

densitometric

analysis

of

Diclofenac

potassium

and

Dicyclomine hydrochloride as the bulk drugs and in the tablet dosage form,
Journal of Pharmacy Research, 2011; 4(9): 3116-3118.
42. Biswas A, Basu A, Simultaneous estimation of paracetamol, chlorzoxazone
and diclofenac potassium in pharmaceutical formulation by RP-HPLC
method, International Journal of Pharma and Bio Sciences, 2010; 1(2): 1-5.
43. Shinde VM, Desai BS, Tendolkar NM, Simultaneous determination of
paracetamol, diclofenac sodium and chlorzoxazone by HPLC from tablet,
Indian Journal of Pharma. Sciences, 1995; 57(1): 35-37.
44. Sherry K, Tina H, Joe B, Determination of 6-hydroxychlorzoxazone and
Chlorzoxazone in Porcine Microsome samples, J. Chromatogr. B, 2003; 78:
111-116.
45. Thakur AD, Hajare AL, Nikhade RD, Chandewar AV, Simultaneous
estimation of tramadol hydrochloride and chlorzoxazone by absorbance
correction method, Journal of Pharmacy Research, 2011,4(6): 1683-1684.
Ganpat University

73

Chapter 2

Review of Literature

46. Zerillia A, Lucas D, Berthou F, Determination of Cytochrome P450 Activity


in Microsomes by Thin-Layer Chromatography Using Chlorzoxazone, J.
Chromatogr. B, 1996, 677: 156-160.
47. Yuvaraj G, Pavan Kumar P, Elangovan N, The Simultaneous Estimation of
Aceclofenac, Paracetamol and Chlorzozazone in Pharmaceutical Dosage
Forms by HPTLC, International Journal of Chemical and Pharmaceutical
Sciences, 2010; 1(2): 24-27.
48. Wahbi AM, Gazy AA, Abdel-Razak O, Mahgoub H, Moneeb MS,
Simultaneous determination of paracetamol and chlorzoxazone using
orthogonal functions- ratio spectrophotometry, Saudi Pharmaceutical Journal,
2003; 11(4): 192-200.
49. Satheeshamanikandan TRS, Wali DC, Banwal J, Kadam SS, Dhaneshwar SR,
Simultaneous spectrophotometric estimation of ibuprofen and methocarbamol
in tablet dosage form, Indian Journal of Pharmaceutical Sciences, 2004; 16:
810-813.
50. Erk N, Zkan Y, Banog E, Simultaneous Determination of Paracetamol and
Methocarbamol in Tablets by Ratio Spectra Derivative Spectrophotometry
and LC, J. Pharm. Biomed. Anal.24, 2000; 14: 469-475.
51. Mallikarjuna RB, Madhavi BR, DurgaDevi NK, Praveen PS, Mrudula BS,
Neelima RT, Determination of Acetaminophen and Methocarbamol in
Bilayered tablets using RP-HPLC, International Journal of Chemical and
Analytical Science 2010; 1(7): 158-160.
52. Zha W, Zhu Z, Determination of methocarbamol concentration in human
plasma

by

high

performance

liquid

chromatographytandem

mass

spectrometry, J Chromatogr B Analyt Technol Biomed Life Sci., 2010;


15(10): 831-835
53. Rosasco MA, Ceresole RS, Forastieri CC, Segall AI, A stability-indicating
high-performance liquid chromatographic method for the determination of
methocarbamol in veterinary preparations, J AOAC Int, 2009; 92(5): 16021605.

Ganpat University

74

Chapter 2

Review of Literature

54. Manmode RS, Dhamankar AK, Manwar JV, and Laddha SS, Stability
indicating HPLC method for simultaneous determination of methocarbamol
and nimesulide from tablet matrix, Pelagia Research Library Der Chemica
Sinica, 2011; 2(4): 81-85.
55. Hafsa D, Chanda S, Prabhu P, Simultaneous HPLC Determination of
Methocarbamol, Paracetamol and Diclofenac Sodium, E Journal of
Chemistry, 2011; 8(4): 1620-1625.
56. Elkady EF, Simultaneous determination of diclofenac potassium and
methocarbamol in ternary mixture with guaifenesin by reversed phase liquid
chromatography, Talanta, 2010; 82(4): 1604-1607.

Ganpat University

75

Chapter 3

Aim of the present work

3. AIM OF THE PRESENT WORK


There are rapidly increasing number of drug groups (and increasing numbers of drugs
within each group) available to treat muscle constriction. A day by day number of newer
muscle relaxant formulations either in single or in combined dosage forms are marketed
as well as is under investigation. Literature reveals that numbers of methods are available
for analysis of muscle relaxant drugs but so far stability indicating methods have not been
reported.
The objective of this work was to develop and validate chromatographic methods, which
would serve as stability indicating assay method for drug product. So the present
investigation was undertaken with a view to develop and validate new chromatographic
methods which could be applied for routine analysis of muscle relaxant drugs in
formulations. It will be tried that the methods should be,
Simple
Accurate
Precise
Selective
Specific
Reproducible
Highly sensitive
Stability indicating
The proposed methods can be made useful in routine analysis of such drugs in bulk and
in pharmaceutical formulations in single as well as in multi drug combinations in a
simple, convenient, and cost effective way.
THUS AIM OF THE PRESENT WORK INCLUDES:
To develop and validate stability indicating RP-HPLC and HPTLC method for
simultaneous

estimation

of

Diclofenac

Potassium,

Chlorzoxazone

and

Paracetamol in bulk powder and their pharmaceutical formulations.


To develop and validate stability indicating RP-HPLC and HPTLC method for
simultaneous

estimation

of

Diclofenac

Potassium,

Paracetamol

and

Methocarbamol in bulk powder and their pharmaceutical formulations.

Ganpat University

76

Chapter 4

Chemicals, glasswares and instruments

4. CHEMICALS, GLASSWARES AND INSTRUMENTS


Chemicals used were of analytical (AR) grade except specified and the glasswares used
in the experimental work were calibrated in the laboratory and out of all only those were
in calibration limit were selected for work[1]. The glasswares were cleaned according to
the pharmacopoeial procedure before they were used for the experiment [2]. While the
instruments were used for the work were passed through routine calibration procedure.
4.1 Chemicals
Water-triple distilled
Methanol-HPLC grade (S.D. Fine Chemicals, Mumbai)
Acetonitrile-HPLC grade (S.D. Fine Chemicals, Mumbai)
Disodium hydrogen phosphate (Merck Ltd, Mumbai )
Sodium dihydrogen phosphate (Merck Ltd, Mumbai)
Dimethyl formamide (Rankem Ltd, New Delhi )
Ammonium acetate (Rankem Ltd, New Delhi)
Sodium hydroxide (Rankem Ltd, New Delhi )
Hydrochloric acid (Merck Ltd, Mumbai )
Hydrogen peroxide (Merck Ltd, Mumbai)
Glacial acetic acid (Rankem Ltd, Mumbai)
O-Phosphoric acid (Merck Ltd, Mumbai)

Ganpat University

77

Chapter 4

Chemicals, glasswares and instruments

4.2 Active Pharmaceutical Ingredients (API)


Table 4.2.1: List of Active Pharmaceutical Ingredient
Sr.

Name

Batch No

A.R.No.

Supplier

DFK/09/0018

Zydus Cadila

No.
1

Diclofenac

DFK/19040018

potassium
2

Chlorzoxazone

Ltd
MNB/94/2

CHA/10/0153

Zydus

Cadila

Ltd
3

Paracetamol

0509/11

FPC/PRL/0509/11

Zydus

Cadila

Ltd
4

Methocarbamol

CN/1127

ARD3170185

Zydus

Cadila

Ltd
4.3 Glasswares
Volumetric flask (10, 25, 50, 100, 250 mL)
Graduated pipette (1, 2, 5, 10 mL)
Measuring cylinder (50, 100 mL)
Thermometer (100C)
Petri-dish
Conical flask (100, 250 mL)
Glass beaker (50, 100, 250, 500 mL)
Plastic beaker (250, 500 mL)
Funnel
Round bottom flask (250 mL)
Condenser
4.4 Instruments
1. HPLC
System: Younglin
Pump: Solvent delivery modal LC-10ATVP
Column: Varian C-18 (250 4.6 mm i.d, 5 m particle size)
Injector: Microliter syringe (Rheodyne)
Ganpat University

78

Chapter 4

Chemicals, glasswares and instruments

Detector: Dual wavelength UV Detector


Software: Autochro-3000
2. HPTLC
System: CAMAG
Sample applicator: Linomat-V
Injector: Hamilton syringe (100l)
Detection: CAMAG TLC Scanner 3
Developing chamber: CAMAG TLC chamber (20x10cm, 20x20cm)
Operation software: WinCATs
3. UV-Visible spectrophotometer
System: ELICO SL 210
Spectral range: 190 to 1100nm
Bandwidth: 1.8nm
Stray light: <0.05%T at 220nm with Nal 10g/L
Light source: Deuterium Lamp (D2) & Tungsten (W) Lamp
Monochromator: Concave holographic grating with 1200lines/mm
Detector: Photo Diode
Control: Microprocessor and Microcontroller (Computer-Optional)
4.5 Other requirements
1. Ultrasonic cleaner
Model-D compact 1.5 litre (EIE Instruments Pvt. Ltd.)
2. Digital analytical balance
Electronic Balance BL-220H Shimadzu Corporation Japan (1mg sensitivity)
3. pH meter (digital)
LI127 Elico Limited

4. FIlter
Ultipore N66 Nylon membrane (Pall India Pvt. Ltd.) (0.45m and 0.2m)

Ganpat University

79

Chapter 4

Chemicals, glasswares and instruments

4.6 References
1. Indian Pharmacopeia, Government of India ministry of health and family welfare,
1996, Vol-II, published by the controller of publications, Delhi, Appendix 1.5, A8.
2. Indian Pharmacopeia, Government of India ministry of health and family welfare,
1996, Vol-II, published by the controller of publications, Delhi, Appendix 12.5,
A-138.

Ganpat University

80

Chapter 5

Stability indicating methods for muscle relaxant drugs

5. STABILITY INDICATING METHOD DEVELOPMENT AND


VALIDATION
DICLOFENAC

FOR

SIMULTANEOUS

POTASSIUM,

ESTIMATION

CHLORZOXAZONE

OF
AND

PARACETAMOL
5.1 STABILITY INDICATING HPLC METHOD DEVELOPMENT AND
VALIDATION FOR SIMULTANEOUS ESTIMATION OF DICLOFENAC
POTASSIUM, CHLORZOXAZONE AND PARACETAMOL
5.1.1 Experimental
5.1.1.1 Solubility
The Solubility of Diclofenac potassium (DIC), Chlorzoxazone (CHL) and Paracetamol
(PCM) were performed in different solvent like distilled water, 0.1N HCl, 0.1N NaOH,
methanol, dimethylformamide, acetonitrile, ethanol and chloroform. All three drugs were
found to be soluble in methanol.
5.1.1.2 Preparation of mobile phase

The mobile phase methanol:phosphate buffer pH 4.0 in the ratio of 70:30, v/v
respectively was used. The pH was adjusted with glacial acetic acid. The mobile phase
was filtered through 0.45 filter paper to remove particulate matter and then degassed by
sonication.
5.1.1.3 Preparation of standard solutions
5.1.1.3.1 Preparation of standard stock solutions
5.1.1.3.1.1 Preparation of standard stock solution of DIC
Accurately weighed DIC (50mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 500g/ml stock solution.
5.1.1.3.1.2 Preparation of standard stock solution of CHL
Accurately weighed CHL (250mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 2500g/ml stock solution.

Ganpat University

81

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.1.1.3.1.3 Preparation of standard stock solution of PCM


Accurately weighed PCM (32.5mg) was transferred in 10ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 3250g/ml stock solution.
5.1.1.3.1.2 Preparation of standard working solutions
5.1.1.3.1.2.1 Preparation of standard working solution of DIC
From the stock solution (500g/ml), an aliquot quantity 1, 2, 3, 4, 5 and 6 ml were
transferred into separate 100ml volumetric flask and final volume was adjusted with
methanol upto mark to prepare 5-30g/ml solutions.
5.1.1.3.2.2 Preparation of standard working solution of CHL

From stock solution (2500g/ml), an aliquot quantity 1, 1.8, 2.6, 3.4, 4.2 and 5.0 ml were
transferred into 100 ml volumetric flask and final volume was adjusted with methanol
upto mark to prepare 25-125g/ml solutions.
5.1.1.3.2.3 Preparation of standard working solution of PCM

From stock solution (3250g/ml), an aliquot quantity 1, 1.9, 2.8, 3.8, 4.7 and 5.6 ml were
transferred into 100 ml volumetric flask and final volume was adjusted with methanol
upto mark to prepare 32.5-182.5 g/ml solutions.
5.1.1.4 Preparation of sample solution

Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50 mg of DIC, 250 mg of CHL and 325 mg of PCM was transferred to a 100 ml
volumetric flask. The powder was dissolved in 60 ml of methanol with sonication for 15
minutes and volume was made up with methanol. 1ml of the solution was transferred into
10ml volumetric flask and diluted upto mark with methanol.
5.1.1.5 Determination of wavelength maxima

Standard solutions of DIC, CHL and PCM were scanned between 200 and 400nm. UV
spectra of all three drugs show maximum absorbance at 280nm. (Figure 5.1.1)

Ganpat University

82

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.1 Overlay UV spectra of DIC, CHL and PCM


5.1.1.6 Chromatographic conditions
The chromatographic separations were performed using Varian C-18 (250 4.6 mm i.d,
5 m particle size) column at room temperature. The optimum mobile phase consisted of
methanol:phosphate buffer pH 4.0 (pH adjusted with glacial acetic acid) in the ratio of
70:30, v/v respectively. A 20 l sample was injected for analysis. The analysis was done
with flow rate of 0.8 ml/min at 280 nm wavelength using Dual wavelength UV Detector.
Method validation[1-2]
5.1.1.7.1 Linearity and range
The calibration curve was plotted over the concentration range of 5-30g/ml for DIC, 25125g/ml for CHL and 32.5-182.5g/ml for PCM. All the solution were filtered through
0.2m membrane filter and injected, chromatograms were recorded and it was repeated
for six times. A calibration graph was plotted between the mean peak area Vs respective
concentration and regression equation was derived.
5.1.1.7.2 Accuracy
The accuracy of the method was determined by calculating recoveries of DIC, CHL and
PCM by standard addition method. Known amount of standard solution of DIC, CHL and
PCM (80, 100 and 120% level) were added to pre-analysed samples.
Ganpat University

83

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.1.1.7.3 Precision
Precision was evaluated in terms of intraday and interday precision. The intraday
precision was investigated using different concentrations of sample solutions in
triplicates. The intraday and interday precision of the proposed method was determined
by analyzing the corresponding concentration three times on the same day and on
different days respectively.
5.1.1.7.4 Robustness
Robustness was determined by the analysis of the samples under a variety of conditions
making small changes in the buffer pH, in the ratio of mobile phase, in the flow rate, in
the temperature conditions and changing the wavelength.
5.1.1.7.5 Limit of detection and quantification
The LOD may be expressed as: DL =3.3 / S
Where = the standard deviation of the response, S = the slope of the calibration curve
The LOQ may be expressed as: QL =10 / S
Where = the standard deviation of the response, S = the slope of the calibration curve
5.1.1.7.6 System suitability
The system suitability parameters like theoretical plates (N), resolution (Rs), retention
time (RT) and tailing factor (Tf) reported in European Pharmacopoeia[2] were calculated
by LC solution software. The HPLC system was equilibrated with the initial mobile
phase composition, followed by six injections of same standard.
5.1.1.8 Analysis of marketed formulation
The response of sample solution was measured under chromatographic condition as
described above in section 5.1.1.6. The amount of DIC, CHL and PCM were calculated
by regression equation.
5.1.1.9 Forced degradation study of drug substance [3]
5.1.1.9.1 Acidic degradation
5.1.1.9.1.1 Acidic degradation of DIC
Accurately weighed (10mg) of DIC was, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N HCl was added and refluxed at 85C for 24
hours. 0.5ml of the solution was diluted upto 10ml with methanol (5g/ml) and analysed
by HPLC.
Ganpat University

84

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.1.1.9.1.2 Acidic degradation of CHL


Accurately weighed CHL (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. 2.5 ml of the solution was diluted upto 10ml with methanol (25g/ml) and
analysed by HPLC.
5.1.1.9.1.2 Acidic degradation of PCM
Accurately weighed PCM (10mg) was transferred into 100ml volumetric flask and
dissolved in 5ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. 3.25ml of the solution was diluted upto 10ml with methanol (32.5g/ml) was
analysed by HPLC.
5.1.1.9.2 Alkaline degradation
5.1.1.9.2.1 Alkaline degradation of DIC
Accurately weighed DIC (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at 80C for 6
hours. 0.5ml of the solution was diluted upto 10ml with methanol (5g/ml) and analysed
by HPLC.
5.1.1.9.2.2 Alkaline degradation of CHL
Accurately weighed CHL (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1 N NaOH was added and refluxed at 80C for 6
hours. 2.5ml of the solution was diluted upto 10ml with methanol (25g/ml) and analysed
by HPLC.
5.1.1.9.2.3 Alkaline degradation of PCM
Accurately weighed PCM (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50 ml of 5N NaOH was added and refluxed at 80C for 6
hours. 3.25ml of the solution was diluted upto 10ml with methanol. The solution
(32.5g/ml) was analysed by HPLC.
5.1.1.9.3 Oxidative degradation
5.1.1.9.3.1 Oxidative degradation of DIC
Accurately weighed DIC (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature

Ganpat University

85

Chapter 5

Stability indicating methods for muscle relaxant drugs

for 6 hours. 0.5ml of the solution was diluted upto 10ml with methanol (5g/ml) and
analysed by HPLC.
5.1.1.9.3.2 Oxidative degradation of CHL
Accurately weighed CHL (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 6 hours. 2.5ml of the solution was diluted upto 10ml with methanol (25g/ml) and
analysed by HPLC.
5.1.1.9.2.3 Oxidative degradation of PCM
Accurately weighed PCM (10mg) was, transferred into 10ml volumetric flask and
dissolved in 5ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 6 hours. 3.25ml of the solution was diluted upto 10ml with methanol (32.5g/ml) and
analysed by HPLC.
5.1.1.9.4 Thermal degradation
5.1.1.9.4.1 Thermal degradation of DIC
Accurately weighed DIC (10mg) was kept at 105C for 8 hours and transferred in 100ml
volumetric flask. Dissolved in 50ml of methanol then dilute upto mark with methanol. 0.5
ml of the solution was diluted upto 10ml with methanol (5g/ml) and analysed by HPLC.
5.1.1.9.4.2 Thermal degradation of CHL
Accurately weighed CHL (10mg) was kept at 105C for 8 hours and transferred in 100ml
volumetric flask. Dissolved in 50ml of methanol then dilute upto mark with methanol. 2.5
ml of the solution was diluted upto 10ml with methanol (25g/ml) and analysed by
HPLC.
5.1.1.9.4.3 Thermal degradation of PCM
Accurately weighed PCM (10mg) was kept at 105C for 8 hours and transferred in 10ml
volumetric flask. Dissolved in 5ml of methanol then dilute upto mark with methanol
(32.5g/ml) and analysed by HPLC.
5.1.1.9.5 Neutral degradation
5.1.1.9.5.1 Neutral degradation of DIC
Accurately weighed DIC (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and refluxed at 85C for 24 hours.

Ganpat University

86

Chapter 5

Stability indicating methods for muscle relaxant drugs

0.5ml of the solution was diluted upto 10ml with methanol (5g/ml) and analysed by
HPLC.
5.1.1.9.5.2 Neutral degradation of CHL
Accurately weighed CHL (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and refluxed at 85C for 24 hours.
2.5ml of the solution was diluted upto 10ml with methanol (25g/ml) and analysed by
HPLC.
5.1.1.9.5.3 Neutral degradation of PCM
Accurately weighed PCM (10mg) was transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hours.
The solution (32.5g/ml) was analysed by HPLC.
5.1.1.10 Forced degradation study of marketed product
5.1.1.10.1 Acidic degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 250mg of CHL and 325mg of PCM were transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 0.1N HCl was added and
refluxed at 85C for 24 hours. 1ml of the solution was diluted upto 100ml with methanol
and analysed by HPLC.
5.1.1.10.2 Alkaline degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 250mg of CHL and 325mg of PCM was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and
refluxed at 80C for 6 hours. 1ml of the solution was diluted upto 100ml with methanol
and analysed by HPLC.
5.1.1.10.3 Oxidative degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 250mg of CHL and 325mg of PCM was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept
at room temperature for 6 hours. 1ml of the solution was diluted upto 100ml with
methanol and analysed by HPLC.

Ganpat University

87

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.1.1.10.4 Thermal degradation of marketed product


Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 250mg of CHL and 325mg of PCM was kept at 105C for 8 hours and
transferred in 100ml volumetric flask. The powder was dissolved in 50ml of methanol
and diluted upto mark with methanol. 1ml of the solution was diluted upto 100ml with
methanol and analysed by HPLC.
5.1.1.10.5 Neutral degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 250mg of CHL and 325mg of PCM was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of water was added and refluxed
at 85C for 24 hours. 1ml of the solution was diluted upto 100ml with methanol and
analysed by HPLC.
5.1.2 Results and discussion
5.1.2.1 Method optimization
The aim of this study was to develop a stability indicating HPLC method for
simultaneous analysis of DIC, CHL and PCM. The review of literature for estimation of
other muscle relaxant drugs either alone or in combination and knowledge of molecule
suggest that reverse phase liquid chromatography (RP-LC) is suitable for simultaneous
analysis of DIC, CHL and PCM. In RP-HPLC various columns are available but C18
column was preferred over other columns. To optimize mobile phase initially methanol
and water in different ratios were tried. But CHL gave broad peak shape, so water was
replaced by phosphate buffer pH 4.0, and mixture of methanol and phosphate buffer pH
4.0 in different ratios were tried. It was found that methanol: phosphate buffer pH 4.0 in
ratio of 70:30 v/v respectively gave acceptable retention time (t R 3.25min for DIC, tR 5.60
min for CHL and tR 9.40 min for PCM) and good resolution for DIC, CHL and PCM with
flow rate of 0.8 ml/min at 280 nm (Table 5.1.1a). The method parameters were optimized
to analyse DIC, CHL and PCM in marketed product. A chromatogram has been shown in
Figure 5.1.2a and 5.1.2b.

Ganpat University

88

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.1.1a Results of optimization of mobile phase


Mobile phase

Ratio

Result

Methanol: water

50:50

Poor elution of drugs

Methanol: water

80:20

No good shape of drug and


poor resolution of peaks

Methanol: phosphate buffer pH 4.0

50:50

High retention time with bad


shape

Methanol: phosphate buffer pH 4.0

70:30

Less retention time with good


shape and better separation

Figure 5.1.2a: Blank Chromatogram

Ganpat University

89

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.2b: Chromatogram of DIC, CHL and PCM by HPLC


5.1.2.2 Method validation
Linearity and range
The calibration curve was plotted over the concentration range of 5-30g/ml for DIC, 25125g/ml for CHL and 32.5-182.5 g/ml for PCM. A calibration graph was plotted
between the mean peak area Vs respective concentration and regression equation was
derived (Figure 5.1.3, 5.1.4, 5.1.5). The results were shown in Table 5.1.1.
160000

P 140000
e 120000
a
100000
k

y = 5018.1x + 362
R = 0.9998

80000

a 60000
r 40000
e 20000
a

0
0

10

15

20

Concentration (g/ml)

25

30

35

Figure 5.1.3: Calibration curve of DIC by HPLC

Ganpat University

90

Chapter 5

Stability indicating methods for muscle relaxant drugs


700000

P
e
a
k

600000
500000
y = 4987.5x - 20588
R = 0.9997

400000
300000

a
r
e
a

200000
100000
0
0

20

40

60

80

100

120

140

Concentration (g/ml)
Figure 5.1.4: Calibration curve of CHL by HPLC

P 1000000
e 900000
a 800000
700000
k 600000
a
r
e
a

y = 5051.4x - 14453
R = 0.9997

500000
400000
300000
200000
100000
0
0

50

100

150

200

Concentration (g/ml)
Figure 5.1.5: Calibration curve of PCM by HPLC
Table 5.1.1b Linearity and range data for DIC, CHL and PCM by HPLC
Drug

Linearity range

DIC
5-30 g/ml
CHL
25-125 g/ml
PCM
32.5-182.5 g/ml
*= Average result of six replicate samples

Y=mx+c
Slope*
Intercept*
5018.1
362
4987.5
2058
5051.4
14445

r2*
0.9998
0.9997
0.9997

5.1.2.2.2 Accuracy (Recovery study)


The recovery for DIC, CHL and PCM were found between 98% and 101%. The results
were shown in Table 5.1.2.

Ganpat University

91

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.1.2 Recovery study of DIC, CHL and PCM by HPLC


Drug

DIC

Conc. Of
Form.
(g/ml)
5

5
5
25
CHL
25
25
32.5
PCM
32.5
32.5
*= Average result of six replicate samples

Conc. Of
Std. added
(g/ml)
4

Conc.
Recovered
(g/ml) SD*
8.890.12

5
6
24
25
26
34
32.5
36

9.970.30
10.920.02
48.490.07
48.900.71
50.580.05
65.541.51
65.280.08
69.070.16

% recoverySD*
98.78%0.18
99.70%0.75
99.27%1.89
98.96%0.01
98.16%0.18
99.17%1.09
98.56%0.86
100.41%0.15
100.83%0.11

5.1.2.2.3 Precision
The %RSD of intraday and inter-day precision study for DIC, CHL and PCM were found
to be <2. The results are shown in Table 5.1.3.
Table 5.1.3 Intra-day & Inter-day precision of DIC, CHL and PCM by HPLC

Intra day
Mean %assay*
%RSD*
DIC
99.34%1.16
0.36
CHL
99.83%1.63
0.12
PCM
98.94%0.01
1.16
*= Average result of six replicate samples
Drug

Inter day
Mean %assay*
%RSD*
99.15%1.82
1.08
100.05%0.57
1.12
98.86%0.12
1.52

5.1.2.2.3 Robustness
To determine the robustness of the developed method, experimental conditions were
deliberately altered and the responses of all drugs were recorded. The results of change in
ratio of mobile phase, flow rate and wavelength are shown in Table 5.1.4.

Ganpat University

92

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.1.4 Robustness data for DIC, CHL and PCM by HPLC
Chromat
ographic
factor

level
0.7ml/
min
1.0ml/
min

Retention time*
DIC

CHL

6.010.0
1
Flow rate
5.040.0
2.950.02
1
6.020.0
Methanol:
75:25 3.910.03
3
phosphate
buffer
5.050.0
85:15 3.960.02
(v/v)
2
5.600.0
Detection 275nm 3.240.04
1
wave5.600.0
length
285nm 3.260.03
1
*= Average result of six replicate samples
3.980.02

Tailing factor*
PCM

DIC

9.960.0
1
8.660.0
3
9.950.0
2

0.950.
02
0.960.
01
0.950.
02
0.960.
01
0.950.
01
0.950.
02

8.61.02
9.410.0
1
8.640.0
2

CHL
1.020.01
1.030.01
1.020.01
1.020.01
1.030.01
1.020.01

PCM
0.970.0
1
0.960.0
1
0.960.0
2
0.970.0
1
0.970.0
3
0.970.0
1

5.1.2.2.4 Limit of detection and quantification


The LOD for DIC, CHL & PCM were found to be 0.15g/ml, 1.82g/ml and 2.40g/ml
respectively, while LOQ were 0.47g/ml, 5.53g/ml and 7.29g/ml respectively.
5.1.2.2.5 System suitability
The parameters like retention time, asymmetric factor, number of theoretical plates and
tailing factors were evaluated for DIC, CHL & PCM. The results were shown in Table
5.1.5.
Table 5.1.5 System suitability parameters for DIC, CHL and PCM by HPLC
No. of
Parameter

RT*

AUC*

theoretical
plates*

DIC
3.250.05
25012.20283.43
4204.49.52
CHL
5.600.06
105410.2312.53 3637.4139.80
PCM
9.400.09 150453.123923.11 1263.127.005
*=Average result of six replicate samples

Ganpat University

Tailing
factor*
0.95.002
1.020.01
0.970.001

93

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.1.2.3 Analysis of marketed product


Market formulation containing 50mg DIC, 250mg CHL and 325mg PCM was analysed
by HPLC.
5.1.2.4 Forced degradation study
Forced degradation studies were performed for bulk drug and marketed product, to
provide an indication of the stability indicating property. The degradation was attempted
to stress conditions like acid hydrolysis, alkaline hydrolysis, oxidative hydrolysis,
thermal treatment and neutral degradation, in order to evaluate the ability of the proposed
method to separate drug from its degradation products [3]. During forced degradation
experiments, DIC showed more degradation under oxidative hydrolysis compared to
other conditions. CHL was degraded in alkaline, oxidative and thermal stress conditions
whereas PCM was degraded only under oxidative stress conditions. Table 5.1.7 indicates
the extent of degradation of marketed product under various stress conditions. Figures
5.1.6 to 5.1.15 shows the chromatograms of forced degraded samples. The degradation
products were well resolved from drug, confirming the stability indicating power of the
method.

Figure 5.1.6a: Chromatogram of acidic degradation of DIC by HPLC

Ganpat University

94

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.6b: Chromatogram of acidic degradation of CHL by HPLC

Figure 5.1.6c: Chromatogram of acidic degradation of PCM by HPLC

Figure 5.1.7a: Chromatogram of alkaline degradation of DIC by HPLC

Ganpat University

95

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.7b: Chromatogram of alkaline degradation of CHL by HPLC

Figure 5.1.7c: Chromatogram of alkaline degradation of PCM by HPLC

Figure 5.1.8a: Chromatogram of oxidative degradation of DIC by HPLC


Ganpat University

96

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.8b: Chromatogram of oxidative degradation of CHL by HPLC

Figure 5.1.8c: Chromatogram of oxidative degradation of PCM by HPLC

Figure 5.1.9a: Chromatogram of thermal degradation of DIC by HPLC

Ganpat University

97

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.9b: Chromatogram of thermal degradation of CHL by HPLC

Figure 5.1.9c: Chromatogram of thermal degradation of PCM by HPLC

Figure 5.1.10a: Chromatogram of neutral degradation of DIC by HPLC

Ganpat University

98

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.10b: Chromatogram of neutral degradation of CHL by HPLC

Figure 5.1.10c: Chromatogram of neutral degradation of PCM by HPLC

Figure 5.1.11: Chromatogram of acidic degradation of marketed product by HPLC


Ganpat University

99

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.12: Chromatogram of alkaline degradation of marketed product by


HPLC

Figure 5.1.13: Chromatogram of oxidative degradation of marketed product by


HPLC

Figure 5.1.14: Chromatogram of thermal degradation of marketed product by


HPLC
Ganpat University

100

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.1.15: Chromatogram of neutral degradation of marketed product by


HPLC

Ganpat University

101

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.1.6 Forced degradation study of marketed product by HPLC


Degradatio
n condition

Acidic

Drug

Conc.
Of
drug(
g/ml)

RT of
observed
peak*

AUC*

%drug*

%degradation
*

DIC

3.25

24018

98.14%1.04

0.00%0.00

5.59

101541

91.98%1.31

0.00%0.00

5.19(I)

8931

0.00%0.00

8.791.31

9.40
3.25

149901
24010

99.65%1.45
94.62%1.24

0.00%0.00
0.00%0.00

2.92(I)

1432

0.00%0.00

5.96%1.21

5.62
5.49(II)
6.12(III)
9.41
3.25

94421
3410
1431
149097
23120

91.56%1.57
0.00%0.00
0.00%0.00
99.57%1.34
91.41%1.32

94.10%1.38
3.61%1.41
1.511.30
0.00%0.00
0.00%0.00

2.72(I)

1981

0.00%0.00

8.98%1.54

5.60
5.12(II)
9.41

102384
2398
148320

97.00%1.03
0.00%0.00
93.84%1.09

0.00%0.00
2.64%0.95
0.00%0.00

10.21(III)

9908

0.00%0.00

6.681.41

CHL

25

PCM

32.5

DIC

Alkaline
CHL

Oxidative

Thermal

25

PCM

32.5

DIC

CHL

25

PCM

32.5

DIC
CHL
PCM

5
25
32.5

3.25
5.61
9.41

25320
105908
149959

99.90%1.12
99.88%1.08
99.84%1.24

0.00%0.00
0.00%0.00
0.00%0.00

DIC

3.25

24906

98.45%1.4

0.00%0.00

103956
9327
104903

91.88%1.23
0.00%0.00
99.84%1.21

0.00%0.00
8.64%0.95
0.00%0.00

Neutral

5.60
4.93(I)
PCM
32.5
9.41
*= Average result of six replicate samples
CHL

Ganpat University

25

102

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.1.3 Conclusion
The RP-HPLC method developed for the analysis of ternary mixtures of DIC, CHL and
PCM as bulk drug and in pharmaceutical preparation is simple, accurate, precise, and
repeatable with short run time. The developed method is stability indicating and can
separate degradants and be used to determine the assay of pharmaceutical preparation and
also stability samples.

Ganpat University

103

Chapter 5

5.2

STABILITY

Stability indicating methods for muscle relaxant drugs

INDICATING

HPTLC

METHOD

FOR

SIMULTANEOUS ESTIMATION OF DICLOFENAC POTASSIUM,


CHLORZOXAZONE AND PARACETAMOL
5.2.1 Experimental
5.2.1.1 Solubility
Solubility of Diclofenac potassium(DIC), Chlorzoxazone(CHL) and Paracetamol(PCM)
were performed in different solvent like distilled water, 0.1N HCl, 0.1N NaOH,
methanol, dimethyl formamide, acetonitrile, ethanol and chloroform. All three drugs were
found to be soluble in methanol.
5.2.1.2 Preparation of mobile phase
The mobile phase Toluene: ethylacetate: glacial acetic acid in the ratio of 1:2:0.5,v/v/v
respectively was used.
5.2.1.3 Preparation of standard solutions
5.2.1.3.1 Preparation of standard stock solutions
5.2.1.3.1.1 Preparation of standard stock solution of DIC
Accurately weighed DIC (10mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 100g/ml stock solution.
5.2.1.3.1.2 Preparation of standard stock solution of CHL
Accurately weighed CHL (10mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 100g/ml stock solution.
5.2.1.3.1.3 Preparation of standard stock solution of PCM
Accurately weighed PCM (10mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 100g/ml stock solution.
5.2.1.4 Preparation of sample solution
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 250mg of CHL and 325mg of PCM were transferred to a 100ml
volumetric flask. The powder was dissolved in 50ml of methanol with sonication for 15
minutes and volume was made up with methanol.
Ganpat University

104

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.2.1.5 Determination of wavelength maxima


Standard solutions of DIC, CHL and PCM were scanned between 200 and 400nm. UV
spectra of all three drugs show maximum absorbance at 280nm.

Figure 5.2.1 Overlay UV spectra of DIC, CHL and PCM


5.2.1.6 Chromatographic conditions
Chromatography was performed on 10x10cm Precoated Silica Gel G60F254 aluminium
sheets. The samples were applied to the plates as 6mm band, 10 mm apart, by means of
Linomat-V sample applicator with help of 100l Hamilton syringe. It was developed in
CAMAG TLC chamber (20x10cm, 20x20cm) which was already saturated for 30 min. with
mobile phase at room temperature. The optimum mobile phase consisted of Toluene: ethyl

acetate: glacial acetic acid in the ratio of 1:2:0.5, v/v/v respectively. After development the
plate was scanned at 280 nm by means of CAMAG TLC Scanner 3 controlled by WinCATs
software.

5.2.1.7 Method validation [1-2]


5.2.1.7.1 Linearity and range
The calibration curve was plotted over the concentration range of 10-60ng/spot for DIC,
50-300ng/spot for CHL and 100-350ng/spot for PCM. All the solution was spotted on
Ganpat University

105

Chapter 5

Stability indicating methods for muscle relaxant drugs

precoated TLC plate, chromatograms were recorded and it was repeated for six times. A
calibration graph was plotted between the mean peak height Vs respective concentration
and regression equation was derived.
5.2.1.7.2 Accuracy
The accuracy of the method was determined by calculating recoveries of DIC, CHL and
PCM by standard addition method. Known amount of standard solution of DIC, CHL and
PCM (80, 100 and 120% level) were added to preanalysed samples.
5.2.1.7.3 Precision
Precision was evaluated in terms of intraday and interday precision. The intraday
precision was investigated using different concentrations of sample solutions in triplicate.
The intraday and interday precision of the proposed method was determined by analyzing
the corresponding concentration three times on the same day and on different days.
5.2.1.7.4 Robustness
Robustness was determined by the analysis of the samples under a variety of conditions
making small changes in the ratio of mobile phase, in the saturation time, changing the
wavelength.
5.2.1.7.5 Limit of detection and quantification
The LOD may be expressed as: DL =3.3 / S
Where = the standard deviation of the response, S = the slope of the calibration curve
The LOQ may be expressed as: QL =10 / S
Where = the standard deviation of the response, S = the slope of the calibration curve
5.2.1.8 Analysis of marketed formulation
8, 10 and 12l of the sample solution as described in 5.2.1.4 were spotted on precoated
TLC plate. 1ml of sample solution (5.2.1.4) was diluted upto 10ml with methanol and 8,
10 and 12l of this solution were spotted on precoated TLC plate. 1ml of above solution
diluted upto 10ml with methanol and 8, 10 and 12l of this solution were spotted on
precoated TLC plate. The TLC plate was developed and analysed as described under
chromatographic condition. The amount of DIC, CHL and PCM were determined by
regression equation.

Ganpat University

106

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.2.1.9 Forced degradation study of drug substance [3]


5.2.1.9.1 Acidic degradation
5.2.1.9.1.1 Acidic degradation of DIC
50mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N HCl was added and refluxed at 85C for 24
hours. 1.0ml of the solution is diluted upto 10ml with methanol then 1.0l of the solution
was spotted on precoated TLC plate (50ng/spot), developed and analysed as described
under chromatographic condition.
5.2.1.9.1.2 Acidic degradation of CHL
25mg of CHL was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. 1ml of the solution diluted upto 10ml with methanol then 8l of the solution was
spotted on precoated TLC plate (250ng/spot), developed and analysed as described under
chromatographic condition.
5.2.1.9.1.3 Acidic degradation of PCM
32.5mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. 1ml of the solution diluted upto 10ml with methanol then 1.0l of the solution was
spotted on precoated TLC plate (325ng/spot), developed and analysed as described under
chromatographic condition.
5.2.1.9.2 Alkaline degradation
5.2.1.9.2.1 Alkaline degradation of DIC
50mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at 80C for 6
hours. 1ml of the solution diluted upto 10ml with methanol then 1.0l of the solution was
spotted on precoated TLC plate (50ng/spot), developed and analysed as described under
chromatographic condition.
5.2.1.9.2.2 Alkaline degradation of CHL
25mg of CHL was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at 80C for 6

Ganpat University

107

Chapter 5

Stability indicating methods for muscle relaxant drugs

hours. 1.0l of the solution was spotted on precoated TLC plate (250ng/spot), developed
and analysed as described under chromatographic condition.
5.2.1.9.2.3 Alkaline degradation of PCM
32.5mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N NaOH was added and refluxed at 80C for 6
hours. 1.0l of the solution was spotted on precoated TLC plate (325ng/spot), developed
and analysed as described under chromatographic condition.
5.2.1.9.3 Oxidative degradation
5.2.1.9.3.1 Oxidative degradation of DIC
50mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 6 hours. 1ml of the solution diluted upto 10ml with methanol. 1.0l of the solution
was spotted on precoated TLC plate (50ng/spot), developed and analysed as described
under chromatographic condition.
5.2.1.9.3.2 Oxidative degradation of CHL
25mg of CHL was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 6 hours. 1.0l of the solution was spotted on precoated TLC plate (250ng/spot),
developed and analysed as described under chromatographic condition.
5.2.1.9.3.3 Oxidative degradation of PCM
32.5mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 6 hours. 1.0l of the solution was spotted on precoated TLC plate (325ng/spot),
developed and analysed as described under chromatographic condition.
5.2.1.9.4 Thermal degradation
5.2.1.9.4.1 Thermal degradation of DIC
50mg of DIC was weighed accurately, kept at 105C for 8 hours and transferred into
100ml volumetric flask. The powder was dissolved in 50ml methanol and diluted upto
mark with methanol. 1ml of the solution diluted upto 10ml with methanol then 1.0l of
the solution was spotted on precoated TLC plate (50ng/spot), developed and analysed as
described under chromatographic condition.
Ganpat University

108

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.2.1.9.4.2 Thermal degradation of CHL


25mg of CHL was weighed accurately, kept at 105C for 8 hours and transferred into
100ml volumetric flask. The powder was dissolved in 50ml methanol and diluted upto
mark with methanol. 8l of the solution was spotted on precoated TLC plate
(250ng/spot), developed and analysed as described under chromatographic condition.
5.2.1.9.4.3 Thermal degradation of PCM
32.5mg of PCM was weighed accurately, kept at 105C for 8 hours and transferred into
100ml volumetric flask. The powder was dissolved in 50ml methanol and diluted upto
mark with methanol. 1.0l of the solution was spotted on precoated TLC plate
(325ng/spot), developed and analysed as described under chromatographic condition.
5.2.1.9.5 Neutral degradation
5.2.1.9.5.1 Neutral degradation of DIC
50mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and refluxed at 85C for 24 hours.
1ml of the solution diluted upto 10ml with methanol then 1.0l of the solution was
spotted on precoated TLC plate (50ng/spot), developed and analysed as described under
chromatographic condition.
5.2.1.9.5.2 Neutral degradation of CHL
25mg of CHL was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hours.
1.0l of the solution was spotted on precoated TLC plate (250ng/spot), developed and
analysed as described under chromatographic condition.
5.2.1.9.5.3 Neutral degradation of PCM
32.5mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hours.
1.0l of the solution was spotted on precoated TLC plate (325ng/spot), developed and
analysed as described under chromatographic condition.

Ganpat University

109

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.2.1.10 Forced degradation study of marketed product


5.2.1.10.1 Acidic degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 25mg of CHL and 32.5mg of PCM was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 0.1N HCl was added and
refluxed at 85C for 24 hours. 1.0ml of the solution diluted upto 10ml with methanol then
1.0l of the solution was spotted on precoated TLC plate (50ng/spot DIC), developed and
analysed as described under chromatographic condition. Moreover, 1.0l of the solution
was spotted on precoated TLC plate (250ng/spot CHL and 325ng/spot PCM), developed
and analysed as described under chromatographic condition.
5.2.1.10.2 Alkaline degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 25mg of CHL and 32.5mg of PCM was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and
refluxed at 80C for 6 hours. 1.0ml of the solution diluted upto 10ml with methanol then
1.0l of the solution was spotted on precoated TLC plate (50ng/spot DIC), developed and
analysed as described under chromatographic condition. Moreover, 1.0l of the solution
was spotted on precoated TLC plate (250ng/spot CHL and 325ng/spot PCM), developed
and analysed as described under chromatographic condition.
5.2.1.10.3 Oxidative degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 25mg of CHL and 32.5mg of PCM was transferred to a 100 ml
volumetric flask and dissolved in 50ml methanol. 50ml of 3% H 2O2 was added and kept
at room temperature for 6 hours. 1.0ml of the solution diluted upto 10ml with methanol
then 1.0l of the solution was spotted on precoated TLC plate (50ng/spot DIC),
developed and analysed as described under chromatographic condition. Moreover, 1.0l
of the solution was spotted on precoated TLC plate (250ng/spot CHL and 325ng/spot
PCM), developed and analysed as described under chromatographic condition.
5.2.1.10.4 Thermal degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 25mg of CHL and 32,5mg of PCM was kept at 105C for 8 hours and
Ganpat University

110

Chapter 5

Stability indicating methods for muscle relaxant drugs

transferred in 100ml volumetric flask. The powder was dissolved in 50ml of methanol
and diluted upto mark with methanol. 1.0ml of the solution diluted upto 10ml with
methanol then 1.0l of the solution was spotted on precoated TLC plate (50ng/spot DIC),
developed and analysed as described under chromatographic condition. Moreover, 1.0l
of the solution was spotted on precoated TLC plate (250ng/spot CHL and 325ng/spot
PCM), developed and analysed as described under chromatographic condition.
5.2.1.10.5 Neutral degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 25mg of CHL and 32.5mg of PCM was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of water was added and refluxed
at 85C for 24 hours. 1.0ml of the solution diluted upto 10ml with methanol then 1.0l of
the solution was spotted on precoated TLC plate (50ng/spot DIC), developed and
analysed as described under chromatographic condition. Moreover, 1.0l of the solution
was spotted on precoated TLC plate (250ng/spot CHL and 325ng/spot PCM), developed
and analysed as described under chromatographic condition.

5.2.2 Results and discussion


5.2.2.1 Method optimization
For optimization, different mobile phases and composition were employed to achieve the
good separation. The method development was initiated with using a mobile phase of
toluene:ethyl acetate:ammonium sulphate in various proportions. In the above conditions
only two drugs were separated. All three drugs were separated using mobile phase
consisting of different ratio of glacial acetic acid. Finally mobile phase consisting mixture
of toluene: ethyl acetate: glacial acetic acid in ratio of 1:2:0.5,v/v/v respectively gave
reasonable resolution (Rf 0.78 for DIC, 0.31 for CHL and 0.53 for PCM) and sharp band
for all three drugs. Saturation of TLC chamber for 30 min assured better reproducibility
and better resolution. All three drugs were detected at 280 nm by means of CAMAG TLC
Scanner 3. A chromatogram is shown in Figure 5.2.2.

Ganpat University

111

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.2 Chromatogram of CHL, PCM and DIC


5.2.2.2 Method validation
5.2.2.2.1 Linearity and range
The calibration curve was plotted over the concentration range of 10-60ng/spot for DIC,
50-300ng/spot for CHL and 100-350ng/spot for PCM. A calibration graph was plotted
between the mean peak height Vs respective concentration and regression equation was
derived (Figure 5.2.3, 5.2.4, 5.2.5). The results were shown in Table 5.2.1.

Ganpat University

112

Chapter 5

Stability indicating methods for muscle relaxant drugs

7000

P
e
a
k

6000
5000
4000

y = 99.626x + 57.267
R = 0.9994

A
r 3000
e
a 2000
1000
0
0

10

20

30

40

50

60

70

Concentration (ng/spot)
Figure 5.2.3 Calibration curve for DIC by HPTLC

16000

P
e 14000
a 12000
k

y = 40.129x + 3023.7
R = 0.9999

10000

A
r
e
a

8000
6000
4000
2000
0
0

50

100

150

200

250

300

350

Concentration (ng/spot)
Figure 5.2.4 Calibration curve for CHL by HPTLC

Ganpat University

113

Chapter 5

Stability indicating methods for muscle relaxant drugs


20000

P 18000
e 16000
a
14000
k

y = 41.006x + 2900.1
R = 0.9997

12000

A 10000
r 8000
e 6000
a 4000
2000
0
0

100

200

300

400

Concentration (ng/spot)
Figure 5.2.5 Calibration curve for PCM by HPTLC
Table 5.2.1 Linearity and range data for DIC, CHL and PCM by HPTLC
Drug

Linearity range

DIC
10-60ng/spot
CHL
50-300ng/spot
PCM
100-350ng/spot
*= Average result of six replicate samples

Y=mx+c
Slope*
Intercept*
99.62
57.26
40.129
3023.7
41.006
2900.1

r2*
0.9994
0.9999
0.9997

5.2.2.2.2 Accuracy (Recovery study)


Recovery studies were performed to validate the accuracy of developed method. To a
pre-analysed sample solution, a definite concentration of standard drug was added and
recovery was studied. The recovery for DIC, CHL and PCM were found between 99%101 %.( Table 5.2.2).

Ganpat University

114

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.2.2 Recovery study of DIC, CHL and PCM by HPTLC


Conc. Of
Conc. Of
Form.
Std. added
(ng/spot)
(ng/spot)
50
40
DIC
50
60
50
50
250
240
CHL
250
250
250
260
325
315
PCM
325
325
325
335
*= Average result of six replicate samples
Drug

Conc.
Recover
(ng/spot)
89.96
108.95
100.02
489.15
497.11
508.32
638.40
648.83
661.47

% recoverySD*
99.96%0.021
99.04%0.032
100.02%0.019
99.82%0.012
99.42%0.01
99.68%0.04
99.75%0.030
99.82%0.018
100.22%0.036

5.2.2.2.3 Precision
The %RSD of intraday and interday precision study for DIC, CHL and PCM were found
to be <2. The results were shown in Table 5.2.3.
Table 5.2.3 Results of Intra-day & Inter-day precision DIC, CHL and PCM by
HPTLC
Intra day
Mean %assay*
%RSD*
DIC
99.98%0.02
0.012
CHL
98.97%0.41
0.014
PCM
99.10%0.04
0.020
*= Average result of six replicate sample
Drug

Inter day
Mean %assay*
%RSD*
97.96%0.048
0.038
98.98%0.047
0.040
99.19%0.01
0.01

5.2.2.2.3 Robustness
To determine the robustness of the developed method, experimental conditions were
deliberately altered and the responses of all drugs were recorded. The results of change in
ratio of mobile phase, flow rate and wavelength are shown in Table 5.2.4.

Ganpat University

115

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.2.4 Robustness data for DIC, CHL and PCM by HPTLC
Chromatographic
factor
Mobile phase ratio

level
1.2:1:4v/v/v
1:1.2:4v/v/v

Saturation time

Detection wavelength

32 minutes
28 minutes
275nm

285nm
*= Average result of six replicate samples

Amont found*(ng/spot)
DIC
CHL
PCM
199.820.03
39.920.051
249.980.086
2
199.840.02
39.900.062
249.160.034
8
199.970.04
39.940.060
249.970.047
3
199.930.04
39.980.062
249.990.037
2
199.950.04
39.950.038
248.950.058
3
39.960.022
201.000.02 250.100.024

5.2.2.2.4 Limit of detection and quantification


The LOD for DIC, CHL and PCM were found to be 0.005 ng/spot, 1.41 ng/spot and 3.17
ng/spot respectively, while LOQ were 0.87 ng/spot, 4.25 ng/spot and 9.61 ng/spot
respectively.
5.2.2.3 Analysis of marketed product
The market formulation containing 50mg DIC, 250mg CHL and 325mg PCM was
analysed by HPTLC.
5.2.2.4 Forced degradation study
The degradation was attempted to stress conditions like acid hydrolysis, alkaline
hydrolysis, oxidative hydrolysis, thermal treatment and neutral degradation, in order to
evaluate the ability of the proposed method to separate drug from its degradation
products[3]. HPTLC studies of the samples obtained during the stress testing of DIC, CHL
and PCM under different conditions using toluene: ethyl acetate: glacial acetic acid in the
ratio of 1:2:0.5, v/v/v respectively as the mobile phase shows different degradation peaks
as shown in Figures 5.2.6 to 5.2.15. CHL degraded under acidic, alkaline, oxidative and
neutral condition. Moreover, it showed highest degradation in alkaline condition and
degradation products appear at Rf of 0.19. DIC was degraded only in alkaline and
oxidative stress condition whereas PCM remain stable in all condition except oxidative

Ganpat University

116

Chapter 5

Stability indicating methods for muscle relaxant drugs

condition. Table 5.2.5 indicates the extent of degradation of marketed product under
various stress conditions.

Figure 5.2.6a: Chromatogram of acidic degradation of DIC by HPTLC

Figure 5.2.6b: Chromatogram of acidic degradation of CHL by HPTLC

Ganpat University

117

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.6c: Chromatogram of acidic degradation of PCM by HPTLC

Figure 5.2.7a: Chromatogram of alkaline degradation of DIC by HPTLC

Figure 5.2.7b: Chromatogram of alkaline degradation of CHL by HPTLC


Ganpat University

118

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.7c: Chromatogram of alkaline degradation of PCM by HPTLC

Figure 5.2.8a: Chromatogram of oxidative degradation of DIC by HPTLC

Figure 5.2.8b: Chromatogram of oxidative degradation of CHL by HPTLC


Ganpat University

119

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.8c: Chromatogram of oxidative degradation of PCM by HPTLC

Figure 5.2.9a: Chromatogram of thermal degradation of DIC by HPTLC

Figure 5.2.9b: Chromatogram of thermal degradation of CHL by HPTLC


Ganpat University

120

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.9c: Chromatogram of thermal degradation of PCM by HPTLC

Figure 5.2.10a: Chromatogram of neutral degradation of DIC by HPTLC

Figure 5.2.10b: Chromatogram of neutral degradation of CHL by HPTLC

Ganpat University

121

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.10c: Chromatogram of neutral degradation of PCM by HPTLC

Figure 5.2.11: Chromatogram of acidic degradation of marketed product by


HPTLC

Ganpat University

122

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.12: Chromatogram of alkaline degradation of marketed product


by HPTLC

Figure 5.2.13: Chromatogram of oxidative degradation of marketed product


by HPTLC

Figure 5.2.14: Chromatogram of thermal degradation of marketed product


by HPTLC
Ganpat University

123

Chapter 5

Stability indicating methods for muscle relaxant drugs

Figure 5.2.15: Chromatogram of neutral degradation of marketed product


by HPTLC

Ganpat University

124

Chapter 5

Stability indicating methods for muscle relaxant drugs

Table 5.2.5 Result of Forced degradation study of drug products


Degradatio
n condition

Conc.
of
Drug
drug(ng
/spot)
DIC

Acidic

50

Thermal

Peak
area*

%of drug*

0.73

2041.2

99.91%0.21

0.29

4694.94

91.89%1.01

0.21(I)

464.1

0.00%0.00

0.51
0.72

6392.9
2064.32

99.92%1.05
89.42%0.94

0.00%0.00
0.00%0.00

0.89(I)

210.6

0.00%0.00

10.17%0.91

0.29
0.19(II)
0.51
0.72

4487.52
208.39
6219.9
2138.71

95.27%0.77
0.00%0.00
99.89%0.34
95.39%1.22

0.00%0.00
4.63%0.29
0.00%0.00
0.00%0.00

0.84(I)

108.8

0.00%0.00

5.05%0.69

4599.6
196.45
5999.34
210.1
2014.56
4689.87
6382.98

96.00%1.03
0.00%0.00
96.95%0.19
0.00%0.00
99.93%0.82
99.87%1.08
99.92%0.24

0.00%0.00
4.26%0.96
0.00%0.00
3.50%0.96
0.00%0.00
0.00%0.00
0.00%0.00

1996.75

98.92%1.03

0.00%0.00

4499.12
291.3
6779.99

99.93%0.63
0.00%0.00
99.91%0.21

0.00%0.00
6.46%0.96
0.00%0.00

CHL

250

PCM

325

DIC

50

CHL

250

PCM

325

DIC

50

CHL

250

PCM

325

DIC
CHL
PCM

50
250
325

0.29
0.38(II)
0.32
0.24(III)
0.72
0.29
0.51

DIC

50

0.71

Alkaline

Oxidative

Rf value
of
observed
peak*

Neutral

0.29
0.21(I)
PCM
325
0.52
*= Average result of six replicate samples
CHL

Ganpat University

250

%of
degradation
*
0.00%0.00
0.00%0.00
9.880.91

125

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.2.3 Conclusion
The developed HPTLC technique is simple, accurate, sensitive, precise, rapid and
repeatable. The method was successfully used for determination of DIC, CHL, and PCM
as bulk drug and in pharmaceutical formulation. After exposing the drugs to different
stress condition, the drug peak area was observed to decrease and also degradants peak
were observed. The developed method is stability indicating and separate degradants and
can be used to determine the assay of pharmaceutical preparation and also stability
samples.

Ganpat University

126

Chapter 5

5.3

Stability indicating methods for muscle relaxant drugs

STATISTICAL

COMPARISON

BETWEEN

OFFICIAL

REPORTED AND PROPOSED METHODS[4-5]


The assay result for DIC, CHL and PCM in their dosage form obtained using HPLC and
HPTLC methods were compared between official reported methods by applying the
paired t-test and F-test. The calculated t-value for DIC (0.0011), CHL (0.030) and PCM
(0.0057) for HPLC as well as DIC (0.096), CHL (0.213), and PCM (0.1854) for HPTLC
is less than the tabulated t-value (2.571) at the 95% confidence interval. Moreover,
calculated F-value for DIC (0.0252), CHL (0.1711) and PCM (0.1741) for HPLC as well
as DIC (0.3567), CHL (0.011) and PCM (0.1854) for HPTLC is also less than the
tabulated F-value (4.28). Therefore, there is no significant difference in a determined
content of DIC, CHL and PCM by HPLC and HPTLC methods. Table 5.3.1 indicates
statistical comparison between proposed methods.
Table 5.3.1 Statistical comparison for DIC, CHL and PCM between proposed
methods
Paired t-test
Drug Methods Mean SD

n Tabulated Calculated Tabulated Calculated


value

DIC

CHL

PCM

Official

99.11

0.06 6

HPLC

99.64

0.19 6

Official

99.11

0.06 6

HPTLC

99.29

0.20 6

Official

99.78

0.27 6

HPLC

99.56

0.12 6

Official

99.78

0.27 6

HPTLC

99.55

0.07 6

Official

99.65

0.24 6

HPLC

99.25

0.14 6

Official

99.65

0.24 6

HPTLC

99.67

0.15 6

Ganpat University

F-test

value

value

0.0011
2.571

0.096

0.0252
4.28

0.030
2.571
0.213

2.571

0.1854

0.3567

0.1711
4.28

0.0057

value

4.28

0.011

0.1741
0.1854

127

Chapter 5

Stability indicating methods for muscle relaxant drugs

5.4 References
1. ICH, Q2(R1) Validation of Analytical Procedure: Text and Methodology,
International Conference on Harmonization, Geneva, October 1994.
2. The United States Pharmacopeia, The National Formulary, USP 30 NF 25, Asian
Edition, Volume 1, 2007; 680-683.
3. Bakshi M, Singh S, Development of validated stability-indicating assay methods
critical review, Journal of Pharmaceutical and Biomedical Analysis, 2002;
28:1010-40.
4. Christian GD, Analytical Chemistry, 6th Ed., University of Washington, 2007; 9097.
5. Sanford Bolton, Charles Bon, Pharmaceutical Statistics Practical and Clinical
Application, 4th Ed., Revised and Expanded, 2004; 417-436.

Ganpat University

128

Chapter 6

Stability indicating methods for muscle relaxant drugs

6. STABILITY INDICATING METHOD DEVELOPMENT AND


VALIDATION

FOR

DICLOFENAC

SIMULTANEOUS

POTASSIUM,

ESTIMATION

PARACETAMOL

OF
AND

METHOCARBAMOL
6.1 STABILITY INDICATING HPLC METHOD DEVELOPMENT FOR
SIMULTANEOUS ESTIMATION OF DICLOFENAC POTASSIUM,
PARACETAMOL AND METHOCARBAMOL

6.1.1 Experimental
6.1.1.1 Solubility
Solubility of Diclofenac potassium (DIC), Paracetamol (PCM) and Methocarbamol
(MET) were performed in different solvent like distilled water, 0.1N HCl, 0.1N NaOH,
methanol, dimethyl formamide, acetonitrile, ethanol and chloroform. All three drugs were
soluble in methanol.
6.1.1.2 Preparation of mobile phase
The mobile phase methanol: water in the ratio of 80:20, v/v respectively was used. The
mobile phase was filtered through 0.45 filter paper to remove particulate matter and
then degassed by sonication.
6.1.1.3 Preparation of standard solutions
6.1.1.3.1 Preparation of standard stock solutions
6.1.1.3.1.1 Preparation of standard stock solution of DIC
Accurately weighed DIC (50mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 500g/ml stock solution.
6.1.1.3.1.2 Preparation of standard stock solution of PCM
Accurately weighed PCM (32.5mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 325g/ml stock solution.

Ganpat University

129

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.1.3.1.3 Preparation of standard stock solution of MET


Accurately weighed MET (50mg) was transferred in 50ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 1000g/ml stock solution.
6.1.1.3.2 Preparation of standard working solutions
6.1.1.3.2.1 Preparation of standard working solution of DIC
From the stock solution (500g/ml), an accurately measured 0.2, 0.4, 0.6, 0.8, 1.0 and
1.2ml was transferred into separate 10ml volumetric flask and final volume was adjusted
with methanol upto mark to prepare 10-60g/ml solutions.
6.1.1.3.2.2 Preparation of standard working solution of PCM
From the stock solution (325g/ml), an accurately measured 2, 4, 6, 8, 10 and 12ml was
transferred into separate 10ml volumetric flask and final volume was adjusted with
methanol upto mark to prepare 65-390g/ml solutions.
6.1.1.3.2.3 Preparation of standard working solution of MET
From the stock solution (1000g/ml), an accurately measured 1, 2, 3, 4, 5 and 6ml was
transfer into separate 10ml volumetric flask and final volume was adjusted with methanol
upto mark to prepare 100-600g/ml solutions.
6.1.1.4 Preparation of sample solution
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 32.5mg of PCM and 500mg of MET was transferred to a 100ml
volumetric flask. The powder was dissolved in 60ml of methanol with sonication for 15
minutes and volume was made up with methanol. 1ml of the solution was transferred into
10ml volumetric flask and diluted upto mark with methanol.
6.1.1.5 Determination of wavelength maxima
Solutions of DIC, PCM and MET were scanned between 200 and 400nm. UV spectra of
all three drugs show maximum absorbance at 225nm. (Figure 6.1.1)

Ganpat University

130

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.1 Overlay UV spectra of DIC, PCM and MET


6.1.1.6 Chromatographic conditions
The chromatographic separations were performed using Varian C-18 (250 4.6 mm i.d,
5 m particle size) column at room temperature. The optimum mobile phase consisted of
methanol: water in the ratio of 80:20, v/v respectively. 20 l sample was injected for
analysis. The analysis was done with flow rate of 0.8 ml/min at 225 nm wavelength using
Dual wavelength UV Detector.
6.1.1.7 Method validation[1-2]
6.1.1.7.1 Linearity and range
The calibration curve was plotted over the concentration range of 10-60g/ml for DIC,
65-390g/ml for PCM and 100-600g/ml for MET. All the solution were filtered through
0.2m membrane filter and injected, chromatograms were recorded and it was repeated
for six times. A calibration graph was plotted between the mean peak area Vs respective
concentration and regression equation was derived.
6.1.1.7.2 Accuracy
The accuracy of the method was determined by calculating recoveries of DIC, PCM and
MET by standard addition method. Known amount of standard solution of DIC, PCM
and MET (80, 100 and 120% level) were added to pre-analysed samples.

Ganpat University

131

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.1.7.3 Precision
Precision was evaluated in terms of intraday and interday precision. The intraday
precision was investigated using three different concentrations of sample solutions. The
intraday and interday precision of the proposed method was determined by analyzing the
corresponding concentration three times on the same day and on different days,
conditions and changing the wavelength.
6.1.1.7.5 Limit of detection and quantification
The LOD may be expressed as: DL =3.3 / S
Where = the standard deviation of the response
S = the slope of the calibration curve
The LOQ may be expressed as: QL =10 / S
Where = the standard deviation of the response
S = the slope of the calibration curve
6.1.1.7.6 System suitability
The system suitability parameters like theoretical plates (N), asymmetry factor (As),
capacity factor (K), resolution (Rs), retention time (RT) and tailing factor (Tf) reported
in European Pharmacopoeia[2] were calculated by LC solution software. The HPLC
system was equilibrated with the initial mobile phase composition, followed by six
injections of same standard.
6.1.1.8 Analysis of marketed formulation
The response of sample solution was measured under chromatographic condition as
described above in section 6.1.1.6. The amount of DIC, PCM and MET were determined
by regression equation.
6.1.1.9 Forced degradation study of drug substance [3]
6.1.1.9.1 Acidic degradation
6.1.1.9.1.1 Acidic degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N HCl was added and refluxed at 85C for 24
hours. 6ml of the solution was diluted upto 10ml with methanol (60g/ml) and analysed
by HPLC.

Ganpat University

132

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.1.9.1.2 Acidic degradation of PCM


32.5mg of PCM was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours (325g/ml) and the solution was analysed by HPLC.
6.1.1.9.1.3 Acidic degradation of MET
50mg of MET was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. The solution (500g/ml) was analysed by HPLC.
6.1.1.9.2 Alkaline degradation
6.1.1.9.2.1 Alkaline degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at 80C for 24
hours. 6ml of the solution was diluted upto 10ml with methanol (60g/ml) and analysed
by HPLC.
6.1.1.9.2.2 Alkaline degradation of PCM
32.5mg of PCM was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1 N NaOH was added and refluxed at 80C for 24
hours (325g/ml) and the solution was analysed by HPLC.
6.1.1.9.2.3 Alkaline degradation of MET
50mg of MET was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N NaOH was added and refluxed at 80C for 24
hours. The solution (500g/ml) was analysed by HPLC.
6.1.1.9.3 Oxidative degradation
6.1.1.9.3.1 Oxidative degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 24 hours. 6.0ml of the solution was diluted upto 10ml with methanol (60g/ml) and
analysed by HPLC.

Ganpat University

133

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.1.9.3.2 Oxidative degradation of PCM


32.5mg of PCM was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 24 hours (325g/ml) and the solution was analysed by HPLC.
6.1.1.9.3.3 Oxidative degradation of MET
50mg of MET was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 24 hours. The solution (500g/ml) was analysed by HPLC.
6.1.1.9.4 Thermal degradation
6.1.1.9.4.1 Thermal degradation of DIC
10mg of DIC weighed accurately, kept at 105C for 8 hours and transferred in 100ml
volumetric flask. The powder was dissolved in 50ml of methanol and diluted upto mark
with methanol. 6ml of the solution was diluted upto 10ml with methanol (60g/ml) and
analysed by HPLC.
6.1.1.9.4.2 Thermal degradation of PCM
32.5mg of PCM weighed accurately, kept at 105C for 8 hours and transfered in 100ml
volumetric flask. The powder was dissolved in 50ml of methanol and diluted upto mark
with methanol (325g/ml) and the solution was analysed by HPLC.
6.1.1.9.4.3 Thermal degradation of MET
50mg of MET weighed accurately, kept at 105C for 8 hours an transfered in 100ml
volumetric flask. The powder was dissolved in 50ml of methanol and diluted upto mark
with methanol (500g/ml) and analysed by HPLC.
6.1.1.9.5 Neutral degradation
6.1.1.9.5.1 Neutral degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and refluxed at 85C for 24 hours.
6ml of the solution was diluted upto 10ml with methanol (60g/ml) and analysed by
HPLC.

Ganpat University

134

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.1.9.5.2 Neutral degradation of PCM


32.5mg of PCM was weighed accurately, transfer into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hour
(325g/ml) and the solution was analysed by HPLC
6.1.1.9.5.3 Neutral degradation of MET
50mg of MET was weighed accurately, transfer into 10ml volumetric flask and dissolved
in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hours. The solution
(500g/ml) was analysed by HPLC.
6.1.1.10 Forced degradation study of marketed product
6.1.1.10.1 Acidic degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 5mg of DIC, 32.5mg of PCM and 50mg of MET was transferred to a 100ml volumetric
flask and dissolved in 50ml methanol. 50ml of 0.1N HCl was added and refluxed at 85C
for 24 hours and analysed by HPLC.
6.1.1.10.2 Alkaline degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 5mg of DIC, 32.5mg of PCM and 50mg of MET was transferred to a 100ml volumetric
flask and dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at
80C for 24 hours.
6.1.1.10.3 Oxidative degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 5mg of DIC, 32.5mg of PCM and 50mg of MET was transferred to a 100ml volumetric
flask and dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room
temperature for 24 hours.
6.1.1.10.4 Thermal degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 5mg of DIC, 32.5mg of PCM and 50mg of MET was kept at 105C for 8 hours and
transferred in 100ml volumetric flask. The powder was dissolved in 50ml of methanol
and diluted upto mark with methanol.

Ganpat University

135

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.1.10.5 Neutral degradation of marketed product


Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 5mg of DIC, 32.5mg of PCM and 50mg of MET was transferred to a 100ml volumetric
flask and dissolved in 50ml methanol. 50ml of water was added and refluxed at 80C for
24 hours.

6.1.2 Results and discussion


6.1.2.1 Method optimization
The aim of this study was to develop a stability indicating HPLC method for
simultaneous analysis of DIC, PCM and MET. The review of literature for estimation of
other muscle relaxant drugs either alone or in combination and knowledge of molecule
suggest that reverse phase liquid chromatography (RPLC) is suitable for simultaneous
analysis of DIC, PCM and MET. In case of RP-HPLC various columns are available but
C18 column was preferred over other columns. Different mobile phases containing
acetonitrile, methanol and water were used. The mobile phase methanol: water in
different ratio was tried. Hence forth, changing the composition of mobile phase
optimized the chromatographic condition. It was found that methanol: water in ratio of
80:20, v/v respectively gave acceptable retention time (tR 3.51min for DIC, tR 6.42 min
for PCM and tR 9.90 min for MET) and good resolution for DIC, PCM and MET with
flow rate of 0.8 ml/min at 225 nm.(Table 6.1.1a) The method parameter was optimized to
analyse DIC, PCM and MET in marketed product. A chromatogram is shown in Figure
6.1.2b
Table 6.1.1a Results of optimization of mobile phase
Mobile phase

Ratio

Result

Acetonitrile: water

50:50

Poor elution of drugs

Acetonitrile: water

70:30

No good shape of drug and


poor resolution of peaks

Methanol: water

50:50

High retention time with bad


shape

Methanol: water

80:20

Less retention time with good


shape and better separation

Ganpat University

136

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.2a: Blank Chromatogram by HPLC

Figure 6.1.2b: Chromatogram of DIC, PCM and MET by HPLC


6.1.2.2.1 Linearity and range
The calibration curve was plotted over the concentration range of 10-60g/ml for DIC,
65-390g/ml for PCM and 100-600g/ml for MET. A calibration graph was plotted
between the mean peak area Vs respective concentration and regression equation was
derived (Figure 6.1.3, 6.1.4, 6.1.5). The results were shown in Table 6.1.1b.

Ganpat University

137

Chapter 6

Stability indicating methods for muscle relaxant drugs

350000

P
e 300000
a 250000
k

y = 5085x - 1345.1
R = 0.9998

200000

a 150000
r
100000
e
a 50000
0

20

40
60
Concentration (g/ml)

80

Figure 6.1.3: Calibration curve of DIC by HPLC

2000000

P 1800000
e 1600000
a 1400000
k 1200000
a
r
e
a

y = 4606.9x + 14242
R = 0.9993

1000000
800000
600000
400000
200000
0
0

100

200

300

400

500

Concentration (g/ml)
Figure 6.1.4: Calibration curve of PCM by HPLC

Ganpat University

138

Chapter 6

Stability indicating methods for muscle relaxant drugs

P
e
a
k
a
r
e
a

3000000
2500000
2000000

y = 4030.6x + 1086
R = 0.9992

1500000
1000000
500000
0
0

100

200

300

400

500

600

700

Conccentration (g/ml)
Figure 6.1.5: Calibration curve of MET by HPLC
Table 6.1.1b Linearity and range data for DIC, PCM and MET by HPLC
Drug

Linearity range

Y=mx+c

Slope*
DIC
10-60g/ml
5085
PCM
65-390g/ml
4606.9
MET
100-600 g/ml
4030.6
*=Average result of six replicate samples

Intercept*
1345.1
14242
1086

r2*
0.9998
0.9993
0.9992

6.1.2.2.2 Accuracy (Recovery study)


The recovery for DIC, PCM and MET were found between 99% and 101%. The results
were shown in Table 6.1.2.
Table 6.1.2 Recovery study for DIC, PCM and MET by HPLC
Conc. Of
Conc. Of
Drug
Form.
Std. added
(g/ml)
(g/ml)
20
20
DIC
20
25
20
30
165
135
PCM
165
165
165
195
200
250
MET
200
200
200
250
*= Average result of six replicate samples
Ganpat University

Conc.
Recover
(g/ml)
40.03
44.84
49.89
299.89
326.94
361.83
450.05
400.87
448.13

% recoverySD*
100.07%0.046
99.64%0.109
99.78%0.046
99.96%0.085
99.07%0.16
100.50%0.11
100.02%0.12
100.21%0.12
99.58%0.10

139

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.2.2.3 Precision
The %RSD of intraday and interday precision study for DIC, PCM and MET were found
to be <2. The results were shown in Table 6.1.3.
Table 6.1.3 Intra-day & Inter-day precision for DIC, PCM and MET by HPLC
Intra day
Mean %assay*
%RSD*
DIC
98.84%0.30
1.22
PCM
98.29%1.18
0.26
MET
98.94%0.12
0.14
*= Average result of six replicate samples
Drug

Inter day
Mean %assay*
%RSD*
98.76%0.25
1.26
99.06%0.36
0.48
99.86%0.19
0.52

6.1.2.2.3 Robustness
To determine the robustness of the developed method, experimental conditions were
deliberately altered and the responses of all drugs were recorded. The results of change in
ratio of mobile phase, flow rate and wavelength are shown in Table 6.1.4.
Table 6.1.4 Robustness data for DIC, PCM and MET by HPLC
Chromato
graphic
factor

level

Retention time*
DIC

PCM

0.6ml/m
in
1.2ml/m
in

3.720. 6.830.
02
01
Flow rate
3.250. 6.140.
03
06
3.700. 6.820.
68:32
09
01
Methanol:
water
3.740. 6.150.
72:28
08
08
3.710. 6.130.
268nm
Detection
08
01
wavelengt
3.710. 6.130.
h
276nm
09
01
*= Average result of six replicate samples

Tailing factor*

MET

DIC

PCM

MET

10.320.
01
9.230.0
8
10.320.
01
10.230.
01
9.410.0
8
9.410.0
8

1.060.
01
1.080.
08
1.080.
05
1.070.
08
1.070.
01
1.070.
09

1.080..
01
1.090.0
8
1.080.0
8
1.080.0
1
1.080.0
1
1.090.0
1

0.970.
01
0.960.
01
0.960.
08
0.970.
08
0.970.
01
0.970.
02

6.1.2.2.4 Limit of detection and quantification


The LOD for DIC, PCM & MET were found to be 0.15g/ml, 2.40g/ml and 1.82g/ml
respectively, while LOQ were 0.48g/ml, 7.29g/ml and 5.53g/ml respectively.

Ganpat University

140

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.2.2.5 System suitability


The parameters like retention time, asymmetric factor, number of theoretical plates and
tailing factors were evaluated for DIC, PCM and MET. The results were shown in Table
6.1.5.
Table 6.1.5 System suitability parameters for DIC, PCM and MET by HPLC
Parameter

RT*

AUC*

No. of
theoretical
plates*

Tailing
factor*

DIC
3.510.03
50152.6503.35 3590.490.52 1.040.02
PCM
6.420.04
312120.2112.54 3437.4139.80 1.080.01
MET
9.900.05
400120.0 3923.11 1263.127.005 0.960.008
*=Average result of six replicate samples
5.1.2.3 Analysis of marketed product
Market formulations containing 50mg DIC, 325mg PCM and 500mg MET was analysed
by HPLC.
6.1.2.4 Forced degradation study
Forced degradation studies were performed for bulk drug and marketed product, to
provide an indication of the stability indicating property. The degradation was attempted
to stress conditions like acid hydrolysis, alkaline hydrolysis, oxidative hydrolysis,
thermal treatment and neutral degradation, in order to evaluate the ability of the proposed
method to separate drug from its degradation products [3]. During forced degradation
experiments, more degradation was observed in DIC samples under acidic, alkaline and
oxidative degradation. Mild degradation was observed in PCM sample under oxidative
conditions whereas degradation was not observed under acidic, thermal and neutral
conditions. Moderate degradation was observed in MET acidic, alkaline, oxidative and
neutral samples under stress conditions. Table 6.1.7 and 6.1.8 indicates the extent of
degradation of drug substance and marketed product under various stress conditions.
Figures 6.1.6 to 6.1.15 shows the chromatograms of forced degraded samples. The
degradation products were well resolved from drug, confirming the stability indicating
power of the method.

Ganpat University

141

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.6a: Chromatogram of acidic degradation of DIC by HPLC

Figure 6.1.6b: Chromatogram of acidic degradation of PCM by HPLC

Figure 6.1.6c: Chromatogram of acidic degradation of MET by HPLC

Ganpat University

142

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.7a: Chromatogram of alkaline degradation of DIC by HPLC

Figure 6.1.7b: Chromatogram of alkaline degradation of PCM by HPLC

Figure 6.1.7c: Chromatogram of alkaline degradation of MET by HPLC


Ganpat University

143

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.8a: Chromatogram of oxidative degradation of DIC by HPLC

Figure 6.1.8b: Chromatogram of oxidative degradation of PCM by HPLC

Figure 6.1.8c: Chromatogram of oxidative degradation of MET by HPLC


Ganpat University

144

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.9a: Chromatogram of thermal degradation of DIC by HPLC

Figure 6.1.9b: Chromatogram of thermal degradation of PCM by HPLC

Figure 6.1.9c: Chromatogram of thermal degradation of MET by HPLC

Ganpat University

145

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.10a: Chromatogram of neutral degradation of DIC by HPLC

Figure 6.1.10b: Chromatogram of neutral degradation of PCM by HPLC

Figure 6.1.10c: Chromatogram of neutral degradation of MET by HPLC

Ganpat University

146

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.11: Chromatogram of acidic degradation of marketed product by


HPLC

Figure 6.1.12: Chromatogram of alkaline degradation of marketed product by


HPLC

Figure 6.1.13: Chromatogram of oxidative degradation of marketed product by


HPLC
Ganpat University

147

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.1.14: Chromatogram of thermal degradation of marketed product


by HPLC

Figure 6.1.15: Chromatogram of neutral degradation of marketed product


by HPLC

Ganpat University

148

Chapter 6

Stability indicating methods for muscle relaxant drugs

Table 6.1.6 Forced degradation study of DIC, PCM and MET by HPLC
RT of
Degradation
Conc. Of
Drug
observed
condition
drug(g/ml)
peak*
DIC
PCM

Alkaline

Oxidative

Thermal

Neutral

%drug*

%
degradation*

50

3.56

241732

99.14%0.00

0.00%0.00

325

6.42

1420526

100%1.01

0.00%0.00

9.90

1807432 94.86%1.04

Acidic
MET

AUC*

500

DIC

50

PCM

325

MET

500

DIC

50

PCM

325

MET

500

DIC
PCM
MET

0.00%0.00

5.49(I)

36123

0.00%0.00

1.99%0.98

11.10(II)

84231

0.00%0.00

4.36%1.62

4.11

231415

95.34%0.98

0.00%0.00

3.52(I)

9329

0.00%0.00

4.03%0.99

6.39

1419311 99.21%1.48

0.00%0.00

9.90
8.92(II)
4.11

1806415 94.12%1.35
95120
0.00%0.00
231811 95.39%1.21

0.00%0.00
5.5%0.25
0.00%0.00

3.49(I)

9653

0.00%0.00

4.16%0.50

50
325
500

6.39
5.62(II)
9.90
10.32(III)
3.51
6.42
9.90

1419769
11981
1806926
91824
251830
1500874
2007891

91.76%1.34
0.00%0.00
99.54%1.21
0.00%0.00
98.99%1.09
99.42%1.09
99.29%1.21

0.00%0.00
8.64%1.21
0.00%0.00
5.06%0.50
0.00%0.00
0.00%0.00
0.00%0.00

DIC

50

4.11

24981

98.44%12

0.00%0.00

PCM

325

6.39
9.90
MET
500
10.29(I)
*=Average result of six replicate samples

Ganpat University

1465896 98.97%1.21
1805341 94.76%1.09
98691
0.00%0.00

0.00%0.00
0.00%0.00
5.46%0.78

149

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.1.3 Conclusion
The isocratic RP-HPLC method developed for the analysis of ternary mixtures of DIC,
PCM and MET as bulk drug and in pharmaceutical preparation is simple, accurate,
precise, and repeatable with short run time. In forced degradation the drug degrades as
shown by the decreased areas in peaks when compared with peak areas of the same
concentration of the non-degraded drug, with giving additional degradation peaks at
different retention time. The developed method is stability indicating and separate
degradants and can be used by quality control department to determine the assay of
pharmaceutical preparation and also stability samples.

Ganpat University

150

Chapter 6

6.2

STABILITY

Stability indicating methods for muscle relaxant drugs

INDICATING

HPTLC

METHOD

FOR

SIMULTANEOUS ESTIMATION OF DICLOFENAC POTASSIUM,


PARACETAMOL AND METHOCARBAMOL
6.2.1 Experimental
6.2.1.1 Solubility
Solubility of Diclofenac potassium (DIC), Paracetamol(PCM) and Methocarbamol
(MET) in different solvent like distilled water, 0.1N HCl, 0.1N NaOH, methanol,
dimethyl formamide, acetonitrile, ethanol and chloroform. All three drugs were soluble in
methanol.
6.2.1.2 Preparation of mobile phase
The mobile phase toluene:ethyl acetate:Methanol in the ratio of 4:3:2, v/v/v respectively
was used.
6.2.1.3 Preparation of standard solutions
6.2.1.3.1 Preparation of standard stock solutions
6.1.1.3.1.1 Preparation of standard stock solution of DIC
Accurately weighed DIC (10mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 100g/ml stock solution.
6.2.1.3.1.2 Preparation of standard stock solution of PCM
Accurately weighed PCM (10mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 100g/ml stock solution.
6.2.1.3.1.3 Preparation of standard stock solution of MET
Accurately weighed MET (10mg) was transferred in 100ml volumetric flask. The drug
was dissolve in methanol with sonication and final volume was adjusted with methanol
upto mark to prepare a 100g/ml stock solution.
6.2.1.4 Preparation of sample solution
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 325mg of PCM and 500mg of MET was transferred to a 100ml
volumetric flask. The powder was dissolved in 60ml of methanol with sonication for 15
minutes and volume was made up with methanol.
Ganpat University

151

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.1.5 Determination of wavelength maxima


Solutions of DIC, PCM and MET were scanned between 200 and 400nm. UV spectra of
all three drugs show maximum absorbance at 225nm.

Figure 6.2.1 Overlay UV spectra of DIC, PCM and MET


6.2.1.6 Chromatographic conditions
Chromatography was performed on 10x10cm Precoated Silica Gel G60F 254 aluminium
sheets. The samples were applied to the plates as 6mm band, 10 mm apart, by means of
Linomat-V sample applicator with help of 100l Hamilton syringe. It was developed in
CAMAG TLC chamber (20x10cm, 20x20cm) which was already saturated for 30 min.
with mobile phase at room temperature. The optimum mobile phase consisted of
toluene:ethyl acetate:methanol in the ratio of 4:3:2, v/v/v respectively. After development
the plate was scanned at 225 nm by means of CAMAG TLC Scanner 3 controlled by
WinCATs software.
6.2.1.7 Method validation[1-2]
6.2.1.7.1 Linearity and range
The calibration curve was plotted over the concentration range of 100-6000ng/spot for
DIC, 150-650ng/spot for PCM and 200-1200ng/spot for MET. All the solutions were
spoted on precoated TLC plate, chromatograms were recorded and it was repeated for six
Ganpat University

152

Chapter 6

Stability indicating methods for muscle relaxant drugs

times. A calibration graph was plotted between the mean peak height Vs respective
concentration and regression equation was derived.
6.2.1.7.2 Accuracy
The accuracy of the method was determined by calculating recoveries of DIC, PCM and
MET by standard addition method. Known amount of standard solution of DIC, PCM
and MET (80, 100 and 120% level) were added to preanalysed samples.
6.2.1.7.3 Precision
Precision was evaluated in terms of intra-day and inter-day precision. The intraday
precision was investigated using three different concentrations of sample solutions. The
intraday and interday precision of the proposed method was determined by analyzing the
corresponding concentration three times on the same day and on different days. The TLC
plate was developed and analysed as described under chromatographic condition.
6.2.1.7.4 Robustness
Robustness was determined by the analysis of the samples under a variety of conditions
making small changes in the ratio of mobile phase, in the saturation time, changing the
wavelength.
6.2.1.7.5 Limit of detection and quantification
The LOD may be expressed as: DL =3.3 / S
Where = the standard deviation of the response, S = the slope of the calibration curve
The LOQ may be expressed as: QL =10 / S
Where = the standard deviation of the response, S = the slope of the calibration curve
6.2.1.8 Analysis of marketed product
0.8, 1.0 and 1.2l of the sample solution as described in 6.2.1.4 were spotted on
precoated TLC plate. 1ml of sample solution (6.2.1.4) was diluted upto 10ml with
methanol and 0.8, 1.0 and 1.2l of this solution were spotted on precoated TLC plate.
The TLC plate was developed and analysed as described under chromatographic
condition. The amount of DIC, PCM and MET were determined by regression equation.

Ganpat University

153

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.1.9 Forced degradation study of drug substance [3]


6.2.1.9.1 Acidic degradation
6.2.1.9.1.1 Acidic degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N HCl was added and refluxed at 85C for 24
hours. 1.0l of the solution was spotted on precoated TLC plate (100ng/spot), developed
and analysed as described under chromatographic condition.
6.2.1.9.1.2 Acidic degradation of PCM
25mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. 1.0l of the solution was spotted on precoated TLC plate (250ng/spot), developed
and analysed as described under chromatographic condition.
6.2.1.9.1.3 Acidic degradation of MET
50mg of MET was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N HCl was added and refluxed at 85C for 24
hours. 1.0l of the solution was spotted on precoated TLC plate (500ng/spot), developed
and analysed as described under chromatographic condition.
6.2.1.9.2 Alkaline degradation
6.2.1.9.2.1 Alkaline degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at 80C for 24
hours. 2.0l of the solution was spotted on precoated TLC plate (100ng/spot), developed
and analysed as described under chromatographic condition.
6.2.1.9.2.2 Alkaline degradation of PCM
25mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and refluxed at 80C for 24
hours. 1.0l of the solution was spotted on precoated TLC plate (250ng/spot), developed
and analysed as described under chromatographic condition.
6.2.1.9.2.3 Alkaline degradation of MET
50mg of MET was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 5N NaOH was added and refluxed at 80C for 24
Ganpat University

154

Chapter 6

Stability indicating methods for muscle relaxant drugs

hours. 1.0l of the solution was spotted on precoated TLC plate (500ng/spot), developed
and analysed as described under chromatographic condition.
6.2.1.9.3 Oxidative degradation
6.2.1.9.3.1 Oxidative degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 24 hours. 1.0l of the solution was spotted on precoated TLC plate (100ng/spot),
developed and analysed as described under chromatographic condition.
6.2.1.9.3.2 Oxidative degradation of PCM
25mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 24 hours. 1.0l of the solution was spotted on precoated TLC plate (250ng/spot),
developed and analysed as described under chromatographic condition.
6.2.1.9.3.3 Oxidative degradation of MET
50mg of MET was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of 3% H2O2 was added and kept at room temperature
for 24 hours. 1.0l of the solution was spotted on precoated TLC plate (500ng/spot),
developed and analysed as described under chromatographic condition.
6.2.1.9.4 Thermal degradation
6.2.1.9.4.1 Thermal degradation of DIC
10mg of DIC was weighed accurately, kept at 105c for 8 hours and transferred into
100ml volumetric flask. The powder was dissolved in 50ml methanol and diluted upto
mark with methanol. 1.0l of the solution was spotted on precoated TLC plate
(100ng/spot), developed and analysed as described under chromatographic condition.
6.2.1.9.4.2 Thermal degradation of PCM
25mg of PCM was weighed accurately, kept at 105C for 8 hours and transferred into
100ml volumetric flask. The powder was dissolved in 50ml methanol and diluted upto
mark with methanol. 1.0l of the solution was spotted on precoated TLC plate
(250ng/spot), developed and analysed as described under chromatographic condition.

Ganpat University

155

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.1.9.4.3 Thermal degradation of MET


50mg of MET was weighed accurately, kept at 105C for 8 hours and transferred into
100ml volumetric flask. The powder was dissolved in 50ml methanol and diluted upto
mark with methanol. 1.0l of the solution was spotted on precoated TLC plate
(500ng/spot), developed and analysed as described under chromatographic condition.
6.2.1.9.5 Neutral degradation
6.2.1.9.5.1 Neutral degradation of DIC
10mg of DIC was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and refluxed at 85C for 24 hours.
1.0l of the solution was spotted on precoated TLC plate (100ng/spot), developed and
analysed as described under chromatographic condition.
6.2.1.9.5.2 Neutral degradation of PCM
25mg of PCM was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hours.
1.0l of the solution was spotted on precoated TLC plate (250ng/spot), developed and
analysed as described under chromatographic condition.
6.2.1.9.5.3 Neutral degradation of MET
50mg of MET was weighed accurately, transferred into 100ml volumetric flask and
dissolved in 50ml methanol. 50ml of water was added and reflux at 85C for 24 hours.
1.0l of the solution was spotted on precoated TLC plate (400ng/spot), developed and
analysed as described under chromatographic condition.
6.2.1.10 Forced degradation study of marketed product
6.2.1.10.1 Acidic degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 325mg of PCM and 500mg of MET was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 0.1N HCl was added and
refluxed at 85C for 24 hours. 1.0ml of the solution diluted upto 10ml with methanol then
2.0l of the solution was spotted on precoated TLC plate (100ng/spot DIC, 650ng/spot
PCM, and 1000ng/spot MET), developed and analysed as described under
chromatographic condition.

Ganpat University

156

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.1.10.2 Alkaline degradation of marketed product


Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50 mg of DIC, 325mg of PCM and 500mg of MET was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of 0.1N NaOH was added and
refluxed at 85C for 24 hours. 1ml of the solution diluted upto 10ml with methanol then
1.0l of the solution was spotted on precoated TLC plate (100ng/spot DIC, 650ng/spot
PCM

and

1000ng/spot

MET)

developed

and

analysed

as

described

under

chromatographic condition
6.2.1.10.3 Oxidative degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 325mg of PCM and 500mg of MET was transferred to a 100 ml
volumetric flask and dissolved in 50ml methanol. 50ml of 3% H 2O2 was added and kept
at room temperature for 24 hours. 1ml of the solution diluted upto 10ml with methanol
then 2.0l of the solution was spotted on precoated TLC plate (100ng/spot DIC,
650ng/spot PCM and 1000ng/spot MET) developed and analysed as described under
chromatographic condition.
6.2.1.10.4 Thermal degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 325mg of PCM and 500mg of MET was kept at 105C for 8 hours and
transferred in 100ml volumetric flask. The powder was dissolved in 50ml of methanol
and diluted upto mark with methanol. 1ml of the solution diluted upto 10ml with
methanol then 2.0l of the solution was spotted on precoated TLC plate (100ng/spot DIC,
650ng/spot PCM and 1000ng/spot MET) developed and analysed as described under
chromatographic condition.
6.2.1.10.5 Neutral degradation of marketed product
Twenty tablets were weighed accurately and finely powdered. Powder exactly equivalent
to 50mg of DIC, 325mg of PCM and 500mg of MET was transferred to a 100ml
volumetric flask and dissolved in 50ml methanol. 50ml of water was added and refluxed
at 85C for 24 hours. 1ml of the solution diluted upto 10ml with methanol then 2.0l of
the solution was spotted on precoated TLC plate (100ng/spot DIC, 650ng/spot PCM and
400ng/spot MET) developed and analysed as described under chromatographic condition.
Ganpat University

157

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.2 Results and discussion


6.2.2.1 Method optimization
For optimization, different mobile phases and composition were employed to achieve the
good separation. The method development was initiated with using a mobile phase of
toluene: methanol:ammonium sulphate in various proportions. In the above conditions
only two drugs were separated. All three drugs were separated using mobile phase
consisting of different ratio of toluene: ethylacetate:methanol. Finally mobile phase
consisting mixture of toluene:ethyl acetate:methanol in ratio of 4:3:2, v/v/v respectively
gave reasonable Rf (Rf 0.78 for DIC, 0.63 for PCM and 0.42 for MET) and sharp band
for all three drugs. Saturation of TLC chamber for 30 min assured better reproducibility
and better resolution. All three drugs were detected at 272 nm by means of CAMAG TLC
Scanner 3. A chromatogram is shown in Figure 6.2.2.

Figure 6.2.2: Chromatogram of DIC, PCM and MET by HPTLC

6.2.2.2 Method validation


6.2.2.2.1 Linearity and range
The calibration curve was plotted over the concentration range of 100-600 ng/spot for
DIC, 150-650 ng/spot for PCM and 200-1200 ng/spot for MET. A calibration graph was
plotted between the mean peak height Vs respective concentration and regression
equation was derived (Figure 6.2.3, 6.2.4, 6.2.5). The results were shown in Table 6.2.1.

Ganpat University

158

Chapter 6

Stability indicating methods for muscle relaxant drugs


10000
9000
8000
7000
6000
5000
4000
3000
2000
1000
0

P
e
a
k
A
r
e
a

y = 15.089x + 47.333
R = 0.9997

100

200

300

400

500

600

700

Concentration (ng/spot)

Figure 6.2.3: Calibration curve for DIC by HPTLC


16000

P 14000
e
a 12000
k 10000
A
r
e
a

y = 22.766x + 53.714
R = 0.9989

8000
6000
4000
2000
0
0

200

400

600

800

Concentration (ng/spot)
Figure 6.2.4: Calibration curve of PCM by HPTLC

Ganpat University

159

Chapter 6

Stability indicating methods for muscle relaxant drugs

P
Pe
ea
ak
k
A
Ar
re
ea
a

20000
18000
16000
14000

y = 14.865x + 196.53
R = 0.9995

12000
10000
8000
6000
4000
2000
0
0

500

Concentration (ng/spot)

1000

1500

Figure 6.2.5: Calibration curve of MET by HPTLC


Table 6.2.1 Linearity and range data for DIC, PCM and MET by HPTLC
Drug

Linearity range

DIC
100-600ng/spot
PCM
150-650ng/spot
MET
200-1200ng/spot
*= Average result of six replicate samples

Y=mx+c
Slope*
Intercept*
15.089
47.33
22.76
53.71
14.865
196.53

r2*
0.9997
0.9989
0.9995

6.2.2.2.2 Accuracy (Recovery study)


The recovery for DIC, PCM and MET were found between 99.0 % and 101%. The results
were shown in Table 6.2.2.
Table 6.2.2 Recovery study for DIC, PCM and MET by HPTLC
Conc. of
Conc. of
Form.
Std. added
(ng/spot)
(ng/spot)
300
250
DIC
300
300
300
350
250
150
PCM
250
250
250
350
600
400
MET
600
500
600
600
*= Average result of six replicate samples
Drug

Ganpat University

Conc.
Recover
(ng/spot)
546.20
594.54
650.73
396.92
496.91
595.22
991.03
1090.1
1188.03

% recoverySD*
99.30%0.047
99.09%0.068
100.11%0.030
99.23%0.123
99.38%0.013
99.20%0.067
99.10%0.050
99.1%0.055
99.00%0.058
160

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.2.2.3 Precision
The %RSD of intraday and inter-day precision study for DIC, PCM and MET were found
to be <2. The results were shown in Table 6.2.3.
Table 6.2.3 Intra-day & Inter-day precision for DIC, PCM and MET by HPTLC
Intra day
Mean %assay*
%RSD*
DIC
98.97%0.015
0.010
PCM
99.97%0.04
0.024
MET
99.90%0.02
0.016
*= Average result of six replicate samples
Drug

Inter day
Mean %assay*
%RSD*
99.97%0.019
0.046
99.98%0.047
0.036
99.99%0.01
0.028

6.2.2.2.3 Robustness
To determine the robustness of the developed method, experimental conditions were
deliberately altered and the responses of all drugs were recorded. The results of change in
ratio of mobile phase, flow rate and wavelength are shown in Table 6.2.4.
Table 6.2.4 Robustness data for DIC, PCM and MET by HPTLC
Chromatographic factor

level

Mobile phase ratio


Toluene: ethyl acetate:
methanol

1.2:1:4v/v/v

Saturation time

Detection wavelength

1:1.2:4v/v/v
32 minutes
28 minutes
275nm
265nm

*= Average result of six replicate samples

Amont found*(ng/spot)
DIC
PCM
MET
299.950.05 399.950.06
599.980.057
4
1
299.940.04 399.930.03
599.960.043
4
8
299.970.03 399.970.04
599.970.047
7
3
299.930.05 399.930.04
599.990.037
0
2
299.930.04 399.950.04
599.950.058
9
3
299.960.04
400.000.02 600.000.033
0

6.2.2.2.4 Limit of detection and quantification


The LOD for DIC, PCM & MET were found to be 0.005 ng/spot, 3.172 ng/spot and
7.617 ng/spot respectively, while LOQ were 0.875 ng/spot, 9.614 ng/spot and 23.083
ng/spot respectively.

6.2.2.3 Analysis of marketed product


The market formulation containing 50mg DIC, 325mg PCM and 500mg MET was
analysed by HPTLC.
Ganpat University

161

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.2.4 Forced degradation study


The degradation was attempted to stress conditions like acid hydrolysis, alkaline
hydrolysis, oxidative hydrolysis, thermal treatment and neutral degradation, in order to
evaluate the ability of the proposed method to separate drug from its degradation
products[3]. HPTLC studies of the samples obtained during the stress testing of DIC,
PCM and MET under different conditions using toluene:ethyl acetate:methanol in the
ratio of 4:3:2, v/v/v respectively as the mobile phase shows different degradation peaks as
shown in Figures 6.2.6 to 6.2.15. DIC showed degradation in alkaline and oxidative stress
conditions at Rf of 0.84 and 0.76 respectively. PCM showed degradation oxidation
condtition at Rf value 0.57. MET showed highest degradation in acidic condition and
degradation products appear at Rf of 0.29 and 0.34 and lower degradation in neutral
condition at Rf of 0.37. Table 6.2.6 indicates the extent of degradation of marketed
product under various stress conditions.

Figure 6.2.6a: Chromatogram of acidic degradation of DIC by HPTLC

Figure 6.2.6b: Chromatogram of acidic degradation of PCM by HPTLC


Ganpat University

162

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.6c: Chromatogram of acidic degradation of MET by HPTLC

Figure 6.2.7a: Chromatogram of alkaline degradation of DIC by HPTLC

Ganpat University

163

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.7b: Chromatogram of alkaline degradation of PCM by HPTLC

Figure 6.2.7c: Chromatogram of alkaline degradation of MET by HPTLC

Figure 6.2.8a: Chromatogram of oxidative degradation of DIC by HPTLC


Ganpat University

164

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.8b: Chromatogram of oxidative degradation of PCM by HPTLC

Figure 6.2.8c: Chromatogram of oxidative degradation of MET by HPTLC

Figure 6.2.9a: Chromatogram of thermal degradation of DIC by HPTLC

Ganpat University

165

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.9b: Chromatogram of thermal degradation of PCM by HPTLC

Figure 6.2.9c: Chromatogram of thermal degradation of MET by HPTLC

Figure 6.2.10a: Chromatogram of neutral degradation of DIC by HPTLC


Ganpat University

166

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.10b: Chromatogram of neutral degradation of PCM by HPTLC

Figure 6.2.10c: Chromatogram of neutral degradation of MET by HPTLC

Figure 6.2.11: Chromatogram of acidic degradation of marketed product by


HPTLC
Ganpat University

167

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.12: Chromatogram of alkaline degradation of marketed product by


HPTLC

Figure 6.2.13: Chromatogram of oxidative degradation of marketed product by


HPTLC

Ganpat University

168

Chapter 6

Stability indicating methods for muscle relaxant drugs

Figure 6.2.14: Chromatogram of thermal degradation of marketed product by HPTLC

Figure 6.2.15: Chromatogram of neutral degradation of marketed product by


HPTLC

Ganpat University

169

Chapter 6

Stability indicating methods for muscle relaxant drugs

Table 6.2.5 Forced degradation study of marketed product by HPTLC


Degradat
ion
condition

acidic

alkaline

oxidative

Thermal

Neutral

Drug

Conc. Of
drug(ng/sp
ot)

Rf value of
observed
peak*

Peak
area*

%drug*

%degradatio
n*

DIC

100

0.78

3234.5

99.12%1.0
8

0.00%0.00

PCM

250

0.63

5318.9

0.42

8965.3

0.29(I)
0.34(II)

692.1
746.7

MET

400

DIC

100

PCM

250

MET

400

DIC

100

PCM

250

MET

400

DIC

0.78
0.84(I)
0.63

3102.8
326.6
5299.36

0.41

8134.9

0.36(II)

425.6

0.78

2894.5

0.76(I)

329.7

0.63

4624.9

0.57(II)

427.4

0.42

8128.8

0.35(III)

692.1

100

0.78

3129.66

PCM

250

0.63

5236.34

MET

400

0.42

8631.34

DIC

100

0.78

2988.75

PCM

250

0.63

4989.6

MET

400

0.42

0.37(I)
*= Average result of six replicate samples
Ganpat University

7984.1
536.9

99.91%1.0
1
84.92%1.0
5
0.00%0.00
0.00%0.00
89.51%0.9
8
0.00%0.00
99.45%1.0
8
94.91%0.2
1
0.00%0.00
88.39%0.2
1
0.00%0.00
90.84%1.3
4
0.00%0.00
91.92%0.2
4
0.00%0.00
99.92%1.0
9
99.96%1.0
9
99.95%0.1
9
98.96%1.0
5
99.92%1.2
1
99.89%0.3
4
0.00%0.00

0.00%0.00
0.00%0.00
7.71%0.99
8.320.99
0.00%0.00
10.50%0.99
0.00%0.00
0.00%0.00
5.22%0.55
0.00%0.00
11.36%0.55
0.00%0.00
9.25%1.26
0.00%0.00
8.51%0.55
0.00%0.00
0.00%0.00
0.00%0.00
4.13%0.99
0.00%0.00
0.00%0.00
6.71%0.99
170

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.2.3 Conclusion
The developed HPTLC technique is simple, accurate, sensitive, precise, rapid and
repeatable. The method was successfully used for determination of DIC, PCM and MET
as bulk drug and in pharmaceutical formulation. After exposing the drugs to different
stress condition, the drug peak area was observed to decrease and also degradant peak
was observed. The developed method is stability indicating and separate degradants and
can be used to determine the assay of pharmaceutical preparation and also stability
samples.

Ganpat University

171

Chapter 6

6.3

Stability indicating methods for muscle relaxant drugs

STATISTICAL

COMPARISON

BETWEEN

OFFICIAL

REPORTED AND PROPOSED METHODS[4-5]


The assay result for DIC, PCM and MET in their dosage form obtained using HPLC and
HPTLC methods were compared with official reported methods by applying the paired ttest and F-test. The calculated t-value for DIC (0.2201), PCM (0.077) and MET (0.3220)
for HPLC as well as DIC (0.3251), PCM (0.0321), and MET (0.1475) for HPTLC is less
than the tabulated t-value (2.571) at the 95% confidence interval. Moreover, the
calculated F-value for DIC (0.1886), PCM (0.2596) and MET (0.1886) for HPLC and
DIC (0.1850), PCM (0.010) and MET (0.1850) for HPTLC is also less than the tabulated
F-value (4.28). Therefore, there is no significant difference in a determined content of
DIC, CHL and PCM by HPLC and HPTLC methods. Table 6.3.1 indicates statistical
comparison between proposed methods.
Table 6.3.1 Statistical comparison for DIC, PCM and MET between proposed
methods
Paired t-test
Drug

DIC

PCM

MET

Methods

Mean

SD

n Tabulated

Official

99.27

0.16 6

HPLC

99.71

0.28 6

Official

99.27

0.16 6

HPTLC

99.28

0.14 6

Official

99.47

0.29 6

HPLC

99.65

0.14 6

Official

99.47

0.29 6

HPTLC

99.15

0.06 6

Official

99.40

0.26 6

HPLC

99.36

0.14 6

Official

99.40

0.14 6

99.48

0.15 6

HPTLC

Ganpat University

F-test

Calculated

Tabulated

Calculated

value

value

value

value

2.571

0.2201

4.28

0.1886

0.3251
2.571

0.077

0.1850
4.28

0.0321
2.571

0.3220
0.1475

0.2596
0.010

4.28

0.1886
0.1850

172

Chapter 6

Stability indicating methods for muscle relaxant drugs

6.4 References
1. ICH, Q2(R1) Validation of Analytical Procedure: Text and Methodology,
International Conference on Harmonization, Geneva, October 1994.
2. The United States Pharmacopeia, The National Formulary, USP 30 NF 25, Asian
Edition, Volume 1, 2007; 680-683.
3. Bakshi M, Singh S, Development of validated stability-indicating assay methods
critical review, Journal of Pharmaceutical and Biomedical Analysis, 2002; 28:
1010-1040.
4. Christian GD, Analytical Chemistry, 6th Ed., University of Washington, 2007; 9097.
5. Sanford Bolton, Charles Bon, Pharmaceutical Statistics Practical and Clinical
Application, 4th Ed., Revised and Expanded, 2004; 417-436.

Ganpat University

173

Chapter 7

Summary

7. SUMMARY
Review was done on muscle relaxant formulations, its cause and drug profile of
the drugs used for the study.
The reported methods for analysis of Diclofenac potassium, Paracetamol,
Chlorzoxazone and Methocarbmol either single or in combination with any one of
mucscle relaxant drugs were reviewed.
Two new simple, precise and accurate methods are described for the
determination of DIC, CHL and PCM in API and in pharmaceutical preparation.
RP-HPLC method was developed and validated using mobile phase consisting of
methanol:phosphate buffer (80:20, v/v respectively), at 0.8ml/min flow rate, C18
column, Autochro-3000 operating software and dual wavelength UV detector.
HPTLC method was developed and validated using mobile phase consisting of
toluene:ethyl acetate:glacial acetic acid(1:2:0.5, v/v/v respectively, at 30 minutes
saturation time, precoated Silica Gel G60F 254 aluminium sheets, CAMAG TLC
Scanner 3 controlled by WinCATs software. Forced degradation study on
individual drug substance and drug product was performed under acidic, alkaline,
oxidative, thermal and neutral condition. The developed analytical methods were
statistically compared using paired t-test and F-test.
Two new simple, precise and accurate methods are described for the
determination of DIC, PCM and MET in both raw material and in laboratory
mixture. RP-HPLC method was developed and validated using mobile phase
consisting of methanol:water (80:20, v/v respectively), at 0.8ml/min flow rate, C 18
column, Autochro-3000 operating software and dual wavelength UV detector.
HPTLC method was developed and validated using mobile phase consisting of
toluene:ethylacetate:methanol(1:2:4, v/v/v respectively), at 30 minutes saturation
time, precoated Silica Gel G60F254 aluminium sheets, CAMAG TLC Scanner 3
controlled by WinCAT software.

Ganpat University

174

Chapter 7

Summary

Forced degradation study on individual drug substance and drug product was
performed under acidic, alkaline, oxidative, thermal and neutral condition. The
developed analytical methods were statistically compared using paired t-test and
F-test.
Forced degradation study on individual drug substance and drug product was
performed under acidic, alkaline, oxidative, thermal and neutral condition.

Ganpat University

175

Chapter 8

Publication

8. PUBLICATION
TITLE

JOURNAL

STATUS

Development and Validation of stability indicating Inventi Rapid:

Published

method for determination of Diclodenac potassium, Pharm Analysis

2012(4): 1-

Chlorzoxazone and Paracetamol in pharmaceutical & Quality


dosage

form

using

high

performance

9, 2012.

liquid Assurance

chromatography
Stability Indicating HPLC method for simultaneous American

Accepted

determination of Diclofenac potassium, Paracetamol Journal of

(in Press)

and Methocarbamol

Pharmaceutical
Technology &
Research

Development and validation of stability indicating Journal of

Under

method for the determination of Paracetamol and Chromatography

review

Methocarbamol in pharmaceutical dosage form B


using HPTLC

Development and validation of simultaneous HPLC Arabian Journal

Under

method

review

for

estimation

of

Paracetamol

and of Chemistry

Methocarbamol.

Ganpat University

176

RESEARCH ARTICLE

Development and Validation of Stability Indicating Method for


Determination of Chlorzoxazone, Diclofenac Potassium and
Paracetamol in Pharmaceutical Dosage form using High
Performance Liquid Chromatography
Maulikkumar R Amin1*, Paresh U Patel2, B N Suhagia3, Madhabhai M Patel4
Abstracts: A stability-indicating HPLC method has been developed and validated for simultaneous estimation chlorzoxazone,
diclofenac potassium and paracetamo in bulk drug and pharmaceutical dosage forms. Stress studies were conducted for all three
drugs under ICH prescribed stress conditions viz. hydrolysis, oxidation, thermal and neutral stress. An isocratic RP-HPLC
method was achieved on younglin HPLC system using Varian C18 (250x4.6 mm i.d, 5 m particle size) column for separation of
drug from its degradation products using mobile phase containing mixture of methanol: phosphate buffer (pH 3.0, adjusted with
glacial acetic acid) (70:30, v/v). The flow rate was 0.8ml/min and the eluent was monitored at 280nm. Linearity was found in
the range of 5-25g/ml for diclofenac potassium (DIC), 25-125g/ml for chlorzoxazone (CHL) and 32.5-182.5g/ml for
paracetamol (PCM). The limit of detection for DIC, CHL & PCM was found to be 0.15g/ml, 1.82g/ml and 2.40g/ml
respectively, while limit of quantification was 0.47g/ml, 5.53g/ml and 7.29g/ml respectively. The developed method was found
to be fast, accurate, precise, reproducible and suitable for analysis of all three drugs in bulk and pharmaceutical dosage forms.

paracetamol). Several UV (9-11) and HPLC (12-16) methods


either single or in combination have been reported for
estimation
of
chlorzoxazone
(CHL),
diclofenac
potassium(DIC) and paracetamol (PCM). Few HPTLC (17-20)
methods are also reported for estimation of DIC and PCM.
The International Conference on Harmonization (ICH)
guideline entitled stability testing of new drug substances
and products requires that stress testing be carried out to
elucidate the inherent stability characteristics of the active
substances. An ideal stability indicating method is one that
resolves the drug and its degradation products efficiently.
Consequently, the implementation of an analytical
methodology to determine CHL, DIC and PCM
simultaneously in presence of its degradation products is
rather a challenge for pharmaceutical analyst. Thus,
thought necessary to study the stability of CHL, DIC and
PCM under acidic, alkaline, neutral hydrolysis, oxidative
and dry heat conditions. This work describes validated
stability-indicating HPLC method for simultaneous
estimation of CHL, DIC, and PCM. Reversed-phase
chromatographic method with UV detection has shown to
be sensitive, accurate and suitable for analyzing a large
support bioavailability and stability studies.

INTRODUCTION
Chlorzoxazone a centrally acting acting agent for painful
musculoskeletal condition it primarily acts at the level of
the spinal cord and subcortical area of brain. It inhibits
degranulation of mast cells, subsequently preventing the
release of histamine and slow-reacting substance of
anaphylaxis, mediators of type I allergic reactions (1). It is
absorbed orally and undergoes first pass metabolism and is
excreted by kidney. t1/2 is 2-3 hrs. It is indicated in
spasticity due to neurological disorders and in painful
muscle spasms of spinal origin (2). Diclofenac potassium [2[(2,6-dichlorophenyl)amino] benzeneacetic acid, mono
potassium salt], is an analgesic which acts by non-steroidal
anti-inflammatory drug (NSAID) that exhibits antiinflammatory, analgesic, and antipyretic activities in animal
models. The mechanism of action of diclofenac potassium
tablets, like that of other NSAIDs, is not completely
understood but may be related to prostaglandin synthetase
inhibition (3-6). It is official in Indian Pharmacopoeia and
United States Pharmacopoeia. It is official in Indian
Pharmacopoeia and United States Pharmacopoeia.
Paracetamol [N-(4-Hydroxyphenyl) acetamide], an
analgesic and antipyretic action with weak anti
inflammatory activity. These effects are related to
inhibition of prostaglandin synthesis (7,8). It is official in
Indian Pharmacopoeia and United States Pharmacopoeia. It
is official in United States Pharmacopoeia (Figure 1
Structure of diclofenac potassium, chlorzoxazone and

EXPERIMENTAL
Reagents and Chemicals
Pure chlorzoxazone, diclofenac potassium and paracetamol
were obtained as a gift samples from Zydus Cadila Ltd
(Ahmedabad, India) with purity of 98.99, 99.87 and 99.93%
respectively. The formulation of the tablet with
combination of diclofenac potassium 50mg, chlorzoxazone
250mg, and paracetamol 325mg is available in market by
brand name Dan-MR. Tablet was purchased from the local
market. Methanol and acetonitrile of HPLC grade were
procured by Merck Ltd., India. Disodium hydrogen
phosphate was procured from Merck Ltd., India. Sodium
hydroxide, hydrochloric acid, disodium hydrogen
phosphate, hydrogen peroxide and glacial acetic acid of
analytical reagent were procured from Merck Ltd., India.

Kalol Institute of Pharmacy, Kalol-Mehsana Highway, Kalol -382721,


Gujarat, India.
E-mail: amin_maulik2003@yahoo.co.in
*Corresponding author
1

Shree S. K. Patel College of Pharmaceutical Education & Research, Ganpat


University, Kherva, Mehsana-382711, Gujarat, India.

Faculty of Pharmacy, D. D. University College Road, Nadiad-387001,


Gujarat, India.

Shankersinh Vaghela Bapu Institute of Pharmacy, Gandhinagar-Mansa


Road, PO. Vasan, Gandhinagar-382650, Gujarat, India.

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
Table 1: Results of System Suitability Parameters
Parameter
DIC
CHL
PCM

RT*
3.250.05
5.600.06
9.400.09

AUC*
25012.20283.43
105410.2312.53
150453.123923.11

No. of Theoretical Plates*


4204.49.52
3637.4139.80
1263.127.005

Tailing Factor*
0.95.002
1.020.01
0.970.001

*=Average result of six replicate samples

Table 2: Linearity and Calibration Curve


Drug

Linearity Range

DIC
CHL
PCM

5-30 g/ml
25-125 g/ml
32.5-182.5 g/ml

Y=mx+c
Slope*
5018.1
4987.5
5051.4

r2*

Intercept*
362
2058
14445

0.9998
0.9997
0.9997

*=Average result of six replicate samples

Table 3: Results of Recovery Studies


Drug

Conc. of Form.(g/ml)
5
5
5
25
25
25
32.5
32.5
32.5

DIC

CHL

PCM

Conc. of Std. Added(g/ml)


4
5
6
24
25
26
34
32.5
36

Conc. Recovered(g/ml) SD*


8.890.12
9.970.30
10.920.02
48.490.07
48.900.71
50.580.05
65.541.51
65.280.08
69.070.16

% RecoverySD*
98.78%0.18
99.70%0.75
99.27%1.89
98.96%0.01
98.16%0.18
99.17%1.09
98.56%0.86
100.41%0.15
100.83%0.11

*= Average result of six replicate samples

Table 4: Results of Intraday & Inter Day Precision


Intra day

Drug

Mean % Assay*
99.34%1.16
99.83%1.63
98.94%0.01

DIC
CHL
PCM

Inter day
%RSD*
0.36
0.12
1.16

Mean % Assay*
99.15%1.82
100.05%0.57
98.86%0.12

% RSD*
1.08
1.12
1.52

*= Average result of six replicate samples

Table 5: Robustness Data for DIC, CHL and PCM


Chromatographic
Factor

level

Flow rate
Methanol:
phosphate buffer (v/v)
Detection wave-length

0.7ml/min
1.0ml/min
75:25
85:15
275nm
285nm

DIC
3.980.02
2.950.02
3.910.03
3.960.02
3.240.04
3.260.03

Retention Time*
CHL
6.010.01
5.040.01
6.020.03
5.050.02
5.600.01
5.600.01

PCM
9.960.01
8.660.03
9.950.02
8.61.02
9.410.01
8.640.02

DIC
0.950.02
0.960.01
0.950.02
0.960.01
0.950.01
0.950.02

Tailing Factor*
CHL
1.020.01
1.030.01
1.020.01
1.020.01
1.030.01
1.020.01

PCM
0.970.01
0.960.01
0.960.02
0.970.01
0.970.03
0.970.01

Table 6: Limit of Detection and Limit of Quantification


Drugs
DIC
CHL
PCM

Standard Deviation
238.7
2759.5
3684.4

Slope of Calibration Curve


5018.1
4987.
5051.4

Instrumentation and Chromatographic Conditions


The HPLC system used was Younglin equipped with
binary solvent managers, manual, dual wavelength UV
detector operated at 280nm. HPLC column incorporated
solvent delivery model LC-10ATVP. Moreover, HPLC
column is Varian C-18 (250x4.6) mm i.d. 5 m particle
size with microliter syringe (rheodyne) injector. The

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

LOD(g/ml)
0.15
1.82
2.40

LOQ(g/ml)
0.47
5.53
7.29

HPLC was connected to personal computer with ClassAutochro-3000 software. The isocratic mobile phase
consisted of methanol : water (80:20 v/v) and was
delivered at a flow rate of 0.8 ml min -1. Detection was
carried out using a UV detector at 280 nm. The column
was maintained at ambient temperature and injection
volume of 20l was used.

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
Table 7: Summary of Validation Parameters
Parameter
Wavelength
Range
Linearity
Intercept
Slope
Intraday precision
Interday precision
LOD
LOQ

DIC
280nm
5-30g/ml
0.9998
362
5018.1
0.36
1.08
0.15
0.47

CHL
280nm
25-125g/ml
0.9997
2058
4987.5
0.12
1.12
1.82
5.53

PCM
280nm
32.5-182.5g/ml
0.9997
14445
5051.4
1.16
1.52
2.40
7.29

Table 8: Analysis of Marketed Formulation of the Product


Brand Name

Drug
DIC
CHL
PCM

DAN-MR

Amount Taken (g/ml)


50
250
325

Amount Found (g/ml)


40.987
249.89
324.26

%Amount found
99.740.22
99.560.11
99.260.21

Table 9: Results of Forced Degradation Study


Degradation Condition

Drug
DIC

Conc. of Drug(g/ml)
5

Acidic

CHL

25

PCM

32.5

DIC

CHL

25

PCM

32.5

DIC

CHL

25

PCM

32.5

DIC
CHL
PCM
DIC

5
25
32.5
5

CHL

25

PCM

32.5

Alkaline

Oxidative

Thermal

Neutral

RTof Observed Peak*


3.25
5.59
5.19(I)
9.40
3.25
2.92(I)
5.62
5.49(II)
6.12(III)
9.41
3.25
2.72(I)
5.60
5.12(II)
9.41
10.21(III)
3.25
5.61
9.41
3.25
5.60
4.93(I)
9.41

% Drug*
% Degradation*
98.14%1.04
0.00%0.00
91.98%1.31
0.00%0.00
0.00%0.00
8.791.31
99.65%1.45
0.00%0.00
94.62%1.24
0.00%0.00
0.00%0.00
5.96%1.21
91.56%1.57
94.10%1.38
0.00%0.00
3.61%1.41
0.00%0.00
1.511.30
99.57%1.34
0.00%0.00
91.41%1.32
0.00%0.00
0.00%0.00
8.98%1.54
97.00%1.03
0.00%0.00
0.00%0.00
2.64%0.95
93.84%1.09
0.00%0.00
0.00%0.00
6.681.41
99.90%1.12
0.00%0.00
99.88%1.08
0.00%0.00
99.84%1.24
0.00%0.00
98.45%1.4
0.00%0.00
91.88%1.23
0.00%0.00
0.00%0.00
8.64%0.95
99.84%1.21
0.00%0.00

Accurately weighed 32.5mg PCM was transferred in 10ml


volumetric flask. The drug was dissolve in methanol with
sonication and final volume was adjusted with methanol
upto mark to prepare a 3250g/ml stock solution.

Preparation of Stock Solutions


Preparation of Standard Stock Solution of DIC
Accurately weighed 50mg DIC was transferred in 100ml
volumetric flask. The drug was dissolve in methanol
with sonication and final volume was adjusted with
methanol upto mark to prepare a 500g/ml stock
solution.

Preparation of Standard Working Solutions


Preparation of Standard Working Solution of DIC
From the stock solution (500g/ml), an aliquot quantity 1,
2, 3, 4, and 5ml were transferred into separate 100ml
volumetric flask and final volume was adjusted with
methanol upto mark to prepare 5-25g/ml solutions.

Preparation of Standard Stock Solution of CHL


Accurately weighed 50mg CHL was transferred in 25ml
volumetric flask. The drug was dissolve in methanol with
sonication and final volume was adjusted with methanol
upto mark to prepare a 2500g/ml stock solution.

Preparation of Standard Working Solution of CHL


From stock solution (2500g/ml), an aliquot quantity 1, 2,
3, 4, and 5 ml were transferred into 100 ml volumetric flask

Preparation of Standard Stock Solution of PCM

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

AUC*
24018
101541
8931
149901
24010
1432
94421
3410
1431
149097
23120
1981
102384
2398
148320
9908
25320
105908
149959
24906
103956
9327
104903

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
Cl

HO

O K+

NH
O
Cl

N
H
Diclofenac Potassium

CH3

Paracetamol
O
OH
N

Cl

Chlorzoxazone
Figure 1: Structure of diclofenac potassium, chlorzoxazone and paracetamol

Figure 2: Overlay UV spectra of DIC, CHL and PCM

Figure 3: A typical Chromatogram for DIC, CHL and PCM


700000

160000
P
e
a
k
a
r
e
a

P
e
a
k

140000
120000
100000
80000

a
r
e
a

y = 5018.1x + 362
R = 0.9998

60000

600000
500000
400000
300000

y = 4987.5x - 20588
R = 0.9997

200000

40000

100000

20000
0

0
0

10

20

30

40

Concentration (g/ml)

50

100

150

Concentration (g/ml)

Figure 4: Calibration curve of DIC

Figure 5: Calibration curve of CHL

and final volume was adjusted with methanol upto mark to


prepare 25-125g/ml solutions.

Preparation of Sample Solution


Twenty tablets were weighed accurately and one tablet
was calculated. The tablets were crushed to furnish a
homogenous powder and quantity equivalent to 50 mg of
DIC, 250 mg of CHL and 325 mg of PCM was transferred to
a 100 ml volumetric flask. The powder was dissolved in 60
ml of methanol with sonication for 15 minutes and volume
was made up with methanol. 1ml of the solution was

Preparation of Standard Working Solution of PCM


From stock solution (3250g/ml), an aliquot quantity 1, 2,
3, 4, and 5 ml were transferred into 100 ml volumetric flask
and final volume was adjusted with methanol upto mark to
prepare 32.5-162.5 g/ml solutions.

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE

P
e
a
k
a
r
e
a

1000000
900000
800000
700000
600000
500000
400000
300000
200000
100000
0

y = 5051.4x - 14453
R = 0.9997

50

100

150

200

Concentration

Figure 7: Forced degradation under acidic condition

Figure 6: Calibration curve of PCM

Figure 8: Forced degradation under alkaline condition

Figure 9: Forced degradation under oxidative condition

Figure 10: Forced degradation under thermal condition

Figure 11: Forced degradation under neutral condition

transferred into 10ml volumetric flask and diluted upto


mark with methanol.

Method Validation
System Suitability Parameters
System suitability parameter was established to ensure
that the validity of the analytical method was maintained
whenever used. Typical variations are the stability of
analytical solution, different equipment, and different
analyzer. In case of liquid chromatography typical
variations are composition of mobile phase, different lots
or supplier of columns, temperature and flow rate. The
parameters like retention time, asymmetric factor, number
of theoretical plates and tailing factors were evaluated for
DIC, CHL and PCM. The results of system suitability
parameters are shown in Table 1.

Selection of Detection Wavelength


Solutions of DIC, CHL and PCM were scanned between 200
and 400nm. UV spectra of all three drugs show maximum
absorbance at 280nm (Figure 2 Overlay UV spectra of DIC,
CHL and PCM).
Selection of Mobile Phase
Different mobile phases were tried and 20L of mixed
standard solution was injected. The mobile phase
consisted of methanol: phosphate buffer pH 4.0(pH
adjusted with glacial acetic acid) in the ratio of 70:30,
v/v was selected respectively was selected as it gave
well resolved sharp peaks with reasonable retention
time for all the drugs. The analysis was carried out using
Younglin series LC-10ATVP binary gradient system,
Varian 5m C-18 column (250x4.6 mm) with flow rate
0.8 mL/min. (Figure 3 A typical Chromatogram for DIC,
CHL and PCM).

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

Linearity and Calibration Graph


The linearity was evaluated by linear regression analysis.
The series of dilution ranging from 5-25g/ml for DIC, 25125g/ml for CHL and 32.5-182.5g/ml for PCM was
prepared and performed for linearity. All the solutions
were filtered through 0.2 m membrane filter and injected,
chromatograms were recorded and it was repeated for six

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
Acidic Degradation of PCM
Accurately weighed PCM (10mg) was transferred into
100ml volumetric flask and dissolved in 5ml methanol.
50ml of 5N HCl was added and refluxed at 85C for 24
hours. 3.25ml of the solution was diluted upto 10ml with
methanol (32.5g/ml) was analysed by HPLC.

times. The results of linearity and calibration curve are


shown in Table 2. (Figure 4 Calibration curve of DIC, Figure
5 Calibration curve of CHL, and Figure 6 Calibration curve
of PCM).
Recovery Studies
The recovery studies were performed to validate the
accuracy of developed method. To a pre analysed sample
solution, a definite concentration of standard drug was
added and recovery was studied. The results of recovery
studies are shown in Table 3.
Table 1 Results of system suitability parameters, Table
2 Linearity and calibration curve, Table 3 Results of
recovery studies.

Alkaline Degradation
Alkaline Degradation of DIC
Accurately weighed DIC (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 0.1N NaOH was added and refluxed at 80C for 6
hours. 0.5ml of the solution was diluted upto 10ml with
methanol (5g/ml) and analysed by HPLC.

Precision
The precision of the procedure was determined by
repeatability (intraday). Intraday precision was evaluated
by assaying same concentration and during the same day.
Repeatability of sample measurement was carried out in
six different sample preparations from same homogenous
blend of sample. Another replicate determination on three
different days to estimate interday precision.

Alkaline Degradation of CHL


Accurately weighed CHL (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 0.1 N NaOH was added and refluxed at 85C for 2
hours. 2.5ml of the solution was diluted upto 10ml with
methanol (25g/ml) and analysed by HPLC.
Alkaline Degradation of PCM
Accurately weighed PCM (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50 ml of 5N NaOH was added and refluxed at 85C for 24
hours. 3.25ml of the solution was diluted upto 10ml with
methanol. The solution (32.5g/ml) was analysed by
HPLC.

Limit of Detection and Limit of Quantification


For HPLC method, the limit of detection (LOD) and limit of
quantification (LOQ) were calculated based on the standard
deviation of the response and the slope by using calibration
curves.
Robustness
For the HPLC method, robustness was determined by
analysis of the samples under a variety of conditions
making small changes in the percentage of mobile phase
compounds (phosphate buffer: methanol in the ratios
68:32 and 42:58), in the flow rate (0.6 and 1.2 ml/min), in
the temperature conditions (35 and 45C), and changing
the wavelength (268 and 276 nm).

Oxidative Degradation
Oxidative Degradation of DIC
Accurately weighed DIC (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 3% H2O2 was added and kept at room temperature
for 6 hours. 0.5ml of the solution was diluted upto 10ml
with methanol (5g/ml) and analysed by HPLC.
Oxidative Degradation of CHL
Accurately weighed CHL (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 30% H2O2 was added and kept at room
temperature for 4 hours. 2.5ml of the solution was diluted
upto 10ml with methanol (25g/ml) and analysed by HPLC.

Forced Degradation Study (21-26)


Forced degradation for all three drugs was carried out
under conditions of acid/base/neutral hydrolysis, oxidation,
and dry heat. For each study, six samples were prepared and
injected then study was extended up to the formulation.
Acidic Degradation
Acidic Degradation of DIC
Accurately weighed (10mg) of DIC was, transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 0.1N HCl was added and refluxed at 80C for 6
hours. 0.5ml of the solution was diluted upto 10ml with
methanol (5g/ml) and analysed by HPLC.

Oxidative Degradation of PCM


Accurately weighed PCM (10mg) was, transferred into
10ml volumetric flask and dissolved in 5ml methanol. 3.25
ml of 30% H2O2 was added and kept at room temperature
for 24 hours. The solution (32.5g/ml) was analysed by
HPLC.
Thermal Degradation
Thermal Degradation of DIC
Accurately weighed DIC (10mg) was kept at 105C for 8
hours and transferred in 100ml volumetric flask. Dissolved
in 50ml of methanol then dilute upto mark with methanol.
0.5 ml of the solution was diluted upto 10ml with methanol
(5g/ml) and analysed by HPLC.

Acidic Degradation of CHL


Accurately weighed CHL (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 5N HCl was added and refluxed at 85C for 24
hours. 2.5 ml of the solution was diluted upto 10ml with
methanol (25g/ml) and analysed by HPLC.

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
for 6 hours. 1ml of the solution was diluted upto 100ml
with methanol and analysed by HPLC.

Thermal Degradation of CHL


Accurately weighed CHL (10mg) was kept at 105C for 8
hours and transferred in 100ml volumetric flask. Dissolved
in 50ml of methanol then dilute upto mark with methanol.
2.5 ml of the solution was diluted upto 10ml with methanol
(25g/ml) and analysed by HPLC.

Thermal Degradation of Marketed Product


Twenty tablets were weighed accurately and finely
powdered. Powder exactly equivalent to 50mg of DIC,
250mg of CHL and 325mg of PCM was kept at 105C for 8
hours and transferred in 100ml volumetric flask. The
powder was dissolved in 50ml of methanol and diluted
upto mark with methanol. 1ml of the solution was diluted
upto 100ml with methanol and analysed by HPLC.

Thermal Degradation of PCM


Accurately weighed PCM (10mg) was kept at 105C for 8
hours and transferred in 100ml volumetric flask. Dissolved
in 5ml of methanol then dilute upto mark with methanol
(32.5g/ml) and analysed by HPLC.

Neutral Degradation of Marketed Product


Twenty tablets were weighed accurately and finely
powdered. Powder exactly equivalent to 50mg of DIC,
250mg of CHL and 325mg of PCM was transferred to a
100ml volumetric flask and dissolved in 50ml methanol.
50ml of water was added and refluxed at 80C for 6 hours.
1ml of the solution was diluted upto 100ml with methanol
and analysed by HPLC.

Neutral Degradation
Neutral Degradation of DIC
Accurately weighed DIC (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of water was added and refluxed at 80C for 6 hours.
0.5ml of the solution was diluted upto 10ml with methanol
(5g/ml) and analysed by HPLC.

RESULTS AND DISCUSSION


The mobile phase consisting of methanol: water (80:20,
v/v) at 0.8ml/min flow rate which gave sharp, wellresolved peak with minimum tailing factor for DIC, CHL and
PCM. The retention time for all three drugs was tR 3.25min
for DIC, tR 5.60 min for CHL and tR 9.40 min for PCM at
wavelength 280nm. The calibration curve for DIC, CHL and
PCM was found to be linear over the range of 5-25Pg/ml for
DIC, 25-125g/ml for CHL and 32.5-162.5 Pg/ml for PCM.
The linearity was found to be 0.9998, 0.9997 and 0.9997
for DIC, CHL and PCM respectively. The proposed method
was successfully applied to the determination of DIC, CHL
and PCM in market formulation. The %RSD of intraday
and interday precision study for DIC, CHL and PCM were
found to be <2 as shown in Table 4. The LOD for DIC, CHL
& PCM was found to be 0.15g/ml, 1.82g/ml and
2.40g/ml respectively, while LOQ were 0.47g/ml,
5.53g/ml and 7.29g/ml respectively as shown in Table
5. The summary of all validated parameters is shown in
Table 6. The summary of method validation and analysis
of marketed formulation is shown in Table 7 and Table 8
respectively.
Table 4 Results of Intraday & Inter day precision, Table
5 Robustness data for DIC, CHL and PCM, Table 6 Limit of
detection and limit of quantification, Table 7 Summary of
validation parameters, Table 8 Analysis of marketed
formulation of the product.
The degradation study indicated that DIC was
susceptible to base and H2O2 under experimental
conditions. Moreover, CHL was not stable towards acidic,
oxidative and neutral degradation conditions. It was
noticed that except oxidation PCM found to stable for all
the experimental conditions. The study revealed that DIC
and PCM showed no degradation in 0.1 N HCl when reflux
at 80c for 10hr condition and chromatogram showed no
additional peak.
Figure 7 Forced degradation under acidic condition,
Figure 8 Forced degradation under alkaline condition.

Neutral Degradation of CHL


Accurately weighed CHL (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of water was added and refluxed at 85C for 24 hours.
2.5ml of the solution was diluted upto 10ml with methanol
(25g/ml) and analysed by HPLC.
Neutral Degradation of PCM
Accurately weighed PCM (10mg) was transferred into
100ml volumetric flask and dissolved in 50ml methanol.
50ml of water was added and reflux at 85C for 24 hours.
The solution (32.5g/ml) was analysed by HPLC.
Forced Degradation Study of Marketed Product
Acidic Degradation of Marketed Product
Twenty tablets were weighed accurately and finely
powdered. Powder exactly equivalent to 50mg of DIC,
250mg of CHL and 325mg of PCM were transferred to a
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 0.1N HCl was added and refluxed at 80C for 6
hours. 1ml of the solution was diluted upto 100ml with
methanol and analysed by HPLC.
Alkaline Degradation of Marketed Product
Twenty tablets were weighed accurately and finely
powdered. Powder exactly equivalent to 50mg of DIC,
250mg of CHL and 325mg of PCM was transferred to a
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 0.1N NaOH was added and refluxed at 80C for 6
hours. 1ml of the solution was diluted upto 100ml with
methanol and analysed by HPLC.
Oxidative Degradation of Marketed Product
Twenty tablets were weighed accurately and finely
powdered. Powder exactly equivalent to 50mg of DIC,
250mg of CHL and 325mg of PCM was transferred to a
100ml volumetric flask and dissolved in 50ml methanol.
50ml of 3% H2O2 was added and kept at room temperature

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
6. Gan TJ. Diclofenac: an update on its mechanism of action and
safety profile. Curr Med Res Opin., 26(7): 1715-1731, 2010.
7. ONeil MJ. The Merck Index: An Encyclopedia of chemicals,
drugs and biological, 14th ed., Merck Research Laboratories,
1033, 2006.
8. Weng N. HPLC and CE Methods for Pharmaceutical Analysis.
Journal of Chromatography B Biomedical Analysis, 654:287292, 1994.
9. Patel MG, Parmar RR, Nayak PP, Shah SA. Simultaneous
estimation of Paracetamol and Tolperisone hydrochloride in
tablet by UV spectrophotometric methods. Journal of
Pharmaceutical Science and Bioscientific Research, 2(2):6367, 2012.
10. Girolamo O, Neill M, Wainer IW. A Validated Method for the
Determination of Paracetamol and its Glucuronide and
Sulphate Metabolites in the Urine of HIV/AIDS Patients Using
Wavelength-Switching
UV
Detection.
Journal
of
Pharmaceutical and Biomedical Analysis, 17:1191-1197, 1998.
11. Jadi MK, Narayan UL. UV spectrophotometric and RP-HPLC
method development for simultaneous determination of
paracetaomol and etodolac in pharmaceutical dosage form,
56(1): 169-174, 2009.
12. Yadav AH, Kothapalli LP, Barhate AN, Pawar I, Pawar CR. RPHPLC method for determination of aceclofenac, chlorzoxazone
and paracetamol in bulk and pharmaceutical formulation.
International Journal of Pharma Research and Development,
1(10):1-7, 2009.
13. Biswas A, Basu A. Simultaneous estimation of paracetamol,
chlorzoxazone and diclofenac potassium in pharmaceutical
formulation by RP-HPLC method. International Journal of
Pharma and Bio Sciences, 1(2):1-5, 2010.
14. Dhaneshwar SR, Bhusari VK. Validated HPLC method for
simultaneous quantitation of
diclofenac sodium and
misoprostol in bulk drug and formulation, Pelagia Research
Library Der Chemica Sinica, 1(2):110-18, 2010.
15. Panda SS, Patnaik d, Ravikumar B, New Stability-Indicating
RP-HPLC method for determination of diclofenac potassium
and metaxalone from their combined dosage form, Scientia
Pharmaceutica, Under Press, 2012.
16. Shinde VM, Desai BS, Tendolkar NM. Simultaneous
determination of paracetamol, diclofenac sodium and
chlorzoxazone by HPLC from tablet. Indian Journal of
Pharmaceutical Sciences, 57(1):35-37, 1995.
17. Rao JR, Mulla TS, Bharekar VV, Yadav SS, Rajput MP.
Simultaneous HPTLC determination of paracetamol and
dexketoprofen trometamol in pharmaceutical dosage form.
Der Pharma Chemica, 3(3): 32-38, 2011.
18. Rajput MP, Bharekar VV, Yadav SS, Mulla TS, Rao JR. Validated
HPTLC method for simultaneous estimation of diclofenac
potassium and metaxalone in bulk drug and formulation,
Pharmacie Globale International Journal of Comprehensive
Pharmacy, 12(4):1-4, 2011.
19. Potawale SE, Nanda RK, Bhagwat VV. Hamane SC, Deshmukh S,
Puttamsetti S, Development and validation of a HPTLC method
for simultaneous densitometric analysis of Diclofenac
potassium and Dicyclomine hydrochloride as the bulk drugs
and in the tablet dosage form, Journal of Pharmacy Research,
4(9):3116-18, 2011.
20. Yuvaraj G, Pavan Kumar P, Elangovan N, The Simultaneous
Estimation of Aceclofenac, Paracetamol and Chlorzozazone in
Pharmaceutical Dosage Forms by HPTLC, International
Journal of Chemical and Pharmaceutical Sciences, 1(2):24-27,
2010.
21. ICH, Q2B Validation of Analytical Procedure: Methodology, in:
Proceeding of the International Conference on Harmonization,
Geneva, March 1996.

In alkaline hydrolysis DIC and CHL degraded as


observed by the decreased area in the peak of the drug
when compared with peak area of the same concentration
of the non-degraded drug, with giving additional
degradation peak. In oxidative degradation all the three
drugs were observed to be labile and 9% of degradation for
DIC was found. There was no degradation found under
thermal and neutral conditions for DIC, CHL and PCM.
(Figure 9 Forced degradation under oxidative condition,
Figure 10 Forced degradation under thermal condition &
Figure 11 Forced degradation under neutral condition).
Percent degradation was calculated by comparing the areas
of the degraded peaks in each degradation condition with
the corresponding areas of the peaks of the drug under non
degradation condition. The summary of degradation
studies is given in Table 9.
CONCLUSION
It was possible in this study to develop a stability indicating
LC assay method for simultaneous estimation of DIC, CHL
and PCM by subjecting the drugs to ICH recommended
stress conditions. The drugs and degradation products got
well separated from each other in an isocratic mode using a
reversed phase. The developed method was found to be
accurate, precise sensitive, selective and repeatable for
analysis of diclofenac potassium, chlorzoxazone
paracetamol in market formulation without any
interference from the excipients. The method was
successfully used for determination of drugs in a
pharmaceutical formulation. It was possible in this study to
develop a stability indicating assay method for the drugs by
subjecting ICH recommended stress conditions. The drugs
and degradation products got well separated from each
other in isocratic mode using a reversed phase C18 column
and mobile phase composed of methanol: phosphate buffer
(70:30, v/v) at 0.8ml/min flow rate and the eluent was
monitored at 280nm.The results indicated suitability of this
method to study stability of three drugs under various
forced degradation conditions like acid, base, dry heat and
oxidative degradation. There was no interference observed
due to excipients or other components present in tablet
dosage form. The developed method is stability indicating
and separate degradants and can be used to determine the
stability of samples.
REFERENCES AND NOTES
1. Wan J, Ernstgard L, Song BJ, Shoaf SE. Chlorzoxazone
metabolism is increased in fasted Sprague-Dawley rats. J
Pharm Pharmacology, 58(1): 51-61, 2006.
2. Park JY, Kim KA, Park PW, Ha JM. Effect of high-dose aspirin on
CYP2E1 activity in healthy subjects measured using
Chlorzoxazone as a probe. J Clinical Pharmacology, 46(1):109114, 2006.
3. Sweetman SC. Martindale: The Complete Drug Reference, 37th
ed. London: Pharmaceutical Press; 32-33, 2011.
4. Solomon DH, Avorn J, Sturmer T, Glynn RJ, Mogun H,
Schneeweiss S. Cardiovascular outcomes in new users of
coxibs and nonsteroidal antiinflammatory drugs: high-risk
subgroups and time course of risk. Arthritis Rheum.,
54(5):1378-1389, 2006.
5. FitzGerald GA, Patrono C. The coxibs selective inhibitors of
cyclooxygenase-2. N Engl J Med., 345(6):433-442, 2000.

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

RESEARCH ARTICLE
Acknowledgments
The authors are greaful to Zydus Cadila Ltd for providing gift
samples of diclofenac potassium, chlorzoxazone and
paracetamol respectively.

22. ICH, Guidance on Analytical Method Validation, in:


Proceedings of International Convention on Quality for the
Pharmaceutical Industry, Toronto, Canada, September 2002.
23. Bakshi M, Singh S, Development of validated stabilityindicating assay methods-critical review, Journal of
Pharmaceutical and Biomedical Analysis, 28:1010-1040, 2002.
24. Rhodes CT, Introductory overview, Drug Stability: Principle
and practices, Marcel Dekker, New York, 1-12, 2000.
25. Matthews BR, Regulatory aspects of Stability Testing in Europe
in: Drug Stability: Principles and Practices, Carstensen JT and
Rhodes C T, 3rd ed., Marcel Dekker, New York, 107: 580, 2000.
26. Hong DD, Shah M, Development and validation of HPLC
stability-indicating assays.in: Drug Stability: Principles and
Practices, J.T. Carstensen and C.T. Rhodes, 3rd ed., Marcel
Dekker, New York, 107:329-384, 2000.

Inventi Rapid: Pharm Analysis & Quality Assurance Vol. 2012, Issue 4
[ISSN 0976-3813]

Cite this article as: Maulikkumar R Amin, Paresh U


Patel, B N Suhagia, Madhabhai M Patel. Development
and Validation of Stability Indicating Method for
Determination of Chlorzoxazone, Diclofenac Potassium
and Paracetamol in Pharmaceutical Dosage form using
High Performance Liquid Chromatography. Inventi
Rapid: Pharm Analysis & Quality Assurance, 2012(4): 19, 2012.

2012 ppaqa 543, CCC: $10 Inventi Journals (P) Ltd


Published on Web 08/10/2012, www.inventi.in

Вам также может понравиться