01.06.2013 Views

Ph. D Thesis - Higher Education Commission

Ph. D Thesis - Higher Education Commission

Ph. D Thesis - Higher Education Commission

SHOW MORE
SHOW LESS

Create successful ePaper yourself

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

<strong>Ph</strong>. D <strong>Thesis</strong><br />

QUANTIFICATION OF ASPIRIN, BRUFEN, DICLOFEN<br />

AND PARACETAMOL IN HUMAN BODY FLUIDS BY<br />

VARIOUS ANALYTICAL TECHNIQUES<br />

A THESIS SUBMITTED TOWARDS THE PARTIAL FULFILLMENT OF<br />

THE REQUIREMENT FOR THE AWARD OF DOCTOR OF PHILOSOPHY<br />

IN ANALYTICAL CHEMISTRY<br />

ABDUL RAUF KHASKHELI<br />

National Centre of Excellence in Analytical Chemistry,<br />

University of Sindh, Jamshoro - PAKISTAN<br />

2011


DEDICATED<br />

To My Affectionate Supervisors, Beloved Parents and Friends<br />

who’s Prayers, Encouragement and Cooperation Enabled Me for<br />

this Achievement<br />

i


Certificate<br />

This is to certify that Mr. ABDUL RAUF KHASKHELI has carried out his research<br />

work on the topic “QUANTIFICATION OF ASPIRIN, BRUFEN, DICLOFEN AND<br />

PARACETAMOL IN HUMAN BODY FLUIDS BY VARIOUS ANALYTICAL TECHNIQUES”<br />

under our supervision at the laboratories of National Centre of Excellence in Analytical<br />

chemistry, University of Sindh, Jamshoro, Pakistan and UNESCO laboratory of<br />

Environmental Electrochemistry, Department of Analytical Chemistry, Charles<br />

University, Prague, Czech Republic under the abroad <strong>Ph</strong>.D split program (IRSIP) of<br />

<strong>Higher</strong> <strong>Education</strong> <strong>Commission</strong>, Pakistan. The work reported in this thesis is original and<br />

distinct. His dissertation is worthy of presentation to the University of Sindh for the award<br />

of degree of Doctor of <strong>Ph</strong>ilosophy in Analytical Chemistry.<br />

Dr. Sirajuddin<br />

Associate Professor<br />

Supervisor<br />

National Centre of Excellence in Analytical<br />

Chemistry, University of Sindh, Jamshoro, Pakistan<br />

Dr. Syed Tufail Hussain Sherazi<br />

Associate Professor<br />

Co-Supervisor<br />

National Centre of Excellence in Analytical<br />

Chemistry, University of Sindh, Jamshoro, Pakistan<br />

ii


ACKNOWLEDGEMENTS<br />

All praises be to Almighty Allah (The Most Merciful, The Most Gracious and The Most<br />

Compassionate) Who is the entire and only source of every knowledge, Who guides me in<br />

the obscurity and helps me in difficulties, and His Prophet Hazrat Mohammad Mustafa<br />

(Salallah-o-Alaihe Wasallim) whose teachings provide the spirit of learning the hidden and<br />

unconcealed facts of nature.<br />

I am highly grateful to my supervisor Dr. Sirajuddin (Associate Professor) and Cosupervisor<br />

Dr. Syed Tufail Hussain Sherazi (Associate Professor) for their genius and<br />

superb ideas, support and untiring efforts for my appreciation during my studies that enabled<br />

me to complete my research work successfully and truly made my research very fruitful and<br />

of high quality.<br />

I would like to extend my gratitude to Prof. Dr. Muhammad Iqbal Bhanger Director,<br />

NCEAC University of Sindh Jamshoro, Pakistan for his sincere advices and facilities to<br />

carry out this research work successfully.<br />

I am particularly indebted to my teachers, Prof. Dr. Tasneem Gul Kazi, Dr. Shahabuddin<br />

Memon, Dr. Najma Memon, Dr. Amber Rehana Solangi, Dr. Farah Naz Talpur, Dr.<br />

Aamna Baloch, Dr. Hassan Imran Afridi and Engr. Mairaj Ahmed Noorani who always<br />

offered their professional skills whenever needed. I am grateful for their encouraging<br />

attitude in the solution of problems faced during the course of my research work.<br />

I am extremely grateful to Prof. Dr. Jiri Barek and Dr. Jan Fischer for their kind<br />

supervision, excellent research assistance in completing a part of my <strong>Ph</strong>.D research work<br />

and in up grading my electrochemical approach during the course of my study in Czech<br />

Republic, at the UNESCO Laboratory of Environmental Electrochemistry, Department<br />

of Analytical Chemistry, Charles University Prague, under IRSIP by the HEC, Pakistan.<br />

I am also very much grateful to <strong>Higher</strong> <strong>Education</strong> <strong>Commission</strong> of Pakistan, for awarding<br />

me the six months scholarship for research training in Czech Republic.<br />

I would extend my sincere and heartily thanks and appreciation to my friends, fellows and<br />

colleagues Dr. Sarfaraz Ahmed Mahesar, Dr. Afzal Shah, Dr. Abdul Niaz, Dr.<br />

Munawar Saeed Qureshi, Sajidullah Abbasi, Kamran Ahmed Abro, Dr. Ghulam<br />

Abbass Kandhro, Ateeq-ur-Rehman Memon, Nazar Hussain Kalwar,<br />

iii


M. Younis Talpur, Imam Bakhsh Solangi, Jameel Ahmed Baig, Zulfiqar Ali Tagar,<br />

Aijaz Ahmed Bhutto, Yawar Latif and Muhammad Ali.<br />

They always encouraged and cooperated with me and made every possible effort to provide<br />

the useful contribution for the improvement of this study.<br />

I would like to place on record sincere thanks to my teachers as my parents especially, Mr.<br />

Abdul Razaque Abro, Prof. Dr. M. Usman Memon, Prof. Dr. Ubedullah M. Abbasi,<br />

Prof. Dr. Mehboob Ali Rind, Mr. Abdul Hakeem Memon and Mr. Ubed-ur- Rehman<br />

Mughal and Mr. Imran-ul-Haq for their assistance and constructive attitude. I am highly<br />

thankful to Dr. Aftab Ahmed Kandhro and Munawar Ali Soomro for their dear and<br />

useful thoughts, and support for editing and printing during this dissertation.<br />

It seems an obligation upon me to record the word of encouragement for my family and<br />

simple words of thankfulness would not cover the genuine affection, absolute support,<br />

remarkable encouragement and profound prayers of my father Muhammad Bakhsh<br />

Khaskheli, brother Abdul Salam Khaskheli and all family members. I am thankful to all of<br />

them, without their co-operation, it would have been impossible for me to devote so much of<br />

time to this study.<br />

I would also like to thank administrative and supportive staff at National Centre of<br />

Excellence in Analytical Chemistry Pir Ziauddin, Nasrullah Kalhoro, Akhtar Vighio, M.<br />

Mudasir Arain, Pir Siraj, Shafiq A. Bhutto, Jawad Ahmed and Junaid Talpur<br />

ABDUL RAUF KHASKHELI<br />

iv


ABSTRACT<br />

A very fast, economical and simple direct spectrophotometric method was investigated for<br />

Paracetamol (PC) determination in aqueous medium without using any reagent. The method<br />

is based on the photo activation of the analyte at 243 nm after dissolution in water. The<br />

change in structure of PC after addition of water was studied by comparing the<br />

corresponding FTIR spectra. Optimization studies were conducted by using a 5 µg ml -1<br />

standard solution of the analyte. Various parameters studied include, time for stability and<br />

measurement of spectra, effect of HCl, NaOH, CH3COOH and NH3 for change in<br />

absorbance and shift in spectra, interference by some analgesic drugs and some polar<br />

solvents and temperature effect. After optimization, Beer’s law was obeyed in the range of<br />

0.3–20 µg ml -1 PC solution with a correlation coefficient of 0.9999 and detection limit of 0.1<br />

µg ml -1 for 3/1 S/N ratio. The newly developed method was successfully applied for PC<br />

determination in some locally available tablets and urinary samples. The proposed method is<br />

very useful for quick analysis of various types of solid and liquid samples containing PC.<br />

Another spectrophotometric work describes a simple, sensitive, rapid and economical<br />

analytical procedure for direct spectrophotometric evaluation of Diclofenac Sodium (DS)<br />

using aqueous medium without using a chemical reagent. Parameters like time, temperature,<br />

acidic and basic conditions and interference by analgesic drugs was studied for a 5µg ml -1<br />

solution of DS at 276 nm. Under optimized parameters, a linear working range of 0.1–30 g<br />

ml -1 with regression coefficient of 0.9998 and lower detection limit of 0.01 g ml -1 was<br />

obtained. The method was applied for DS contents in tablets, serum and urine samples.<br />

v


A new method was developed for the determination of paracetamol by differential pulse<br />

voltammetry (DPV) at carbon film electrode (CFE). The experimental parameters, such as<br />

pH of Britton-Robinson buffer and potentials for regeneration of electrode surface were<br />

optimized. Under optimized conditions in Britton-Robinson buffer pH 4.0 a linear<br />

calibration curve was obtained in the range 0.02–100 μmol L -1 . The limit of determination<br />

was 0.034 μmol L -1 which showed high sensitivity of developed method. The method was<br />

applied for the quantitative determination of Paracetamol in pharmaceutical formulations as<br />

well as urine samples.<br />

A rapid, reliable and economical analytical procedure for the estimation of ibuprofen in<br />

pharmaceutical formulations and human urine samples was developed using transmission<br />

Fourier Transform Infrared (FT-IR) spectroscopy. For the determination of ibuprofen, a KBr<br />

window with 500 µm spacer was used to acquire the FT-IR spectra of standards,<br />

pharmaceuticals as well as urine samples. The Partial Least Squares (PLS) calibration model<br />

was developed based on carbonyl region (C=O) from 1807-1461 cm −1 in the range from 10-<br />

1000 ppm. The developed model was checked by cross-validation steps to diminish standard<br />

error of the models, such as root mean square error of calibration (RMSEC), root mean<br />

square error of cross validation (RMSECV) and root mean square error of prediction<br />

(RMSEP). The good coefficient of determination (R 2 ) was achieved 0.999 with minimum<br />

standard errors RMSEC, RMSECV and RMSEP 1.89, 1.956 and 1.38, respectively.<br />

The other method was based on indirect determination of acetylsalicylic acid (aspirin)<br />

utilizing differential pulse voltammetry at carbon film electrode as working electrode. The<br />

theory of indirect determination of ASA is based on the hydrolysis of aspirin in salicylic<br />

vi


acid (SA) for detection. Moreover, we optimized conditions such as pH of Britton-Robinson<br />

buffer, potentials for regeneration and activation of electrode surface, amplitude and scan<br />

rate. Under optimized conditions in Britton-Robinson buffer pH 2.0 a linear calibration<br />

curve was obtained in the range 0.2 – 100 µmol L -1 . The limit of determination was 0.15<br />

µmol L -1 which showed high sensitivity of developed method. The method for indirect<br />

determination of ASA was thus developed for the quantification of pharmaceutical<br />

formulations as well in human urine model samples.<br />

vii


List of Contents<br />

Dedication i<br />

Certificate ii<br />

Acknowledgement iii<br />

Abstract v<br />

List of Contents viii<br />

List of Tables xii<br />

List of Figures xiv<br />

Abbreviations xviii<br />

Chapter - One INTRODUCTION<br />

1.1. Analgesics / NSAIDs -An overview 1<br />

1.1.1. Mechanism of Action (NSAIDs) 2<br />

1.2. Paracetamol 3<br />

1.2.1. <strong>Ph</strong>armacopoeias 3<br />

1.2.2. Uses and Administration 4<br />

1.2.3. Adverse Effects 4<br />

1.2.4. Overdosage and its Treatment 5<br />

1.2.5. Precautions 5<br />

1.3. Aspirin 6<br />

1.3.1. <strong>Ph</strong>armacopoeias 6<br />

1.3.2. Uses and Administration 7<br />

1.3.3. Adverse Effects 8<br />

1.3.4. Overdosage and its Treatment 9<br />

1.3.5. Precautions 9<br />

1.4. Diclofen 10<br />

1.4.1. <strong>Ph</strong>armacopoeias 10<br />

1.4.2. Uses and Administration 10<br />

1.4.3. Adverse Effects 11<br />

1.4.4. Precautions 12<br />

1.5. Brufen 12<br />

1.5.1. <strong>Ph</strong>armacopoeias 12<br />

1.5.2. Uses and Administration 12<br />

1.5.3. Adverse Effects 13<br />

1.5.4. Overdosage and its Treatment 14<br />

1 – 14<br />

viii<br />

viii


Chapter - Two<br />

LITERATURE REVIEW<br />

15 – 42<br />

2.1. Analytical Techniques for Assessment of NSAIDs 15<br />

2.2. Electrochemical Techniques 16<br />

2.3. Spectroscopic Methods 21<br />

2.4. Chromatographic Methods 25<br />

2.5. Other Techniques 27<br />

2.6. Quantification of Paracetamol in Biological Samples and<br />

<strong>Ph</strong>armaceuticals<br />

27<br />

2.7. Quantification of Aspirin in <strong>Ph</strong>armaceutical and Biological<br />

Samples<br />

31<br />

2.8. Quantification of Diclofenac Sodium in <strong>Ph</strong>armaceutical and<br />

Biological Samples<br />

35<br />

2.9. Quantification of Ibuprofen in <strong>Ph</strong>armaceutical and<br />

Biological Samples<br />

39<br />

Chapter - Three EXPERIMENTAL 43 – 53<br />

3.1. Material and Methods for Paracetamol Using UV-Visible<br />

Spectrophotometry<br />

43<br />

3.1.1. Reagents and Chemicals 43<br />

3.1.2. Instruments and Apparatus 43<br />

3.1.3. Procedure for Determining PC 44<br />

3.1.4. Analysis of PC in Urine Samples 44<br />

45<br />

3.2. Material and Methods for Diclofenac Sodium Using UV-<br />

Visible Spectrophotometry<br />

3.2.1. Apparatus 45<br />

3.2.2. Washing of Glassware 45<br />

3.2.3. Reagents and Solutions 45<br />

3.2.4. Procedure for Determining DS in Tablets 46<br />

3.2.5. Procedure for DS in Serum and Urine Samples 46<br />

3.3. Material and Methods for the Determination of<br />

Paracetamol Using Differential Pulse Voltammetry<br />

47<br />

3.3.1. Reagents and Solutions 47<br />

3.3.2. Apparatus 47<br />

3.3.3. Procedures 48<br />

49<br />

3.4. Material and Methods for the Analysis of Ibuprofen<br />

Using FT-IR Spectroscopy<br />

3.4.1. Reagents and Samples 49<br />

3.4.2. FT-IR Spectral Measurements 49<br />

ix


3.4.3. FT-IR Calibrations 50<br />

3.4.4. Sample Preparation Procedure 50<br />

3.4.5. Collection and Preparation of Urine Samples 50<br />

3.5. Material and Methods for Investigation of Aspirin Using<br />

Voltammetry<br />

51<br />

3.5.1. Reagents 51<br />

3.5.2. Apparatus 51<br />

3.5.3. Voltammetric Procedure 52<br />

3.5.4. Indirect Determination of ASA 52<br />

3.5.5. Indirect Determination of ASA in <strong>Ph</strong>armaceutical<br />

Drugs and Urine Samples by DPV at CFE<br />

52<br />

Chapter - Four Result and Discussions 54 - 108<br />

4.1. Simpler Spectrophotometric Assay of PC in Tablets and<br />

Urine Samples<br />

54<br />

54<br />

4.1.1. Effect of Water Addition to PC (FTIR studies)<br />

4.1.2. Optimization of Time for Measurement and Stability<br />

of Analytical Signal<br />

55<br />

4.1.3. Effect of Temperature 56<br />

4.1.4. Effect of Polar Solvents 57<br />

4.1.5. Interference by Various Analgesic Drugs 58<br />

4.1.6. Effect of Acidic and Basic Solutions 60<br />

4.1.7. Calibration Range 61<br />

4.1.8. Analysis of Tablets 62<br />

4.1.9. Analysis of Urine Samples 63<br />

4.2. Simpler and Faster Spectrophotometric Determination of<br />

Diclofenac Sodium (DS) in Tablets, Serum and Urine<br />

Samples<br />

65<br />

4.2.1. Influence of Time 65<br />

4.2.2. Effect of Temperature 66<br />

4.2.3. Interference by Various Analgesic Drugs 67<br />

4.2.4. Effect of Acidic and Alkaline Conditions 68<br />

4.2.5. Calibration Plot 71<br />

4.2.6. Comparison with Other Reported Spectroscopic<br />

Methods<br />

71<br />

4.2.7. Analysis of Tablets 72<br />

4.2.8. Analysis of Urine and Serum Samples 74<br />

4.3. Differential Pulse Voltammetric Determination of PC in<br />

Tablet and Urine Samples at Carbon Film Electrode<br />

4.3.1. Influence of pH on PC at DC and DP Voltammetry 77<br />

77<br />

x


4.3.2. Optimization of Parameters and Calibration Curve 78<br />

4.3.3. Analysis of <strong>Ph</strong>armaceutical Drugs 82<br />

4.3.4. Analysis of Urine Samples 84<br />

4.4. Quantification of Ibuprofen in <strong>Ph</strong>armaceuticals and<br />

Biological Samples by FTIR Transmission Spectroscopy.<br />

4.4.1. Analysis of <strong>Ph</strong>armaceutical Samples<br />

86<br />

4.4.2. Analysis of Urine Samples 89<br />

4.4.3. Limits of Detection and Quantitation 92<br />

4.5. Differential Pulse Voltammetric Determination of<br />

Salicylic acid and Acetylsalicylic acid in Tablet and Urine<br />

Samples at Carbon Film Electrode<br />

93<br />

93<br />

4.5.1. Influence of pH on Salicylic Acid in DC and DP<br />

Voltammetry<br />

4.5.2. Reproducibility of Salicylic Acid 94<br />

4.5.3. Linear Calibration Curves of Salicylic Acid 95<br />

4.5.4. Hydrolysis of Acetylsalicylic acid 97<br />

4.5.5. Linear Calibration Curves of Hydrolyzed<br />

Acetylsalicylic Acid at CFE<br />

98<br />

4.5.6. Determination of Salicylic Acid in Different Drugs 100<br />

4.5.7. Determination of Acetylsalicylic Acid from Different<br />

Drugs<br />

101<br />

4.5.8. Determination of Salicylic Acid in Urine Samples at<br />

CFE<br />

103<br />

4.5.9. Determination of Acetylsalicylic Acid in Urine at<br />

CFE<br />

104<br />

4.5.10. Interference Study 106<br />

4.6. Conclusion 107<br />

References 109-132<br />

86<br />

xi


LIST OF TABLES<br />

Table. 4.1.1. Effect of different polar solvents on absorbance of PC.<br />

Table. 4.1.2. % interference by various analgesics in different ratios on<br />

5 µg ml -1 PC<br />

59<br />

Table. 4.1.3. Effect of strong and weak acids and bases on PC<br />

determination 60<br />

Table. 4.1.4. Determination of PC in tables of various companies by<br />

proposed method<br />

63<br />

Table. 4.1.5. Comparative analyses of PC in urine samples. 63<br />

Table. 4.2.1. % interference by various analgesics in different ratios on<br />

5 µg ml -1 DS<br />

67<br />

Table. 4.2.2. Effect of strong and weak acids and bases on DFS<br />

determination<br />

70<br />

Table. 4.2.3. Comparison of current method with other spectroscopic<br />

methods for determination of DS<br />

72<br />

Table. 4.2.4. Determination of DS in tablets of various companies by<br />

proposed method<br />

73<br />

Table. 4.2.5. Concentration of determined DS and its relation with<br />

ingested DS by oral administration<br />

76<br />

Table 4.3.1. The influence of pH on DCV and DPV of Paracetamol<br />

(c = 100 µmol L -1 )<br />

Table. 4.3.2. Parameters of the calibration straight lines for the<br />

78<br />

determination of Paracetamol in Britton-Robinson buffer<br />

pH 4.0 using DPV at CFE with regeneration potential<br />

Ereg1 = –400 mV, and Ereg2 = 1300 mV.<br />

Table. 4.3.3. The amount of Paracetamol determined by DPV in tablets<br />

79<br />

of commercial drugs with declare contents 500 mg of<br />

Paracetamol.<br />

Table. 4.3.4. Parameters of the calibration straight lines for the<br />

determination of model samples of Paracetamol in urine,<br />

83<br />

media of Britton-Robinson buffer pH 4.0 using DPV at<br />

CFE with regeneration potentials Ereg1 = –400 mV and<br />

Ereg2 = 1300 mV.<br />

84<br />

Table. 4.4.1. Results for the ibuprofen concentration found in the tablet<br />

samples<br />

87<br />

Table. 4.4.2. Recovery result of ibuprofen from tablet samples after<br />

spiking with known concentrations of standards<br />

89<br />

Table. 4.4.3. Recovery results of ibuprofen from urine samples after<br />

spiking with known concentrations of standard<br />

90<br />

Table 4.4.4. Recovery results of ibuprofen in urine samples after<br />

spiking with known concentrations of standard<br />

91<br />

Table. 4.5.1. The influence of pH on DCV and DPV of Salicylic Acid (c<br />

= 100 µmol L -1 )<br />

94<br />

xii


Table 4.5.2. Parameters of the calibration straight lines for the<br />

determination of Salicylic Acid in a Britton-Robinson<br />

buffer pH 2.0 using DPV at CFE with activation potential<br />

2200 mV<br />

Table 4.5.3. Parameters of the calibration straight lines for the<br />

determination of hydrolyzed Acetylsalicylic Acid in a<br />

Britton-Robinson buffer pH 2.0 using DPV at CFE with<br />

activation potential 2200 mV<br />

Table 4.5.4. The amount of Salicylic Acid determined by DPV in<br />

tablets<br />

Table 4.5.5. The amount of hydrolyzed Acetylsalicylic Acid<br />

determined by DPV in tablets<br />

Table 4.5.6. Parameters of the calibration straight lines for the<br />

determination of salicylic acid in 0.1 ml urine samples in a<br />

Britton-Robinson buffer pH 2.0 using DPV at CFE with<br />

activation potential 2200 mV<br />

Table 4.5.7. Parameters of the calibration straight lines for the<br />

determination of hydrolyzed Acetylsalicylic acid in 0.1 ml<br />

urine sample in a Britton-Robinson buffer pH 2.0 using<br />

DPV at CFE with activation potential 2200 mV<br />

97<br />

100<br />

101<br />

102<br />

104<br />

106<br />

xiii


LIST OF FIGURES<br />

Figure. 1.1. Structural formula of Paracetamol (PC) 3<br />

Figure. 1.2. Structural formula of Aspirin (ASA) 6<br />

Figure. 1.3. Structural formula of Diclofenac Sodium (DS) 10<br />

Figure. 1.4. Structural formula of Ibuprofen (IBP) 12<br />

Figure. 4.1.1. FTIR spectra of (A), pure PC (B), aqueous PC paste and<br />

(C) aqueous solution of 5 µgml -1 PC.<br />

55<br />

Figure. 4.1.2. Dependence of absorbance of PC on time 56<br />

Figure. 4.1.3. Effect of temperature on absorbance of PC 57<br />

Figure. 4.1.4. Shift of wavelength with pH: (A) 3.3 and (B) 12. 3 for<br />

5µgml -1 PC solution.<br />

61<br />

Figure. 4.1.5. Calibration range of absorbance vs. concentration for PC<br />

from 0.3 to 20 µg ml -1 .<br />

61<br />

Figure. 4.1.6. A 1000 times diluted sample of urine (a) with PC and (b)<br />

without PC.<br />

64<br />

Figure. 4.2.1. Time effect on absorbance of Diclofenac Sodium 65<br />

Figure. 4.2.2. Temperature effect on UV absorbance of 5 µg ml -1 DS<br />

solution.<br />

66<br />

Figure. 4.2.3. UV spectra of DS at acidic pH, 3.56 (lower) and basic pH,<br />

11.68 (higher).<br />

Figure. 4.2.4. Calibration plot of absorbance verses concentration for DS<br />

69<br />

solutions from below to above as 0.1, 0.5, 1, 2, 3, 4, 5, 6, 7,<br />

8, 9, 10, 15, 20, 25 and 30 µg ml -1 .<br />

71<br />

Figure. 4.2.5. Representative UV-spectrum of expected 5 µg ml -1 DS in<br />

(Diclofen) tablet.<br />

73<br />

Figure. 4.2.6. UV spectra showing blank (black), DS containing (blue)<br />

and DS spiked (red) urine.<br />

74<br />

Figure. 4.2.7. UV spectra showing blank (black), DS containing (blue)<br />

and DS spiked (red) serum.<br />

Figure. 4.3.1. DC voltammograms (A) and DP voltammograms (B) of<br />

Paracetamol (c = 100 μmol<br />

75<br />

-1 .l) at CFE in Britton-Robinson<br />

buffer pH 2 to 12 (numbers above curves correspond to<br />

given pH) without electrode regeneration. Inset is<br />

corresponding dependence of peak potential on the pH.<br />

Figure. 4.3.2. Repetitive measurements of 100 µmol L<br />

77<br />

-1 Paracetamol<br />

using DPV at CFE in Britton-Robinson buffer pH 4.0 with<br />

regeneration potential Ereg1 = –400 mV and Ereg2 = 1300<br />

mV Inset is corresponding dependence of peak current on<br />

the number of scans.<br />

Figure. 4.3.3. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(PC): (1) 0, (2) 20 (3)<br />

40, (4) 60, (5) 80, (6) 100 µmol L<br />

79<br />

-1 . Regeneration<br />

potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding<br />

calibration dependence.<br />

80<br />

xiv


Figure. 4.3.4. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(PC): (1) 0, (2) 2 (3)<br />

4, (4) 6, (5) 8, (6) 10 µmol L -1 . Regeneration potential<br />

Ereg1 = –400 mV and Ereg2 = 1300 mV. Background<br />

current = 1.94 nA. Inset is corresponding calibration<br />

dependence.<br />

Figure. 4.3.2. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(Paracetamol): 0 (1),<br />

81<br />

0.2 (2), 0,4 (3), 0,6 (4), 0,8 (5), 1 µmol L -1 (6).<br />

Regeneration potential Ereg1 = –400 mV and Ereg2 = 1300<br />

mV. Inset is corresponding calibration dependence.<br />

Figure. 4.3.6. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(PC): (1) 0, (2) 0,02<br />

81<br />

(3) 0,04, (4) 0,06, (5) 0,08, (6) 0,1 µmol L -1 . Regeneration<br />

potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding<br />

calibration dependence.<br />

Figure. 4.3.3. Differential pulse voltammograms of Paracetamol 10 µmol<br />

82<br />

L -1 with Ascorbic acid from 10 to 100 µmol L -1<br />

(concentrations are written above curves in plot) at CFE in<br />

Britton-Robinson buffer pH 4.0, regeneration potential<br />

Ereg1 = –400 mV, and Ereg2 = 1300 mV. Inset is<br />

83<br />

corresponding dependence of peak current of<br />

Paracetamol on concentration of Ascorbic acid.<br />

Figure. 4.3.4. Differential pulse voltammograms of 0.1 ml urine with<br />

model sample of Paracetamol (2 – 10 µmol L -1 (A), and<br />

20 – 100 µmol L -1 (B), concentration is written next to<br />

curves in plots) sample at CFE in Britton-Robinson buffer<br />

pH 4.0, regeneration potentials Ereg1 = –400 mV, and Ereg2<br />

= 1300 mV. Inset is corresponding calibration dependence.<br />

84<br />

Figure. 4.4.1. Calibration plot in the range of 10-100 ppm for<br />

pharmaceutical samples<br />

87<br />

Figure. 4.4.2. <strong>Ph</strong>armaceutical Tablet sample and spikes of 30 ppm, 50<br />

ppm, 70 ppm ibuprofen<br />

88<br />

Figure. 4.4.3. Blank urine and 3 spikes of 10, 20, 30 ppm ibuprofen 90<br />

Figure. 4.4.5. Group Spectra of Ibuprofen Standards 92<br />

Figure. 4.5.1. A and B. DC & DP voltammograms of Salicylic Acid<br />

(c = 100 µmol L -1 ) at CFE in Britton-Robinson buffer pH<br />

2 to 12 (numbers in above curves correspond to given pH).<br />

Inset is corresponding dependence of peak potential on the<br />

pH<br />

Figure. 4.5.2. Measurements of 100 µmol L -1 Salicylic Acid using DPV<br />

at CFE in Britton-Robinson Buffer pH 2.0 with activation<br />

potential (1) 2000,(2) 1500 and (3) 2200 mV<br />

93<br />

95<br />

xv


Figure. 4.5.3. Differential pulse voltammograms of Salicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

20 (3) 40, (4) 60, (5) 8,0 (6) 100 µmol L -1 .<br />

Activation of potential=2200mV for 120 sec<br />

Inset is corresponding calibration dependence<br />

Figure. 4.5.4. Differential pulse voltammograms of Salicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2) 2<br />

(3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation of<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure. 4.5.5. Differential pulse voltammograms of Salicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

0.2 (3) 0.4, (4) 0.6, (5) 0.8, (6) 1 µmol L -1 . Activation of<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure.4.5.6. Differential pulse voltammograms of 10 µmol L -1<br />

hydrolyzed Acetylsalicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, (1) Electrolyte, (2) 10 µmol L -1<br />

without hydrolysis (3) 10 µmol L -1 Salicylic Acid (4) 10<br />

µmol L -1 after hydrolysis Acetylsalicylic acid. Activation<br />

potential=2200mV for 120 sec<br />

Figure. 4.5.7. DP voltammograms of hydrolyzed Acetylsalicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(ASA): (1) 0, (2)<br />

20 (3) 40, (4) 60, (5) 80, (6) 100 µmol L -1 . Activation<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure. 4.5.8. DP voltammograms of hydrolyzed Acetylsalicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2. c (ASA): (1) 0, (2) 2<br />

(3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure. 4.5.9. DP voltammograms of hydrolyzed Acetylsalicylic Acid at CFE in<br />

Britton-Robinson buffer pH 2.0, c(ASA): (1) 0, (2) 0.2 (3) 0.4, (4)<br />

0.6, (5) 0.8, (6) 1 µmol L -1 . Activation potential=2200mV for 120<br />

sec. Inset is corresponding calibration dependence<br />

Figure.4.5.10.Differential pulse voltammograms of 4 µmol L -1 Duofilm<br />

sample with spikes of salicylic acid standard at CFE in<br />

Britton-Robinson buffer pH 2.0, c(SA): (1) electrolyte, (2)<br />

expected 4 μmol L -1 salicylic acid in sample of Duofilm<br />

(3) 6 μmol L -1 spike, (4) 8 μmol L -1 spike and (5) 10 μmol<br />

L -1 spike. Activation potential 2200 mV<br />

Figure. 4.5.11. Differential pulse voltammograms of 4 µmol L -1 Aspirin<br />

sample with spikes of hydrolyzed acetylsalicylic acid<br />

standard at CFE in Britton-Robinson buffer pH 2.0, c(SA):<br />

(1) electrolyte, (2) expected 4 μmol L -1 salicylic acid in<br />

sample of Aspirin (3) 6 μmol L -1 spike, (4) 8 μmol L -1<br />

spike and (5) 10 μmol L -1 spike. Activation potential 2200<br />

mV<br />

96<br />

96<br />

97<br />

98<br />

99<br />

99<br />

100<br />

101<br />

102<br />

xvi


Figure. 4.5.12. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

20 (3) 40, (4) 60, (5) 80, (6) 100 µmol L -1 . Activation<br />

potential 2200 mV. Inset is corresponding calibration<br />

dependence<br />

Figure. 4.5.13. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

2 (3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation potential<br />

2200 mV. Inset is corresponding calibration dependence<br />

Figure. 4.5.14. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(ASA): (1) 0,<br />

(2) 20 (3) 40, (4) 60, (5) 80, (6) 100 µmol L -1 . Activation<br />

potential 2200 mV. Inset is corresponding calibration<br />

dependence<br />

Figure. 4.5.15. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(ASA): (1) 0,<br />

(2) 2 (3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation<br />

potential 2200 mV. Inset is corresponding calibration<br />

dependence<br />

Figure.4.5.16.Differential pulse voltammograms of hydrolyzed<br />

acetylsalicylic acid 5 μmolL -1 with ascorbic acid 5-100<br />

µmol L -1 at CFE in Britton-Robinson buffer pH 2.0.<br />

Activation potential 2200 mV<br />

103<br />

103<br />

105<br />

105<br />

106<br />

xvii


List of Abbreviations<br />

AdSV Adsorptive Stripping Voltammetry<br />

AgA–CFE Silver Amalgam Carbon Film Electrode<br />

AgA–PE Silver Amalgam Paste Electrode<br />

AgE Silver Electrode<br />

AOAC Association of Official Analytical Chemists<br />

ASA Acetylsalicylic Acid<br />

ATR Attenuated Total Reflection<br />

AuE Gold Electrode<br />

CFE Carbon Film Electrode<br />

CNS Central Nerves System<br />

COX Cyclo-Oxygenase<br />

CPE Carbon Paste Electrode<br />

CV Cyclic Voltammetry<br />

DDP Differential Pulse Polarography<br />

DME Dropping Mercury Electrode<br />

DPV Differential Pulse Voltammetry<br />

DS Diclofenac Sodium<br />

E1/2 Half Wave Potential<br />

FIA Flow Injection Analysis<br />

FT-IR Fourier Transform Infrared Spectroscopy<br />

FT-NIR Fourier Transform Near-Infrared<br />

GC Gas Chromatography<br />

GC– MS Gas Chromatography Mass Spectroscopy<br />

GCE Glassy Carbon Electrode<br />

GGE Glassy Graphite Electrode<br />

HEC <strong>Higher</strong> <strong>Education</strong> <strong>Commission</strong><br />

HMDE Hanging Mercury Drop Electrode<br />

HPLC High Performance Liquid Chromatography<br />

HT-GLC High-Temperature Gas–Liquid Chromatography<br />

Hz Hertz<br />

IBP Ibuprofen<br />

LOD Limit of Detection<br />

LOQ Limit of Quantification<br />

LSV Linear Sweep Voltammetry<br />

mg/kg Milligram per Kilogram<br />

mg/kg /b.w/day Milligram per Kilogram per Body Weight per Day<br />

mg/L Milligram per Liter<br />

mm Millimeter<br />

mol.L -1 Mole per liter<br />

mV Milli volt<br />

mV s -1 Milli volt per second<br />

nA Nano Ampere<br />

NIR Near-Infrared Spectroscopy<br />

nM Nano Molar<br />

NMR Nuclear Magnetic Resonance<br />

NPV Normal Pulse Voltammetry<br />

NSAIDs Non Steroidal Anti-Inflammatory Drugs<br />

PC Paracetamol<br />

xviii


PLS Partial Least Square<br />

ppb parts per billion<br />

ppm parts per million<br />

PtSE Platinum Solid Electrode<br />

PVC Polyvinyl Chloride<br />

RE Reference Electrode<br />

RMSEC Root Mean Standard Error of Calibration<br />

RMSEP Root Mean Square Error of Prediction<br />

RP-HPLC Reserved <strong>Ph</strong>ase-High-Performance Liquid Chromatography<br />

rpm revolutions per minute<br />

RSD Relative Standard Deviation<br />

SA Salicylic Acid<br />

SPME Solid <strong>Ph</strong>ase Microextraction<br />

SWV Square Wave Voltammetry<br />

TLC Thin Layer Chromatography<br />

TOF-MS Time-of-Flight Mass Spectrometry<br />

TQ Turbo Quant<br />

V/s Volt per second<br />

v/v Volume by volume<br />

WE Working Electrode<br />

WHO World Health Organization<br />

μg/kg Microgram per kilogram<br />

µg L -1 Microgram per liter<br />

µg/g Microgram per gram<br />

µL Micro liter<br />

µm Micrometer<br />

µM Micro mole<br />

µmol L -1 Micro mole per liter<br />

xix


List of Contents<br />

Dedication i<br />

Certificate ii<br />

Acknowledgement iii<br />

Abstract v<br />

List of Contents viii<br />

List of Tables xii<br />

List of Figures xiv<br />

Abbreviations xviii<br />

Chapter - One INTRODUCTION 1 - 14<br />

1.1. Analgesics / NSAIDs -An overview 1<br />

1.1.1. Mechanism of Action (NSAIDs) 2<br />

1.2. Paracetamol 3<br />

1.2.1. <strong>Ph</strong>armacopoeias 3<br />

1.2.2. Uses and Administration 4<br />

1.2.3. Adverse Effects 4<br />

1.2.4. Overdosage and its Treatment 5<br />

1.2.5. Precautions 5<br />

1.3. Aspirin 6<br />

1.3.1. <strong>Ph</strong>armacopoeias 6<br />

1.3.2. Uses and Administration 7<br />

1.3.3. Adverse Effects 8<br />

1.3.4. Overdosage and its Treatment 9<br />

1.3.5. Precautions 9<br />

1.4. Diclofen 10<br />

1.4.1. <strong>Ph</strong>armacopoeias 10<br />

1.4.2. Uses and Administration 10<br />

1.4.3. Adverse Effects 11<br />

1.4.4. Precautions 12<br />

1.5. Brufen 12<br />

1.5.1. <strong>Ph</strong>armacopoeias 12<br />

1.5.2. Uses and Administration 12<br />

1.5.3. Adverse Effects 13<br />

1.5.4. Overdosage and its Treatment 14


Chapter - Two LITERATURE REVIEW ix<br />

15 - 42<br />

2.1. Analytical Techniques for Assessment of NSAIDs 15<br />

2.2. Electrochemical Techniques 16<br />

2.3. Spectroscopic Methods 21<br />

2.4. Chromatographic Methods 25<br />

2.5. Other Techniques 27<br />

2.6. Quantification of Paracetamol in Biological Samples and<br />

<strong>Ph</strong>armaceuticals<br />

27<br />

2.7. Quantification of Aspirin in <strong>Ph</strong>armaceutical and Biological<br />

Samples<br />

31<br />

2.8. Quantification of Diclofenac Sodium in <strong>Ph</strong>armaceutical and<br />

Biological Samples<br />

35<br />

2.9. Quantification of Ibuprofen in <strong>Ph</strong>armaceutical and<br />

Biological Samples<br />

39<br />

Chapter - Three EXPERIMENTAL 43 - 53<br />

3.1. Material and Methods for Paracetamol Using UV-Visible<br />

Spectrophotometry<br />

43<br />

3.1.1. Reagents and Chemicals 43<br />

3.1.2. Instruments and Apparatus 43<br />

3.1.3. Procedure for Determining PC 44<br />

3.1.4. Analysis of PC in Urine Samples 44<br />

3.2. Material and Methods for Diclofenac Sodium Using UV-<br />

Visible Spectrophotometry<br />

45<br />

3.2.1. Apparatus 45<br />

3.2.2. Washing of Glassware 45<br />

3.2.3. Reagents and Solutions 45<br />

3.2.4. Procedure for Determining DS in Tablets 46<br />

3.2.5. Procedure for DS in Serum and Urine Samples 46<br />

3.3. Material and Methods for the Determination of<br />

Paracetamol Using Differential Pulse Voltammetry<br />

47<br />

3.3.1. Reagents and Solutions 47<br />

3.3.2. Apparatus 47<br />

3.3.3. Procedures 48<br />

49<br />

3.4. Material and Methods for the Analysis of Ibuprofen<br />

Using FT-IR Spectroscopy<br />

3.4.1. Reagents and Samples 49


3.4.2. FT-IR Spectral Measurements 49<br />

3.4.3. FT-IR Calibrations 50<br />

3.4.4. Sample Preparation Procedure 50<br />

3.4.5. Collection and Preparation of Urine Samples 50<br />

3.5. Material and Methods for Investigation of Aspirin Using<br />

Voltammetry<br />

51<br />

3.5.1. Reagents 51<br />

3.5.2. Apparatus 51<br />

3.5.3. Voltammetric Procedure 52<br />

3.5.4. Indirect Determination of ASA 52<br />

3.5.5. Indirect Determination of ASA in <strong>Ph</strong>armaceutical<br />

Drugs and Urine Samples by DPV at CFE<br />

52<br />

Chapter - Four EXPERIMENTAL 54 - 108<br />

4.1. Simpler Spectrophotometric Assay of PC in Tablets and<br />

Urine Samples<br />

54<br />

4.1.1. Effect of Water Addition to PC (FTIR studies) 54<br />

4.1.2. Optimization of Time for Measurement and Stability<br />

of Analytical Signal<br />

55<br />

4.1.3. Effect of Temperature 56<br />

4.1.4. Effect of Polar Solvents 57<br />

4.1.5. Interference by Various Analgesic Drugs 58<br />

4.1.6. Effect of Acidic and Basic Solutions 60<br />

4.1.7. Calibration Range 61<br />

4.1.8. Analysis of Tablets 62<br />

4.1.9. Analysis of Urine Samples 63<br />

4.2. Simpler and Faster Spectrophotometric Determination of<br />

Diclofenac Sodium (DS) in Tablets, Serum and Urine<br />

Samples<br />

65<br />

4.2.1. Influence of Time 65<br />

4.2.2. Effect of Temperature 66<br />

4.2.3. Interference by Various Analgesic Drugs 67<br />

4.2.4. Effect of Acidic and Alkaline Conditions 68<br />

4.2.5. Calibration Plot 71<br />

4.2.6. Comparison with Other Reported Spectroscopic<br />

Methods<br />

71<br />

4.2.7. Analysis of Tablets 72<br />

4.2.8. Analysis of Urine and Serum Samples 74<br />

4.3. Differential Pulse Voltammetric Determination of PC in 77


4.3.1.<br />

Tablet and Urine Samples at Carbon Film Electrode<br />

Influence of pH on PC at DC and DP Voltammetry 77<br />

4.3.2. Optimization of Parameters and Calibration Curve 78<br />

4.3.3. Analysis of <strong>Ph</strong>armaceutical Drugs 82<br />

4.3.4. Analysis of Urine Samples 84<br />

4.4. Quantification of Ibuprofen in <strong>Ph</strong>armaceuticals and<br />

Biological Samples by FTIR Transmission Spectroscopy.<br />

86<br />

4.4.1. Analysis of <strong>Ph</strong>armaceutical Samples 86<br />

4.4.2. Analysis of Urine Samples 89<br />

4.4.3. Limits of Detection and Quantitation 92<br />

4.5. Differential Pulse Voltammetric Determination of<br />

Salicylic acid and Acetylsalicylic acid in Tablet and Urine<br />

Samples at Carbon Film Electrode<br />

93<br />

4.5.1. Influence of pH on Salicylic Acid in DC and DP<br />

Voltammetry<br />

93<br />

4.5.2. Reproducibility of Salicylic Acid 94<br />

4.5.3. Linear Calibration Curves of Salicylic Acid 95<br />

4.5.4. Hydrolysis of Acetylsalicylic acid 97<br />

4.5.5. Linear Calibration Curves of Hydrolyzed<br />

Acetylsalicylic Acid at CFE<br />

98<br />

4.5.6. Determination of Salicylic Acid in Different Drugs 100<br />

4.5.7. Determination of Acetylsalicylic Acid from Different<br />

Drugs<br />

101<br />

4.5.8. Determination of Salicylic Acid in Urine Samples at<br />

CFE<br />

103<br />

4.5.9. Determination of Acetylsalicylic Acid in Urine at<br />

CFE<br />

104<br />

4.5.10. Interference Study 106<br />

4.6. Conclusion 107<br />

References 109-132


LIST OF TABLES<br />

Table. 4.1.1. Effect of different polar solvents on absorbance of PC. 58<br />

Table. 4.1.2. % interference by various analgesics in different ratios on<br />

5 µg ml -1 PC<br />

59<br />

Table. 4.1.3. Effect of strong and weak acids and bases on PC<br />

determination 60<br />

Table. 4.1.4. Determination of PC in tables of various companies by<br />

proposed method<br />

63<br />

Table. 4.1.5. Comparative analyses of PC in urine samples. 63<br />

Table. 4.2.1. % interference by various analgesics in different ratios on<br />

5 µg ml -1 DS<br />

67<br />

Table. 4.2.2. Effect of strong and weak acids and bases on DFS<br />

determination<br />

70<br />

Table. 4.2.3. Comparison of current method with other spectroscopic<br />

methods for determination of DS<br />

72<br />

Table. 4.2.4. Determination of DS in tablets of various companies by<br />

proposed method<br />

73<br />

Table. 4.2.5. Concentration of determined DS and its relation with<br />

ingested DS by oral administration<br />

76<br />

Table 4.3.1. The influence of pH on DCV and DPV of Paracetamol<br />

(c = 100 µmol L -1 )<br />

Table. 4.3.2. Parameters of the calibration straight lines for the<br />

78<br />

determination of Paracetamol in Britton-Robinson buffer<br />

pH 4.0 using DPV at CFE with regeneration potential<br />

Ereg1 = –400 mV, and Ereg2 = 1300 mV.<br />

Table. 4.3.3. The amount of Paracetamol determined by DPV in tablets<br />

79<br />

of commercial drugs with declare contents 500 mg of<br />

Paracetamol.<br />

Table. 4.3.4. Parameters of the calibration straight lines for the<br />

determination of model samples of Paracetamol in urine,<br />

83<br />

media of Britton-Robinson buffer pH 4.0 using DPV at<br />

CFE with regeneration potentials Ereg1 = –400 mV and<br />

Ereg2 = 1300 mV.<br />

84<br />

Table. 4.4.1. Results for the ibuprofen concentration found in the tablet<br />

samples<br />

87<br />

Table. 4.4.2. Recovery result of ibuprofen from tablet samples after<br />

spiking with known concentrations of standards<br />

89<br />

Table. 4.4.3. Recovery results of ibuprofen from urine samples after<br />

spiking with known concentrations of standard<br />

90


Table 4.4.4. Recovery results of ibuprofen in urine samples after<br />

spiking with known concentrations of standard<br />

Table. 4.5.1. The influence of pH on DCV and DPV of Salicylic Acid (c<br />

= 100 µmol L -1 )<br />

Table 4.5.2. Parameters of the calibration straight lines for the<br />

determination of Salicylic Acid in a Britton-Robinson<br />

buffer pH 2.0 using DPV at CFE with activation potential<br />

2200 mV<br />

Table 4.5.3. Parameters of the calibration straight lines for the<br />

determination of hydrolyzed Acetylsalicylic Acid in a<br />

Britton-Robinson buffer pH 2.0 using DPV at CFE with<br />

activation potential 2200 mV<br />

Table 4.5.4. The amount of Salicylic Acid determined by DPV in<br />

tablets<br />

Table 4.5.5. The amount of hydrolyzed Acetylsalicylic Acid<br />

determined by DPV in tablets<br />

Table 4.5.6. Parameters of the calibration straight lines for the<br />

determination of salicylic acid in 0.1 ml urine samples in a<br />

Britton-Robinson buffer pH 2.0 using DPV at CFE with<br />

activation potential 2200 mV<br />

Table 4.5.7. Parameters of the calibration straight lines for the<br />

determination of hydrolyzed Acetylsalicylic acid in 0.1 ml<br />

urine sample in a Britton-Robinson buffer pH 2.0 using<br />

DPV at CFE with activation potential 2200 mV<br />

91<br />

94<br />

97<br />

100<br />

101<br />

102<br />

104<br />

106


LIST OF FIGURES<br />

Figure. 1.1. Structural formula of Paracetamol (PC) 3<br />

Figure. 1.2. Structural formula of Aspirin (ASA) 6<br />

Figure. 1.3. Structural formula of Diclofenac Sodium (DS) 10<br />

Figure. 1.4. Structural formula of Ibuprofen (IBP) 12<br />

Figure. 4.1.1. FTIR spectra of (A), pure PC (B), aqueous PC paste and<br />

(C) aqueous solution of 5 µgml -1 PC.<br />

55<br />

Figure. 4.1.2. Dependence of absorbance of PC on time 56<br />

Figure. 4.1.3. Effect of temperature on absorbance of PC 57<br />

Figure. 4.1.4. Shift of wavelength with pH: (A) 3.3 and (B) 12. 3 for<br />

5µgml -1 PC solution.<br />

61<br />

Figure. 4.1.5. Calibration range of absorbance vs. concentration for PC<br />

from 0.3 to 20 µg ml -1 .<br />

61<br />

Figure. 4.1.6. A 1000 times diluted sample of urine (a) with PC and (b)<br />

without PC.<br />

64<br />

Figure. 4.2.1. Time effect on absorbance of Diclofenac Sodium 65<br />

Figure. 4.2.2. Temperature effect on UV absorbance of 5 µg ml -1 DS<br />

solution.<br />

66<br />

Figure. 4.2.3. UV spectra of DS at acidic pH, 3.56 (lower) and basic pH,<br />

11.68 (higher).<br />

Figure. 4.2.4. Calibration plot of absorbance verses concentration for DS<br />

69<br />

solutions from below to above as 0.1, 0.5, 1, 2, 3, 4, 5, 6, 7,<br />

8, 9, 10, 15, 20, 25 and 30 µg ml -1 .<br />

71<br />

Figure. 4.2.5. Representative UV-spectrum of expected 5 µg ml -1 DS in<br />

(Diclofen) tablet.<br />

73<br />

Figure. 4.2.6. UV spectra showing blank (black), DS containing (blue)<br />

and DS spiked (red) urine.<br />

74<br />

Figure. 4.2.7. UV spectra showing blank (black), DS containing (blue)<br />

and DS spiked (red) serum.<br />

Figure. 4.3.1. DC voltammograms (A) and DP voltammograms (B) of<br />

Paracetamol (c = 100 μmol<br />

75<br />

-1 .l) at CFE in Britton-Robinson<br />

buffer pH 2 to 12 (numbers above curves correspond to<br />

given pH) without electrode regeneration. Inset is<br />

corresponding dependence of peak potential on the pH.<br />

Figure. 4.3.2. Repetitive measurements of 100 µmol L<br />

77<br />

-1 Paracetamol<br />

using DPV at CFE in Britton-Robinson buffer pH 4.0 with<br />

regeneration potential Ereg1 = –400 mV and Ereg2 = 1300<br />

mV Inset is corresponding dependence of peak current on<br />

the number of scans.<br />

79


Figure. 4.3.3. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(PC): (1) 0, (2) 20 (3)<br />

40, (4) 60, (5) 80, (6) 100 µmol L -1 . Regeneration<br />

potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding<br />

calibration dependence.<br />

Figure. 4.3.4. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(PC): (1) 0, (2) 2 (3)<br />

80<br />

4, (4) 6, (5) 8, (6) 10 µmol L -1 . Regeneration potential<br />

Ereg1 = –400 mV and Ereg2 = 1300 mV. Background<br />

current = 1.94 nA. Inset is corresponding calibration<br />

dependence.<br />

Figure. 4.3.2. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(Paracetamol): 0 (1),<br />

81<br />

0.2 (2), 0,4 (3), 0,6 (4), 0,8 (5), 1 µmol L -1 (6).<br />

Regeneration potential Ereg1 = –400 mV and Ereg2 = 1300<br />

mV. Inset is corresponding calibration dependence.<br />

Figure. 4.3.6. Differential pulse voltammograms of Paracetamol at CFE<br />

in Britton-Robinson buffer pH 4.0, c(PC): (1) 0, (2) 0,02<br />

81<br />

(3) 0,04, (4) 0,06, (5) 0,08, (6) 0,1 µmol L -1 . Regeneration<br />

potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding<br />

calibration dependence.<br />

Figure. 4.3.3. Differential pulse voltammograms of Paracetamol 10 µmol<br />

82<br />

L -1 with Ascorbic acid from 10 to 100 µmol L -1<br />

(concentrations are written above curves in plot) at CFE in<br />

Britton-Robinson buffer pH 4.0, regeneration potential<br />

Ereg1 = –400 mV, and Ereg2 = 1300 mV. Inset is<br />

83<br />

corresponding dependence of peak current of<br />

Paracetamol on concentration of Ascorbic acid.<br />

Figure. 4.3.4. Differential pulse voltammograms of 0.1 ml urine with<br />

model sample of Paracetamol (2 – 10 µmol L -1 (A), and<br />

20 – 100 µmol L -1 (B), concentration is written next to<br />

curves in plots) sample at CFE in Britton-Robinson buffer<br />

pH 4.0, regeneration potentials Ereg1 = –400 mV, and Ereg2<br />

= 1300 mV. Inset is corresponding calibration dependence.<br />

84<br />

Figure. 4.4.1. Calibration plot in the range of 10-100 ppm for<br />

pharmaceutical samples<br />

87<br />

Figure. 4.4.2. <strong>Ph</strong>armaceutical Tablet sample and spikes of 30 ppm, 50<br />

ppm, 70 ppm ibuprofen<br />

88<br />

Figure. 4.4.3. Blank urine and 3 spikes of 10, 20, 30 ppm ibuprofen 90<br />

Figure. 4.4.5. Group Spectra of Ibuprofen Standards 92


Figure. 4.5.1. A and B. DC & DP voltammograms of Salicylic Acid<br />

(c = 100 µmol L -1 ) at CFE in Britton-Robinson buffer pH<br />

2 to 12 (numbers in above curves correspond to given pH).<br />

Inset is corresponding dependence of peak potential on the<br />

pH<br />

Figure. 4.5.2. Measurements of 100 µmol L -1 Salicylic Acid using DPV<br />

at CFE in Britton-Robinson Buffer pH 2.0 with activation<br />

potential (1) 2000,(2) 1500 and (3) 2200 mV<br />

Figure. 4.5.3. Differential pulse voltammograms of Salicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

20 (3) 40, (4) 60, (5) 8,0 (6) 100 µmol L -1 .<br />

Activation of potential=2200mV for 120 sec<br />

Inset is corresponding calibration dependence<br />

Figure. 4.5.4. Differential pulse voltammograms of Salicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2) 2<br />

(3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation of<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure. 4.5.5. Differential pulse voltammograms of Salicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

0.2 (3) 0.4, (4) 0.6, (5) 0.8, (6) 1 µmol L -1 . Activation of<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure.4.5.6. Differential pulse voltammograms of 10 µmol L -1<br />

hydrolyzed Acetylsalicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, (1) Electrolyte, (2) 10 µmol L -1<br />

without hydrolysis (3) 10 µmol L -1 Salicylic Acid (4) 10<br />

µmol L -1 after hydrolysis Acetylsalicylic acid. Activation<br />

potential=2200mV for 120 sec<br />

Figure. 4.5.7. DP voltammograms of hydrolyzed Acetylsalicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2.0, c(ASA): (1) 0, (2)<br />

20 (3) 40, (4) 60, (5) 80, (6) 100 µmol L -1 . Activation<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure. 4.5.8. DP voltammograms of hydrolyzed Acetylsalicylic Acid at<br />

CFE in Britton-Robinson buffer pH 2. c (ASA): (1) 0, (2) 2<br />

(3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation<br />

potential=2200mV for 120 sec. Inset is corresponding<br />

calibration dependence<br />

Figure. 4.5.9. DP voltammograms of hydrolyzed Acetylsalicylic Acid at CFE in<br />

Britton-Robinson buffer pH 2.0, c(ASA): (1) 0, (2) 0.2 (3) 0.4, (4)<br />

0.6, (5) 0.8, (6) 1 µmol L -1 . Activation potential=2200mV for 120<br />

sec. Inset is corresponding calibration dependence<br />

93<br />

95<br />

96<br />

96<br />

97<br />

98<br />

99<br />

99<br />

100


Figure.4.5.10.Differential pulse voltammograms of 4 µmol L -1 Duofilm<br />

sample with spikes of salicylic acid standard at CFE in<br />

Britton-Robinson buffer pH 2.0, c(SA): (1) electrolyte, (2)<br />

expected 4 μmol L -1 salicylic acid in sample of Duofilm<br />

(3) 6 μmol L -1 spike, (4) 8 μmol L -1 spike and (5) 10 μmol<br />

L -1 spike. Activation potential 2200 mV<br />

Figure. 4.5.11. Differential pulse voltammograms of 4 µmol L -1 Aspirin<br />

sample with spikes of hydrolyzed acetylsalicylic acid<br />

standard at CFE in Britton-Robinson buffer pH 2.0, c(SA):<br />

(1) electrolyte, (2) expected 4 μmol L -1 salicylic acid in<br />

sample of Aspirin (3) 6 μmol L -1 spike, (4) 8 μmol L -1<br />

spike and (5) 10 μmol L -1 spike. Activation potential 2200<br />

mV<br />

Figure. 4.5.12. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

20 (3) 40, (4) 60, (5) 80, (6) 100 µmol L -1 . Activation<br />

potential 2200 mV. Inset is corresponding calibration<br />

dependence<br />

Figure. 4.5.13. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2)<br />

2 (3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation potential<br />

2200 mV. Inset is corresponding calibration dependence<br />

Figure. 4.5.14. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(ASA): (1) 0,<br />

(2) 20 (3) 40, (4) 60, (5) 80, (6) 100 µmol L -1 . Activation<br />

potential 2200 mV. Inset is corresponding calibration<br />

dependence<br />

Figure. 4.5.15. Differential pulse voltammograms of 0.1 ml urine sample<br />

at CFE in Britton-Robinson buffer pH 2.0, c(ASA): (1) 0,<br />

(2) 2 (3) 4, (4) 6, (5) 8, (6) 10 µmol L -1 . Activation<br />

potential 2200 mV. Inset is corresponding calibration<br />

dependence<br />

Figure.4.5.16.Differential pulse voltammograms of hydrolyzed<br />

acetylsalicylic acid 5 μmolL -1 with ascorbic acid 5-100<br />

µmol L -1 at CFE in Britton-Robinson buffer pH 2.0.<br />

Activation potential 2200 mV<br />

101<br />

102<br />

103<br />

103<br />

105<br />

105<br />

106


List of Abbreviations<br />

AdSV Adsorptive Stripping Voltammetry<br />

AgA–CFE Silver Amalgam Carbon Film Electrode<br />

AgA–PE Silver Amalgam Paste Electrode<br />

AgE Silver Electrode<br />

AOAC Association of Official Analytical Chemists<br />

ASA Acetylsalicylic Acid<br />

ATR Attenuated Total Reflection<br />

AuE Gold Electrode<br />

CFE Carbon Film Electrode<br />

CNS Central Nerves System<br />

COX Cyclo-Oxygenase<br />

CPE Carbon Paste Electrode<br />

CV Cyclic Voltammetry<br />

DDP Differential Pulse Polarography<br />

DME Dropping Mercury Electrode<br />

DPV Differential Pulse Voltammetry<br />

DS Diclofenac Sodium<br />

E1/2 Half Wave Potential<br />

FIA Flow Injection Analysis<br />

FT-IR Fourier Transform Infrared Spectroscopy<br />

FT-NIR Fourier Transform Near-Infrared<br />

GC Gas Chromatography<br />

GC– MS Gas Chromatography Mass Spectroscopy<br />

GCE Glassy Carbon Electrode<br />

GGE Glassy Graphite Electrode<br />

HEC <strong>Higher</strong> <strong>Education</strong> <strong>Commission</strong><br />

HMDE Hanging Mercury Drop Electrode<br />

HPLC High Performance Liquid Chromatography<br />

HT-GLC High-Temperature Gas–Liquid Chromatography<br />

Hz Hertz<br />

IBP Ibuprofen<br />

LOD Limit of Detection<br />

LOQ Limit of Quantification<br />

LSV Linear Sweep Voltammetry<br />

mg/kg Milligram per Kilogram<br />

mg/kg /b.w/day Milligram per Kilogram per Body Weight per Day<br />

mg/L Milligram per Liter<br />

mm Millimeter<br />

mol.L -1 Mole per liter<br />

mV Milli volt<br />

mV s -1 Milli volt per second<br />

nA Nano Ampere<br />

NIR Near-Infrared Spectroscopy


nM Nano Molar<br />

NMR Nuclear Magnetic Resonance<br />

NPV Normal Pulse Voltammetry<br />

NSAIDs Non Steroidal Anti-Inflammatory Drugs<br />

PC Paracetamol<br />

PLS Partial Least Square<br />

ppb parts per billion<br />

ppm parts per million<br />

PtSE Platinum Solid Electrode<br />

PVC Polyvinyl Chloride<br />

RE Reference Electrode<br />

RMSEC Root Mean Standard Error of Calibration<br />

RMSEP Root Mean Square Error of Prediction<br />

RP-HPLC Reserved <strong>Ph</strong>ase-High-Performance Liquid Chromatography<br />

rpm revolutions per minute<br />

RSD Relative Standard Deviation<br />

SA Salicylic Acid<br />

SPME Solid <strong>Ph</strong>ase Microextraction<br />

SWV Square Wave Voltammetry<br />

TLC Thin Layer Chromatography<br />

TOF-MS Time-of-Flight Mass Spectrometry<br />

TQ Turbo Quant<br />

V/s Volt per second<br />

v/v Volume by volume<br />

WE Working Electrode<br />

WHO World Health Organization<br />

μg/kg Microgram per kilogram<br />

µg L -1 Microgram per liter<br />

µg/g Microgram per gram<br />

µL Micro liter<br />

µm Micrometer<br />

µM Micro mole<br />

µmol L -1 Micro mole per liter


INTRODUCTION<br />

1.1. Analgesics / NSAIDs -An overview<br />

Chapter-01<br />

A drug designed to control pain. Analgesic comes from the Greek word an means without<br />

and algesis means sense of pain collectively called without sense of pain. They are the<br />

commonly prescribed and widely consumed (over-the-counter) formulation in the world.<br />

Although analgesics are generally supposed to be safe agents, harmful effect may occur<br />

in the case of chronic abuse and acute overdose (Suzanne & Steven, 1998). Analgesics<br />

are used in the treatment of a broad range of therapeutic situations, they are used in<br />

treatment of both chronic and acute pain symptom such as dysmenorrheal or cephalgia.<br />

Antipyresis is also common sign for the administration of these drugs, mainly in the<br />

children.<br />

Non steroidal anti-inflammatory drugs (NSAIDs) are the chemically heterogeneous group<br />

of compounds, often chemically unrelated (although most of them are organic acids)<br />

which nevertheless share certain therapeutic actions and adverse effects Goodman &<br />

gilmans (2007). It is given as single dose or in short-term alternating therapy; it can<br />

reduce mild to moderate ache. The anti-inflammatory effects become apparent when it<br />

may give up to 21 day. The combined anti-inflammatory and analgesic effects make them<br />

helpful for the symptomatic relief of painful situation including rheumatic disorders such<br />

as rheumatoid arthritis, osteoarthritis, and the spondyloarthropathies, and also in peri-<br />

articular disorders, and soft-tissue rheumatism. Some NSAIDs are used in the<br />

management of postoperative pain (Bidaut-Russell & Gabriel, 2001).<br />

1


Anti-inflammatory activity of drug shows the small differences, between the NSAIDs and<br />

preference is mostly empirical. If one NSAID fail to respond the patient, in that case<br />

another drug may be selected for treatment. However, it has been recommended that<br />

NSAIDs associated with a low risk of gastrointestinal toxicity should generally be<br />

preferred and the lowest effective dose used. NSAIDs are usually given by mouth, with<br />

or after food, although some such as diclofenac, ketorolac, lornoxicam, parecoxib, and<br />

tenoxicam can be given intravenously and intramuscularly. Some NSAIDs are applied<br />

topically or given rectally as suppositories. Several NSAIDs are used in ophthalmic<br />

preparations for the inhibition of intra-operative miosis, control of postoperative ocular<br />

inflammation.<br />

1.1.1. Mechanism of Action (NSAIDs)<br />

Cyclo-oxygenases play an important role in the biosynthesis of prostaglandins. NSAIDs<br />

inhibit cyclo-oxygenase-1 (COX-1) and cyclo-oxygenase-2 (COX-2) and it is thought<br />

that inhibition of COX-1 is connected with unfavorable gastrointestinal effects while<br />

inhibition of COX-2 is related with anti-inflammatory activity (Hayllar, et al., 1995,<br />

Richardson and Emery, 1996) hence the interest in preferential or selective inhibitors of<br />

COX-2. COX-2 inhibitors may also have a potential use in other diseases in which COX-<br />

2 might be implicated (Jouzeau, et al., 1997, Hawkey, 1999). There is evidence that<br />

NSAIDs may also have a central mechanism of action that augments the peripheral<br />

mechanism (Gupta & Tarkkila 1998; Cashman, 1996).<br />

In view of the analgesic (Paracetamol, Aspirin, Diclofenac and Brufen) studied during the<br />

current project it is essential to focus on the properties, uses and toxic effects of these<br />

drugs.<br />

2


1.2. Paracetamol<br />

1.2.1. <strong>Ph</strong>armacopoeias:<br />

Fig. 1.1. Structural formula of Paracetamol (PC)<br />

Chemical name: 4´-Hydroxyacetanilide; N-(4-Hydroxyphenyl)acetamide<br />

Molecular formula: C8H9NO2 =151.2<br />

According to British, European and United States <strong>Ph</strong>armacopoeias Paracetamol<br />

(Acetaminophen) is a white crystalline odourless powder. It is sparingly soluble in water,<br />

freely in alcohol and sodium hydroxide and very slightly soluble in dichloromethane<br />

(Jacoby, 2000). It should be protected from light and stored in airtight containers.<br />

Paracetamol (N-acetyl-p-aminophenol) is an effective alternative to aspirin as an<br />

analgesic, antipyretic agent and safe up to therapeutic doses. It work as painkiller by<br />

control prostaglandin’s synthesis in the central nervous system and reduced fever by<br />

painkilling hypothalamic heat-regulating center (Goyal and Singh 2006; de los, et al.,<br />

2005; Moreira, et al., 2005; Campanero, et al., 1999). Paracetamol is readily absorbed<br />

after administration and widely distributed throughout most body fluids (Criado, et al.,<br />

2000), as a weak acid (pKa value 9.5), it gets quickly absorbed and distributed after oral<br />

administration and is immediately emitted through urine (Goyal and Singh 2006;<br />

Parojčić, et al., 2003). Generally paracetamol does not shows any unsafe side effects but<br />

hypersensitivity or overdoses in few cases lead to the formation of some liver and<br />

nephrotoxic metabolites (Goyal and Singh 2006; Patel, 1992).<br />

3


1.2.2. Uses and Administration<br />

Paracetamol is often self-prescribed and given by mouth or as a rectal suppository for<br />

relief of moderate pain, fever, lumber pain, backache, migraine or non-specific<br />

indications without any medical control (Mcgregir, et al., 2003; Nikles, et al., 2005). It<br />

has been reported as a useful drug in osteoarthritis therapy and in recent years,<br />

postoperative pain as well (Brandt, 2003). After ingestion of an overdose amount of<br />

paracetamol, the accumulation of harmful metabolite may cause severe and occasionally<br />

fatal nephrotoxicity and hepatotoxicity (Xu and Li, 2004; Maria, et al., 2005). However,<br />

the frequent use of paracetamol in late pregnancy may be associated with an increased<br />

risk of persistent wheezing in the infant (Farquhar; 2009; Shaheen, et al., 2002).<br />

Usual doses in children are: under one to five years, 120 to 250 mg and six to twelve<br />

years, 250-500 mg. The normally adult dose by 30 days is 0.5-1 g every four to six hrs up<br />

to a highest of 4 g each day. These doses may be given every four to six hrs when<br />

necessary up to a maximum of four doses in one day (Prescott, 1996).<br />

1.2.3. Adverse Effects<br />

Side-effects of paracetamol are exceptional and typically soft, although haematological<br />

reactions including leucopenia, thrombocytopenia, neutropenia, agranulocytosis and<br />

pancytopenia have been described. Skin rashes and other hypersensitivity reactions occur<br />

rarely (Xu and Li, 2004; Martin & McLean, 1998; KocaoÄŸlu, et al., 1997; Mugford &<br />

Tarloff, 1997; Nagasawa, et al., 1996; Ishida, et al., 1997).<br />

4


1.2.4. Overdosage and its Treatment<br />

Ingestion of as little as ten to fifteen g of paracetamol by adults may cause severe<br />

hepatocellular necrosis and less often renal tubular necrosis. However, chronic use of<br />

supratherapeutic doses in children has resulted in unintentional overdoses and severe<br />

hepatotoxicity (Miles, et al., 1999; American Academy of Pediatrics Committee on<br />

Drugs, 2001; Wyszecka-Kaszuba, et al., 2003).<br />

Activated charcoal may be used to reduce gastrointestinal absorption, if it can be given<br />

within 1 hour of the overdose, and if greater than 150 mg/kg of paracetamol has been<br />

consumed. However, if acetylcysteine or methionine is to be given by mouth the charcoal<br />

is best cleared from the stomach to prevent its reducing the absorption of the antidote.<br />

There is little evidence that gastric lavage is of benefit in those who have overdosed<br />

solely with paracetamol.<br />

1.2.5. Precautions<br />

Paracetamol should be prescribed with carefully to patients with harmed liver or kidney<br />

and alcohol dependence patients. About 0.01% population of the United state and 0.02%<br />

population of the Australian were appraised in hospital every year because of PC<br />

poisoning (de los et al., 2005; Dargan & Jones, 2003).<br />

5


1.3. Aspirin<br />

1.3.1. <strong>Ph</strong>armacopoeias<br />

Fig. 1.2. Structural formula of Aspirin (ASA)<br />

Chemical name: O-Acetylsalicylic acid; 2-Acetoxybenzoic acid<br />

Molecular formula: C9H8O4 =180.2<br />

According to British, European and United States <strong>Ph</strong>armacopoeias Acetylsalicylic Acid<br />

or Aspirin is white crystalline powder, commonly tubular or needle-like, stable in dry, air<br />

has a faint odour or odourless, while in moist air it steadily hydrolyzes to salicylic and<br />

acetic acids. It is slightly soluble in water but freely in alcohol, chloroform and sparingly<br />

soluble in absolute ether (Jacoby, 2000), store in airtight containers.<br />

Acetylsalicylic acid (ASA), shown in Figure 2, more popularly known as aspirin, is one<br />

of the oldest medicines that still plays an important role in modern therapeutics. It is<br />

widely employed in pharmaceutical formulations for the relief of headaches, fever,<br />

muscular pain, and inflammation (Xianwen, et al., 2009). Acetylsalicylic acid or aspirin,<br />

was introduced in the late 1890s (Dreser, 1899). It was first synthesized in 1897, by Felix<br />

Hoffmann, in the Farbenfabrik Freidrich Bayer laboratories, in Elberfeld, Germany<br />

(Hammerschmidt, 1998; Sneader, 2000; Moore, et al., 1995; Kibbey, et al., 1992).<br />

Salicylates, in the form of willow bark, were used as an analgesic during the time of<br />

Hippocrates (Pirker, et al., 2004). This substance is consumed worldwide (Erica et al.,<br />

2010; Elwood, 2001) indicates the importance of the development of new analytical<br />

methods to assess not only the quality but also the authenticity of the product (Erica et<br />

al., 2010).<br />

6


The analgesic and anti-pyretic efficiency of aspirin was promptly comproved, however<br />

only after the years 1940 it began to be employed in higher doses as anti-inflammatory<br />

agent. When its mechanism of action began to be understood the possibility for use<br />

against cardiac and circulatory disturbances became evident (Muralidharan et al., 2008;<br />

Eccles, et al., 1998).<br />

The rate of decomposition of ASA to salicylic acid (SA) and acetic acid (AA) is reliant<br />

on solution pH and temperature (Lu & Tsai, 2010). In the pH range 11-12 ASA is quickly<br />

hydrolyzed, in the pH range 4-8 its hydrolysis rate is slow while highest constancy is<br />

attained at pH 2-3 (Connors, et al., 1979).<br />

1.3.2. Uses and Administration<br />

It is used in acute conditions such as arthralgia, headaches, myalgia and other cases<br />

involving mild analgesia. Once ingested, ASA is quickly hydrolysed in the body to<br />

formulate salicylic acid (SA), the compound that is primarily responsible for the<br />

pharmacological activity of ASA (Houshmand, et al., 2008). It is also used for reduce the<br />

excitements of viral or bacterial origin (Houshmand, et al., 2008; Boopathi, et al., 2004,<br />

De Carvalho, et al., 2004). In general, the salicylates action is achieved by the SA<br />

contents, although some of the characteristic properties of the ASA are their capability<br />

for protein acetylation. The esters in the phenolic or carboxylic functional groups alter the<br />

power in the toxicity of salicylates (Torriero, et al., 2004; Hardman, et al., 1996).<br />

Aspirin act as inhibitors of the enzyme cyclo-oxygenase, which outcome in the direct<br />

inhibition of the bio-synthesis of thromboxanes and prostaglandins from arachidonic acid.<br />

7


It has also been used in the management of inflammation and pain in acute and chronic<br />

rheumatic disorders such as juvenile idiopathic arthritis, rheumatoid arthritis, ankylosing<br />

spondylitis and osteoarthritis. In the treatment of minor febrile conditions, such as colds<br />

or influenza, aspirin can reduce temperature and relieve headache and joint and muscular<br />

pains (Vane, et al., 2006, Thun, et al., 2002).<br />

Aspirin is usually taken by mouth. Various dosage forms are available including plain<br />

uncoated tablets, buffered tablets, dispersible tablets, enteric-coated tablets, and<br />

modified-release tablets. In some instances aspirin may be given rectally by suppository.<br />

The usual oral dose of aspirin as an analgesic and antipyretic is 300-900 mg, repeated<br />

every 4 to 6 hours according to clinical needs, to a maximum of 4 g daily. The dose as<br />

suppositories is 600-900 mg every four hrs to a maximum of 3.6 g every day.<br />

1.3.3. Adverse Effects<br />

The most common adverse effects of therapeutic doses of aspirin are gastrointestinal<br />

disturbances such as nausea, dyspepsia, and vomiting (Dirckx, et al., 2009). This<br />

substance is highly consumed in all over the world, and its remedial act and its toxic<br />

effects make it a drug that is subjected to continuous researches (Majdi, et al., 2007;<br />

Elwood, 2001), and its antipyretic effects have been recognized for more than 200 years<br />

(Stone, 1763). However, the antiplatelet activity of this agent was not recognized until<br />

almost 70 years later (Awtry & Loscalzo, 2000; Wong, et al., 2004). Researchers have<br />

also demonstrated therapeutic benefit of aspirin in a variety of cardiovascular diseases<br />

with its doses of 30 to 1500 mg/d (Awtry & Loscalzo, 2000, Wong, et al., 2004, Patrono,<br />

1994, Fuster et al., 1993, Hennekens, et al., 1997, Manson, et al., 1991).<br />

8


The effect of salicylates on cancer treatment has been studied, too (Thun et al., 2002,<br />

Shen, et al., 2004, Keloff, et al., 2004, Hardwick, et al., 2004, Martnett, 1992). Moreover,<br />

inhibition of growth of bacteria Helicobacter pylori (Wang, et al., 2003) and<br />

Staphyloccocus aureus (Kupferwasser, et al., 2003) has been also described. Kidney<br />

damage related with the therapeutic utilization of aspirin alone appears to be relatively<br />

rare (Dubach, et al., 1991, Perneger, et al., 1994). Aspirin-induced liver injury is<br />

frequently reversible on stopping the drug (Lewis, 1984, Freeland, et al., 1988). Aspirin<br />

burns when ulceration of the mucosal layer of the lips) developed in a 26-year old woman<br />

after getting an aspirin-containing powder for a migraine (Dellinger and Livingston,<br />

1998).<br />

1.3.4. Overdosage and its Treatment<br />

In case of acute oral salicylate overdosage, repeated doses of activated charcoal may be<br />

given by mouth if the patient is suspected of ingesting more than 250 mg/kg of salicylate.<br />

Intravenous sodium bicarbonate is given to enhance urinary salicylate excretion if plasma<br />

salicylate concentration exceeds 500 micrograms/mL (350 micrograms/mL in children).<br />

Haemodialysis or haemoperfusion are also effective methods of removing salicylate from<br />

the plasma.<br />

1.3.5. Precautions<br />

Aspirin should not be given to patients with haemophilia or other haemorrhagic disorders<br />

or the patients with a history of sensitivity reactions to aspirin, including those in whom<br />

attacks of asthma, angioedema, urticaria, or rhinitis have been precipitated by such drugs<br />

although low-dose aspirin might be given in some pregnant patients as an analgesic.<br />

9


1.4. Diclofen<br />

1.4.1. <strong>Ph</strong>armacopoeias<br />

Fig. 1.3. Structural formula of Diclofenac Sodium (DS)<br />

Chemical name: Sodium 2-[(2, 6-dichlorophenyl) amino] phenyl acetate<br />

Molecular formula: C14H10Cl2NNaO2 =318.1<br />

According to British, European and United States <strong>Ph</strong>armacopoeias diclofenac Sodium is<br />

a white to slightly yellowish (off-white), slightly hygroscopic, crystalline powder. It is<br />

sparingly soluble in water, alcohol, slightly in acetone and freely in methyl alcohol,<br />

practically insoluble in chloroform and in ether (Bahram, 2008). pH of a 1% solution in<br />

water is between 7.0 and 8.5. Store in airtight containers and protect from light (Rients &<br />

Jan, 2008).<br />

1.4.2 Uses and Administration<br />

A phenylacetic acid derivative, Diclofenac is an NSAID (Matin, et al., 2005). It is used<br />

mostly as the sodium salt for the release of inflammation and pain in many situations:<br />

joint disorders and musculoskeletal such as osteoarthritis, rheumatoid arthritis, and<br />

ankylosing spondylitis; peri-articular disarray such as tendinitis and bursitis; soft-tissue<br />

disarray such as strains and sprains; and other painful circumstances such as renal colic,<br />

dysmenorrhoea, migraine and acute gout subsequent surgical processes (Lala, et al.,<br />

2002). Eye drops of diclofenac sodium are used for the prevention of intra-operative<br />

10


miosis for the duration of cataract removal, for the medication of inflammation following<br />

surgery or laser treatment of the eye, for pain in corneal epithelial defects following<br />

surgery or accidental trauma, and for the relief of ocular signs and symptoms of seasonal<br />

allergic conjunctivitis.<br />

The usual dose of diclofenac sodium by mouth or rectally is 75 to 150 mg daily in<br />

divided doses. In children of 1 to 12 years old, the dose by mouth for juvenile idiopathic<br />

arthritis is one to three mg/kg daily in divided doses.<br />

1.4.3. Adverse Effects<br />

There may be pain and, occasionally, tissue damage at the site of injection when<br />

diclofenac is given intramuscularly. Diclofenac suppositories may cause local irritation.<br />

Transient burning and stinging may occur with diclofenac ophthalmic solution; more<br />

serious corneal adverse effects have also occurred. The main effects on the blood reports<br />

are haematological abnormalities including haemolytic anaemia (López, et al., 1995)<br />

agranulocytosis (Colomina & Garcia, 1989) neutropenia (Kim & Kovacs, 1995) and<br />

thrombocytopenia (George & Rahi, 1995) happening in patients given diclofenac.<br />

Localized slow bleeding and inhibition of platelet aggregation (Price & Obeid, 1989)<br />

bruising and prolonged bleeding time (Khazan, et al., 1990) have also been reported.<br />

Rectal management of diclofenac suppositories may origin local reactions such as<br />

burning and itching. Nephrotic syndrome (Beun, et al., 1987; Yinnon, et al., 1987;<br />

Tattersall, et al., 1992) described in patients taking diclofenac. Rise of clinical hepatitis<br />

and serum amino transferase activity (Ryley, et al., 1989) including fatal fulminant<br />

hepatitis (Purcell, et al., 1991) have occurred in patients getting diclofenac.<br />

11


1.4.4 Precautions<br />

Use of intravenous diclofenac is contra-indicated in patients with moderate or severe<br />

renal impairment, hypovolaemia, or dehydration. Intravenous diclofenac should not be<br />

used in patients with a history of haemorrhagic diathesis, cerebrovascular bleeding<br />

(including suspected), or asthma nor in patients undergoing surgery with a high risk of<br />

haemorrhage.<br />

1.5. Brufen<br />

1.5.1. <strong>Ph</strong>armacopoeias<br />

Fig. 1.4. Structural formula of Ibuprofen (IBP)<br />

Chemical name: [2-{4-(2-methylpropyl)phenyl} propanoic acid]<br />

Molecular formula: C13H18O2 =206.3<br />

According to British, European and United States <strong>Ph</strong>armacopoeias ibuprofen is a<br />

colourless crystals or white crystalline powder having a minute characteristic odor. It is<br />

practically insoluble in water, freely soluble in acetone, in dichloromethane, chloroform<br />

and in methyl alcohol. It dissolves in dilute solutions of alkali hydroxides and carbonates.<br />

Store in airtight containers.<br />

1.5.2. Uses and Administration<br />

Ibuprofen, a propionic acid derivative, is an important anti-inflammatory, analgesic and<br />

antipyretic medicine with considerably less gastrointestinal adverse effect than other<br />

NSAIDs (Velasco, et al., 2010; Donald, et al., 2010; Whittle, et al., 2003) employed in<br />

12


the administration of mild to moderate pain in situation such as headache,<br />

dysmenorrhoea, postoperative pain, including migraine, musculoskeletal and dental pain;<br />

joint disorders such as osteoarthritis, rheumatoid arthritis and ankylosing spondylitis;<br />

peri-articular disorders such as tenosynovitis and bursitis; soft-tissue disorders such as<br />

strains and sprains. NSAIDs is also used to lower fever (Clara, et al., 2009; Santini, et al.,<br />

2006).<br />

Patients with rheumatoid arthritis generally require higher doses of ibuprofen than those<br />

with osteoarthritis. The recommended dose for fever reduction in adults is 200 to 400 mg<br />

every 4 to 6 hours to a highest of 1.2 g every day (Nanda et al., 2010). In children the<br />

usual dose by mouth for the treatment of pain or fever is 20 to 30 mg/kg daily in divided<br />

doses. Ibuprofen is also applied topically as a 5% cream, foam, gel, or spray solution; a<br />

10% gel is also available.<br />

1.5.3. Adverse Effects<br />

Symptoms of vomiting, tinnitus and nausea have been detailed after ibuprofen an<br />

excessive dose. Moreover severe toxicity is unusual, but gastric emptying chased by<br />

supportive measures is suggested if the amount ingested within the previous hour exceeds<br />

four hundred mg/kg.<br />

Effect on the blood including agranulocytosis, aplastic anaemia (Gryfe & Rubenzahl,<br />

1976) pure white-cell aplasia (Mamus, et al., 1986) and thrombocytopenia ( Jain, 1994 )<br />

have been reported in patients taking ibuprofen. Fatal haemolytic anaemia occurred in a<br />

man taking ibuprofen and oxazepam ( Guidry et al., 1979 ). A review of NSAID-related<br />

CNS adverse effects summarized twenty three literature reports of NSAID-associated<br />

13


aseptic meningitis (Hoppmann, et al., 1991), while seventeen reports involved ibuprofen<br />

patients with a diagnosis of systemic lupus erythematosus. Typically the reaction is seen<br />

in patients who have just restarted NSAID therapy after a gap in their treatment. The<br />

authors believed this to be the first reported case of ibuprofen-induced aseptic meningitis<br />

in a patient with rheumatoid arthritis (Horn & Jarrett, 1997). Hyponatraemia has been<br />

described in patients receiving ibuprofen (García, et al., 2003). Ibuprofen may be<br />

associated with a lower risk of upper gastrointestinal effects than some other NSAIDs,<br />

but nonetheless it can cause vomiting, nausea, dyspepsia, gastrointestinal bleeding,<br />

perforation and peptic ulcers. Colitis and its exacerbation have also occurred (Ravi, et al.,<br />

1986, Clements, et al., 1990). Adverse renal effects with ibuprofen include an increase in<br />

serum creatinine concentration (Whelton, et al., 1990), Acute renal failure (Fernando, et<br />

al., 1994) and nephrotic syndrome (Justiniani, 1986) cystitis, haematuria, and interstitial<br />

nephritis may occur as well.<br />

1.5.4. Overdosage and Treatment<br />

Usually after ingestion of large dose of ibuprofen serious toxicity may occur, including<br />

seizures, hypotension, apnoea, coma, and renal failure. Treatment of Ibuprofen<br />

overdosage is entirely supportive. Gastric lavage and activated charcoal may be of benefit<br />

within 1 hour of ingestion. Multiple doses of activated charcoal may be useful in<br />

enhancing elimination of Ibuprofen with long half-lives.<br />

14


LITERATURE REVIEW<br />

2.1. Analytical Techniques for Assessment of NSAIDs<br />

Chapter-02<br />

Various methods are has been developed for NSAIDs determination in different types of<br />

samples (Sirajuddin, 2007). These include, titrimetry , thin layer chromatography (TLC),<br />

colorimetry, Gas Chromatography/ Mass Spectrometry (GC/MS), high performance<br />

liquid chromatography (HPLC), Voltammetry, Flow injection analysis (FIA),<br />

Spectrofluorimetry, UV/visible Spectrophotometry, Chemiluminescence, NMR, FTIR,<br />

Raman and Solid-state Linear Dichroic Infrared (IR-LD) Spectroscopy.<br />

Ayora et al., 2000, reported a direct sensor based determination for paracetamol analysis<br />

using easy flow-through ultraviolet-visible optosensing device based on the solid support<br />

packed in the flow cell after that continuously observing their absorbance on the solid<br />

phase at 264 nm. The proposed method was profitably applied to the PC in<br />

pharmaceuticals formulations.<br />

Fujiwara et al., 2002, developed a systematic approach for the aqueous crystallization of<br />

paracetamol. The solubility curve and the solution concentration of paracetamol in water<br />

were determined using Fourier transform infrared (FTIR) spectroscopy coupled with<br />

attenuated total reflection (ATR) mode and chemometrics.<br />

Sun et al., 2003, reported a sensitive method on HPLC coupled with UV detector for the<br />

simultaneously find out of non-steroidal anti-inflammatory drugs (NSAIDs) possessing<br />

an arylpropionic acid moiety in plasma and pharmaceutical formulations.<br />

15


Šatínský et al., 2004, been described the molecularly imprinted polymer as a carbon fiber<br />

microelectrode coating for determining paracetamol using cyclic voltammetry (CV).<br />

A voltammetric method for the determination of ascorbic acid with Carbon Paste<br />

Electrodes (CPEs), was applied with good results to several dosage forms (tablets, vials)<br />

and even to an effervescent dosage form containing a mixture of ascorbic and<br />

acetylsalicylic acid (Sandulescu et al., 2000).<br />

2.2. Electrochemical Techniques<br />

Use of electrochemical methods, especially the voltammetric and amperometric in<br />

analysis paid attention as sensitive, cost-effective and accurate way of analysis in last<br />

decades.<br />

Voltammetric methods have been developed due to high sensitivity these techniques<br />

offer, principally when a variety of redox mediators (Rover et al., 2000; Lobo et al.,<br />

1997, Fung & Luk, 1989) are employed, decreasing the interference levels in the<br />

determinations of compounds present in complex matrices such as pharmaceuticals and<br />

body fluids (Rover et al., 2000). Square wave voltammetry (SWV) is regarded as very<br />

responsive and direct analytical techniques, which has used for the fast and sensitive<br />

determination of a wide range of organic molecules, with low nonfaradaic current and<br />

high sensitivity. In most instances this method presents further advantages such as no<br />

need for sample pretreatment and less sensitivity to matrix effects than other analytical<br />

techniques. (Codognoto et al., 2002; Souza et al., 2003 ; Pedrosa et al., 2003;<br />

Osteryoung., 1985).<br />

16


Nowadays a number of polymer films are used in the electroanalytical field due to easy<br />

generated on the electrode surface than monolayers. To improve the selectivity and<br />

sensitivity for the determination of PC various polymeric based films modified electrodes<br />

has been developed (Chenghang Wang, et al., 2006; Sandik & Wallace, 1993; Piro et al.,<br />

2000).<br />

The electroanalytical features and performances of CPEs for fabrication of chemo and<br />

biosensors through the modification of carbon paste and their analytical applications are<br />

well documented (Goyal, & Singh, 2006; Vire et al., 1994; Kalcher, 1990; Kalcher et al.,<br />

1995; Gorton, 1995; Campanella et al., 1992; Kalcher et al., 1997).<br />

It is well known from the molecular structure of paracetamol, that it is electrochemically<br />

active, and its electrochemical properties on a choice of electrodes, such as glassy carbon<br />

electrodes (GCE) (Wang et al., 2006; Bolado et al., 2009; Santos et al., 2008;<br />

Wangfuengkanagul et al., 2002; Gimenes et al., 2010), screen print electrode (Fanjul-<br />

Bolado et al., 2009), gold electrodes (Santos et al., 2008, Pedrosa et al., 2006), and<br />

diamond electrode have been investigated (Wangfuengkanagul et al., 2002).<br />

The cyclic voltammetric studies concerning the electrochemical oxidation of<br />

acetaminophen were also explained (Sandulescu, 2000; Miner et al., 1981; Benschoten et<br />

al., 1983).<br />

Zen et al., 1997, used the Nafion/ruthenium oxide pyrochlore chemically modified<br />

electrode using SWV for the simultaneous determination of acetaminophen and caffeine<br />

in drug formulations. The detection limits were 2.2 and 1.2 μM for caffeine and<br />

17


acetaminophen, respectively. The analytical application of the proposed method was<br />

applied in several commercially available drugs, without any preliminary treatment. Fang<br />

Fang et al., 1997, proposed an oxidation process of acetaminophen at a platinum<br />

electrode by parallel incident spectroelectrochemistry. The results exposed that the<br />

acetaminophen redox reaction was a diffusion-controlled one-electron quasi reversible<br />

process, this process confirm by cyclic voltammetric experiments. Capsules were<br />

analyzed without pretreatment using linear range of acetaminophen with a R 2 =0.9985.<br />

The voltammetric and spectrophotometric methods which were used to determine<br />

ascorbic acid and acetaminophen in different quantity forms (tablets, effervescent and<br />

vials). The electroanalytical study of acetaminophen, ascorbic and some mixtures of these<br />

compounds in different ratios has been made by using a (CPE-graphite:solid paraffin 2:1)<br />

as working electrode (Sandulescu et al., 2000). Wangfuengkanagul et al., 2002, reported<br />

the electrochemical behavior of acetaminophen in phosphate (pH 8) buffer solution at a<br />

boron-doped diamond thin film electrode using hydrodynamic voltammetry. Torriero, et<br />

al., 2004, reported a cyclic and DPV method for the electrochemical oxidation of SA on a<br />

glassy carbon electrode (GCE). The method was linear over the salicylic acid<br />

concentration at the range of 1–60 µgml -1 .<br />

Goyal, et al., 2005, a reproducible method for determined the paracetamol using a<br />

nanogold modified indium tin oxide electrode were developed. Under conditions of<br />

differential pulse voltammetry, 2.0x10 -7 –1.5x10 -3 M was range of linear calibration curve<br />

with a correlation coefficient of 0.997 was obtained.<br />

18


An additional approach by Goyal, et al., 2006, for voltammetric analysis of PC was<br />

carried out at C60-modified glassy carbon electrode, which demonstrates steady response<br />

with better sensitivity and selectivity. A linear calibration curve was obtained in the range<br />

of 0.05–1.5 mM paracetamol concentration with sensitivity of the method as 13.04<br />

µAmM −1 having correlation coefficient of 0.985.<br />

Chenghang et al., 2006, fabricated a novel L-cysteine film modified electrode effectively<br />

applied for the analysis of acetaminophen by means of an electrochemical oxidation<br />

procedure in tablets and human urine.<br />

Jia et al., 2007, reported the electrochemical determination of acetaminophen on the<br />

AMP SAMs/Au in Britton–Robinson (BR) solution of buffer by SWV. The modified<br />

electrode showed a significant enhancement in the oxidation current response for<br />

acetaminophen in comparison to a bare gold electrode. Linear calibration curve were<br />

obtained in the range of 2.0 ×10 –6 – 4.0 × 10 –3 M.<br />

Kachoosangi et al., 2008 reported a sensitive and selective electroanalytical method for<br />

the analysis of paracetamol utilizizng adsorptive stripping voltammetric technique at a<br />

multi walled carbon nanotube modified basal plane pyrolytic graphite electrode<br />

(Kachoosangi, et al., 2008).<br />

Saraswathyamma et al., 2008, reported gold electrode modified with dipyrromethene-Cu<br />

(II) derivatives possessing two dodecane alkyl chains. The presence of dipyrromethene-<br />

Cu(II) redox centers on the electrode surface was proved by CV and Osteryoung square-<br />

wave voltammetry for determination of PC in plasma. Skeika et al., 2008, investigated<br />

19


the simultaneous electrochemical determination of paracetamol and dypirone by<br />

differential pulse voltammetry technique (DPV) using an unmodified carbon paste<br />

electrode. The multivariate calibration methodology based on partial least square<br />

regression (PLSR) was employed for the voltammetric peaks of paracetamol and<br />

dypirone, due to overlapping.<br />

Parviz et al., 2009 proposed the highly stable, convenient method for the electrochemical<br />

oxidation of naproxen and PC using dysprosium nanowire modified carbon paste<br />

electrode With SWV. A low detection limit for PC and naproxen, were observed by this<br />

method.<br />

Elen et al., 2009, reported a direct determination of acetylsalicylic acid (ASA) in<br />

pharmaceutical formulations using SWV and a boron-doped diamond electrode (BDD).<br />

The obtained relative standard deviation was smaller than 1.4% with a detection limit of<br />

2.0 µmol L -1 . Nada & Mehar, 2009 reported a promising voltammetric sensors based on<br />

the modification of platinum (Pt) and poly (3 methylthiophene) (PMT) electrodes with<br />

palladinum (Pd) nanoparticles were achieved for the determination of paracetamol.<br />

Shahrokhian & Elham, 2010, have simultaneous determined acetaminophen and ascorbic<br />

acid in the presence of isoniazid by CV and DPV using CPE modified with thionine<br />

immobilized on multi-walled carbon nanotube (MWCNT).<br />

Irena Baranowska & Marta, 2009, proposed a sensitive and fast method for the<br />

quantification of PC and its glucuronide (PG) and sulfate (PS) metabolites. The<br />

electrochemical properties of the compounds were examined by CV on GCE.<br />

20


Bruna et al., 2009, developed simple and highly selective SWV or DP voltammetric<br />

method for the single or simultaneous determination of caffeine and paracetamol in<br />

aqueous media (acetate buffer, pH 4.5) on a BDDE. The limits of detection for the<br />

simultaneous determination of caffeine and paracetamol were 3.5×10 −8 mol L −1 and<br />

4.9×10 −7 mol L −1 , respectively.<br />

It was also planned to develop a selective methods for PC analysis in pharmaceutical<br />

determinations, and the methods were based on reaction between ethylacetoacetate and<br />

PC by sulfuric acid (dehydrating agent) producing a coumarinic compound, which was<br />

spectrofluorimetrically calculated (Schirmer et al., 1991; Walily et al., 1999; Gikas et al.,<br />

2003).<br />

Xu et al., 2009, FI analysis for the determination of acetaminophen investigated by an<br />

amperometric detector with gold nanoparticle modified carbon paste electrode. The<br />

obtained results were found to be comparable with HPLC.<br />

A vast deal of interest has been point out on the Substances immobilized onto the<br />

electrode surface and chemically modified electrodes under the effect of external electric<br />

fields able to mediating fast electron transfer (Majdi, 2007; Pournaghi-Azar & Sabzi,<br />

2004, Golabi & Irannejad, 2005).<br />

2.3. Spectroscopic Methods<br />

Spectrofluorimetry for the UV–visible region can be employed to achieve the<br />

measurements in solid matrix (Moreira et al., 2004), leading to favorable characteristics<br />

of sensitivity, straightforwardness, selectivity, rapidity and ruggedness etc.<br />

21


Nondestructive analyzes were carried out that follow the current tendency towards clean<br />

chemistry. Moreover, the opportunity of optical-fiber accessories for in situ and/or on-<br />

line analysis (Bosch, 2006; Utzinger and Richards-Kortum, 2003) becomes possible.<br />

Now a day, this approach has scarcely been exploited in relation to solid samples of<br />

pharmaceutical importance. Regarding the paracetamol analysis, a literature review tells<br />

that it generally involves the derivative reactions (Pulgarin & Bermejo, 1996; Murillo &<br />

Garcia, 1996) because paracetamol was not basically fluorescent in aqueous solutions. In<br />

this view beginning experiments confirmed that paracetamol was fluorescent in the solid<br />

phase (Bosch, 2006).<br />

Various spectrofluorimetric methods reported for the quantification of single PC or as a<br />

mixture with other drugs in pharmaceutical preparations, such as indirect determination<br />

using Ce (IV) as an oxidant agent (de los, 2005; Amann et al., 1980), reaction with<br />

fluorescamine (Nakamura & Tamura., 1980), 1-nitroso-2-naphthol (Shah & Balaraman,<br />

1999), oxidation with 2,2-dihydroxy- 5,5-diacetyldiaminebiphenyl (Vilchez et al., 1995),<br />

potassium hexacyanoferate (III) (Pulgarín & Bermejo, 1996). Conversely, several of<br />

these methods show low selectivity and interference with other medicines and excipients<br />

can be predicted.<br />

Continuous flow systems are valuable devices for the automation, miniaturization and<br />

preliminary operations, particularly as regards simplification of analytical processes.<br />

These systems afford the development of various chemical reactions and the<br />

implementation of reliable separation techniques with a view to increasing sensitivity and<br />

selectivity (Criado et al., 2000; Valcarcel et al., 1998;). The on-line microwave assisted<br />

22


hydrolysis followed by chemical reaction was used in the analysis of PC (Bouhsain et al.,<br />

1996).<br />

Usually it not possible to determined the PC by both batch and FIA modesin direct US<br />

spectrophotomtery in the presence of compounds that were often found along with it, due<br />

to the interference caused by them. To solve this problem occurred from the non-specific<br />

absorption in this spectral region, derivative reactions have often been used in order to<br />

give colored compounds (Ayora Canada, 2000; Murfin, & Wragg, 1972; Hassan et al.,<br />

1981).<br />

An additional optical determinative technique such as spectrofluorimetry more sensitive,<br />

also needs the use of derivative reactions (Ayora, 2000; Vilchez et al., 1995), because<br />

paracetamol was not an intrinsic fluorophor and, in any case, the determination will be<br />

quicker and cheaper than that obtained by spectrofluorimetry if direct UV<br />

spectrophotometry is used.<br />

Pereira et al., 1998, proposed a method based on the on-line microwave-assisted alkaline<br />

hydrolysis of acetylsalicylic acid to salicylic acid that reacts with Fe (III) to form a<br />

complex that absorbs at 525 nm. The precision for ten successive measurements of 200<br />

μg/ml acetylsalicylic acid presented a relative standard deviation of 0.40%. The detection<br />

limit was 4.0 μg/ml and recoveries of 99.1–101.0% were obtained for acetylsalicylic acid.<br />

Another approach described by Criado, et al., 2000, for the analysis of PC and its major<br />

metabolites using fully automated screening system in human urine samples. The<br />

detection limit reached, 0.1 mg ml -1 .The proposed method is based on direct acid<br />

23


microwave assisted hydrolysis of the drug to p-aminophenol after that the reaction with<br />

o-cresol in alkaline medium.<br />

The benefit of derivative spectroscopy has reported by Bermejo et al., 1991, for the<br />

simultaneous determination and identification of PC and plasma salicylate using second<br />

order derivative spectra after a normal extraction process (Jelena, et al., 2003). Damiani<br />

et al., 1995, also reported the applications of the first derivative spectrophotometric<br />

technique for the analysis of PC in blood serum.<br />

Ragno et al., 2004, have reported that the UV analysis of a multicomponent mixture<br />

contains tripelenamine, paracetamol, salicylamide and caffeine using partial least-squares<br />

regression (PLS) and principal component regression (PCR). The PCR and PLS models<br />

were compared and their predictive performance was inferred by a successful application<br />

to the assays of synthetic mixtures and pharmaceutical formulations (Ragno, 2004).<br />

Knochen et al., 2003 setted a extremely precise and sensitive FI method depends on the<br />

nitration of paracetamol with sodium nitrite. The method was useful to the determination<br />

of PC in oral solutions and tablets.<br />

In view of reality that a luminol based CL system is applicable due to permanganate is an<br />

oxidant and in a basic medium the oxidation of paracetamol can be done<br />

(Easwaramoorthy, 2001; Seitz & Crit., 1981; Martinez & Gomez-Benito, 1990; Vilchez<br />

et al., 1995).<br />

A novel spectrophotometric method has developed by Afshari, et al., 2001, for rapid<br />

quantification of acetaminophen in serum. Free unconjugated acetaminophen is separated<br />

24


from other endogenous interferents by extracting the drug into ethyl acetate and<br />

hydrolysis to p-aminophenol by treatment with acid and heat. The proposed method is<br />

suitable for screening of drug overdose in an emergency situation with a linearity range<br />

from 25 to 600 µgml −1 .<br />

2.4. Chromatographic Methods<br />

There were various methods has been reported for the quantification of paracetamol in<br />

biological fluids as well in pharmaceuticals mainly, liquid chromatography coupled with<br />

mass spectrometry (Lohmann & Karst., 2006; Lou et al., 2010), high performance liquid<br />

chromatography (HPLC) (Šatínský et al., 2004; Goicoechea et al., 1995; Bari et al.,<br />

1998), gas chromatography (Santos et al., 2007), and Gas Chromatography/ Mass<br />

Spectrometry (El Haj et al., 1999).<br />

However, all these process involve derivatisation or e extraction procedures. Urinary<br />

screening of paracetamol was generally carried out by using acids (Bosch, 2006; Ray et<br />

al., 1987; Davey & Naidoo, 1993) or enzymatic (Hammond et al., 1984; Edwardson et<br />

al., 1989; Dasgupta & Kinnaman, 1993). The presented methods for the determination of<br />

PC in biological fluids (urine, blood, plasma) largely use chromatographic technique<br />

(HPLC and GC) (Wong et al., 1976; Lo and Bye, 1979; Ameer et al., 1981; Al-Obaidy et<br />

al., 1995; Lau and Critchley, 1994; Goicoechea et al., 1995), and other chromatographic<br />

methods. Plasma proteins are precipitated by a sulfotungstic acid reagent, and the<br />

supernatant liquid was mixed with pyridine containing the internal standard, and<br />

determined by gas chromatography. No interference was found, while the method was<br />

suitable for 1x10 -4 M levels of paracetamol in plasma (Pegon & Vallon, 1981).<br />

25


Evans et al., 1991, determined salicylic acid in serum samples using liquid<br />

chromatography, with amperometric detection. The serum extracts was found to be 0.06<br />

mol dm –3 acetate buffer in 8% methanol (pH 5.0). The average recovery from serum was<br />

found to be 60% with a relative standard deviation of 5.8%.<br />

A easy and fast HPLC method by phenacetin as internal standard for simultaneous<br />

determination of chlorphenamine maleate, caffeine and acetaminophen in the product<br />

compound PC and chlorphenamine maleate granules (Sun et al., 2006). The accuracy,<br />

precision and linearity of the method were satisfactory for the analyzed drugs.<br />

The application of the ratio spectra derivative spectrophotometry and HPLC has been by<br />

(Erk et al., 2001) for the direct analysis of PC and methocarbamol jointly in<br />

pharmaceutical tablets. Erdal et al., 2001, developed the HPLC based first derivative ratio<br />

amplitudes method for determination of paracetamol, caffeine and propyphenazon in<br />

ternary mixtures and tablets, the amounts of caffeine and paracetamol in the ternary<br />

mixture were determined using propyphenazon as a divisor.<br />

A chromatographic method (HPLC) for the analysis of 4-aminophenol and major<br />

adulteration of paracetamol has reported by Wyszecka-Kaszuba et al., 2003. The method<br />

was sensitive up to 4 ng ml -1 and 1 ng ml -1 in capsules and tablets respectively. The<br />

proposed method was effectively useful for the determination of commercially available<br />

drugs.<br />

26


2.5. Other Techniques<br />

In addition to above techniques there are several of analytical methods existing for the<br />

analysis of PC in various types of samples. These include colorimetric (Knochen et al.,<br />

2003), capillary electrophoresis (Chu et al., 2008, Heitmeier et al., 1999), micellar liquid<br />

chromatography (Love et al., 1985), and many others.<br />

Due to the hydrolysis of PC to 4-Aminophenol, it produced a colored compound with the<br />

suitable reactions, they are time-consuming, however these kind of produres are not<br />

enough convenient for PC determination in <strong>Ph</strong>armaceutical preparation. Flow-injection<br />

(FI ) with chemiluminescense method was therefore sought to serve this purpose.<br />

Chemiluminescence methods are regarded as sensitive and selective having various<br />

advantages for pharmaceutical formulations (Townshend, 1990; Robards & Worsfold,<br />

1992; Hindson & Barnett, 2001). The analysis of PC in biological fluids by using FI with<br />

chemiluminescence based on the oxidation of paracetamol with cerium (IV) were also<br />

reported (Koukli et al., 1989; Easwaramoorthy, 2001).<br />

2.6. Quantification of Paracetamol in Biological Samples and<br />

<strong>Ph</strong>armaceuticals<br />

Several analytical methodologies have been proposed for the determination of<br />

paracetamol in pharmaceutical formulations and biological samples (Bosch et al., 2006,<br />

Fanjul-Bolado et al., 2009). Clinical (blood and urine) determination of PC formulations<br />

using urinary excretion data has been well documented (Goyal, 2006; Welch & Conney,<br />

27


1965; Wong et al., 1976; Mattok et al., 1971; Sotiropoulus et al., 1981; Vila-Jato et al.,<br />

1986; DomÃnguez et al., 2000; Su and Cheng, 2010).<br />

The combination of mefenamic acid and paracetamol was described for the simultaneous<br />

determination by (Dinç et al., 2002).<br />

Criado et al., 2000, reported the method for the analysis of PC which was further diluted<br />

and continually hydrolyzed in an alkaline medium by spectroscopic method. The average<br />

R.S.D. of 2.4% shows the good validity of the method.<br />

For the first time, a multiparameter responding flow-through system with solid phase<br />

detection (a multiparameter optosensor) was described for the Simultaneous<br />

determination of a mixture of three active principles (caffeine, paracetamol and<br />

propyphenazone) using univariate calibration by UV spectrophotometric coupled with a<br />

multiparameter optosensor detector. The application of the detector successfully<br />

determined the analyte in commercial pharmaceuticals (DomÃnguez et al., 2000).<br />

Jelena et al., 2003, reported a method to facilitate the direct and simple determination of<br />

total paracetamol in urine by UV at the wavelength range 220–400nm.<br />

The selective and simple spectrofluorimetrical method has been designed by de los et al.,<br />

2005, for paracetamol determination in tablets.<br />

Altair et al., 2005 demonstrated a rapid and simple method for direct analysis of<br />

paracetamol in the solid state pharmaceutical formulations by fluorescence. Results were<br />

28


compared with those obtained by the BP recommended method at the 95% confidence<br />

level.<br />

The FI analysis of PC was setted, based on its inhibitory effect on a luminol-<br />

permanganate chemiluminescence system (Easwaramoorthy, 2001). The paracetamol<br />

contents were analyzed in locally available pharmaceuticals. The recovery obtained was<br />

in the range of 98.2–104.4%. The viability of the method was confirmed on actual<br />

samples (Easwaramoorthy et al., 2001).<br />

Ruengsitagoon et al., 2006 developed a simple chemiluminometric method for the<br />

determination of paracetamol in pharmaceutical formulations using FI based on the CL<br />

produced by the reduction of tris (2,2-bipyridyl) ruthenium(III) (Ruengsitagoon, 2006).<br />

Pedrosa et al., 2006, reported FIA method for the analysis of PC in pharmaceutical<br />

Formulation with a modified self-assembled monolayer (SAM) gold electrode with a of<br />

3-mercaptopropionic acid. Fifteen days lifetime of the modified electrode was found.<br />

Obtained results of amperometric proposed method will found comparable with<br />

spectrophotomtery.<br />

FIA with amperometric detection has been employed by Felix et al., 2007, for<br />

quantification of paracetamol using a carbon film resistor electrode. This sensor exhibited<br />

sharp and reproducible current peaks for acetaminophen without chemical modification<br />

of its surface.<br />

Cervini et al., 2008, evaluated a bare graphite-polyurethane composite as an<br />

amperometric FI detector for the quantification of paracetamol in pharmaceuticals. The<br />

29


esults showed that graphite-polyurethane composite can be used as an amperometric<br />

detector for flow analysis in the proposed determination.<br />

Santos et al., 2008, reported a simple and fast approach for simultaneous determination in<br />

pharmaceutical formulations of ascorbic acid and PC using FI method with multiple pulse<br />

amperometric detection. In this method chemometric technique were also employed for<br />

the better achievement of the results.<br />

A novel type of modified glass carbon electrodes with the nickel magnetic nanoparticles<br />

was made-up to determine the electrochemical properties of N-acetyl-p-aminophenol.<br />

Differential pulse voltammetry (DPV) was used for the determination of ACOP in<br />

effervescent dosage samples (Wang et al., 2007).<br />

Safavi et al., 2008, investigated an easy electrochemical method for the simultaneous<br />

determination of p-aminophenol and paracetamol in drugs. The peak potentials (oxidation<br />

and reduction) in cyclic voltammetry for paracetamol on carbon ionic liquid electrode<br />

were significantly improved in contrast to the usual carbon paste electrode.<br />

Zhao et al., 2006 reported the application of the indirect determination of paracetamol by<br />

a capillary electrophoresis–chemiluminescence (CE–CL) detection based on its inhibitory<br />

effect on a luminol-potassium hexacyanoferrate (III).<br />

Santhosh et al., 2009, determined the concentration of paracetamol in human blood<br />

plasma and commercial drugs by modification of Au electrode with tetraoctylammonium<br />

bromide stabilized gold nanoparticles attached to 1,6-hexanedithiol. (ATR)-FT-IR were<br />

use to confirmed the modified Au surface.<br />

30


Recently Lou Hong-gang et al., 2010, developed a highly sensitive and simple LC–<br />

MS/MS method after one-step precipitation for the simultaneous determination of<br />

pseudoephedrine, paracetamol, chlorpheniramine and dextrophan,, in human plasma<br />

using diphenhydramine as internal standard. The method was successfully validated for<br />

the determination of pharmaceutical formulation.<br />

2.7. Quantification of Aspirin in <strong>Ph</strong>armaceutical and Biological Samples<br />

Aspirin was begin famous in the late 1890s and has been deal with range of inflammatory<br />

conditions (Supalkova et al., 2006). Many folk remedies used since pre-historic times<br />

have depended upon salicylates for their effect. 100 years ago aspirin was prepared from<br />

acetic and salicylic acids. It was the first drug to be synthesized and its formulation is<br />

regarded as the base of the current pharmaceutical industry.<br />

Commonly, acetylsalicylic acid (ASA) is indirectly determined after its conversion to<br />

salicylic acid (SA) and acetic acid by alkaline hydrolysis according to a British<br />

<strong>Ph</strong>armacopoeia method, (British <strong>Ph</strong>armacopoeia, 1980). Another reference method for the<br />

determination of salicylic acid was the spectrophotometric method based on the Trinder<br />

reaction, (Trinder, 1954).<br />

The SA was the most important metabolite of the ASA, which obtained by the hydrolysis.<br />

A large number of analytical approaches such as potentiometry (Kubota et al., 1999),<br />

spectrophotometry (Sena et al., 2000; Merckle & Kovar 1998; Loh et al., 2005; Vidal et<br />

al., 2002; Ruiz- medina et al., 2001), amperometry (Rover et al., 2000; Rover et al.,<br />

1998; Pasekova et al., 2001), chromatography (Nogowska et al., 1999), UV detection<br />

31


(Glombitza & Schmidt, 1994; Matias et al., 2004) and fluorescence (Martos et al., 2001),<br />

spectrofluorimetry (Criado et al., 2000; Arancibia et al., 2002; Moreira et al., 2004),<br />

methods have been also expressed for the analysis of ASA and SA in pharmaceutical<br />

formulation. Conversely it seems that they are not commonly used, maybe due the<br />

tedious and difficult sample preparations, usually liquid chromatographic methods were<br />

used (Pirola et al., 1998; Franeta et al., 2002; Šatínský et al., 2004; Stolker et al., 2004).<br />

Some time atomic absorption spectroscopic methods were also used but such methods are<br />

expensive and need extraction procedures. However ion selective electrode showing high<br />

specificity, good detection limit and comparably cheapest (Pasekova et al., 2001). Some<br />

other electrode has been also used for the determination aspirin such as nickel oxide-<br />

modified nickel electrode in alkaline solution (Kowal et al., 1997; Jafarian et al., 2003;<br />

Yousef Elahi et al., 2006; Majdi et al., 2007), and glassy carbon electrode (Houshmand et<br />

al., 2008). A number of flow injection spectrophotometric methods were also use on-line<br />

hydrolysis (Koupparis & Anagnostopoulou, 1988; Quintino et al., 2002; Catarino et al.,<br />

2003; Quintino et al., 2004, Garrido et al., 2000; Rover et al., 1998).<br />

Many developed methods has shows benefit regarding this containing some limitation<br />

also for the particular demand of analysis. In contrast with methods, electrochemical<br />

sensors are selective option for electroactive species because of high sensitivity and<br />

economical (Prasek et al., 2006, Babula et al., 2006).<br />

Supalkova et al., 2006, developed the method for indirect determination of acetylsalicylic<br />

acid in pharmaceutical drug. Electrochemical sensor for acetylsalicylic detection was also<br />

32


suggested. SWV using both CPE and graphite pencil as working electrode was<br />

successfully applied for determination of ASA.<br />

Majdi et al., 2007 and Houshmand et al., 2008, studied the electrocatalytic oxidation of<br />

aspirin on a nickel oxide-modified nickel electrode and nanoparticles of cobalt hydroxide<br />

electro-deposited on the surface of a glassy carbon electrode respectively in alkaline<br />

solution by chronoamperometry, cyclic voltammetry and electrochemical impedance<br />

spectroscopy techniques as well as steady-state polarization measurements. The<br />

investigative facility helped for the determination of aspirin by using of modified<br />

electrodes. The method was proven to be valid for analyzing these drugs in urine samples<br />

(Houshmand et al., 2008).<br />

López-Cueto et al., 2002, proposed that when ASA was added to a bromine solution,<br />

slow decompose of the bromine concentration occurs, while hydrolysis of aspirin yields<br />

SA slowly, and bromine reacts quickly with salicylic acid. This behavior can be utilized<br />

to develop kinetic methods for resolution of mixtures of SA and PC.<br />

Matias et al., 2004, described the quantitative reflectance spot test procedure for the<br />

analysis of acetylsalicylic acid in pharmaceutical formulations. In this method the<br />

reaction of SA were carried out by the hydrolysis of acetylsalicylic acid with Fe (III)<br />

forming a deep blue-violet complex. Drugs having acetylsalicylic acid can be easily<br />

determined by the suggested method without any separation.<br />

Šatínský et al., 2004 reported the simultaneous determination of paracetamol, caffeine,<br />

acetylsalicylic acid, and internal standard benzoic acid on a novel reversed-phase<br />

33


sequential injection chromatography technique with UV detection. The analysis time was<br />

about 6 min. The method was found to be applicable for the routine analysis of the active<br />

compounds paracetamol, caffeine, and acetylsalicylic acid in pharmaceutical tablets.<br />

A simple and rapid analytical procedure was proposed by Sena & Poppi 2004, for<br />

simultaneous determination of caffeine paracetamol and acetylsalicylic acid by UV<br />

spectrophotometric method using multivariate calibration and measurements at 210–300<br />

nm.<br />

The on-line microwave assisted alkaline hydrolysis of acetylsalicylic acid has been<br />

developed by Pereira et al., 1998, where the hydrolysis product (salicylic acid) reacts<br />

with iron (III) to form a complex that absorbs at 525 nm. The method was applied to the<br />

determination of the drug in tablets.<br />

Tsikas et al., 1998, described gas chromatographic–tandem mass spectrometric method<br />

for the accurate analysis of ASA in human plasma after a only low-dose oral<br />

administration of 2 guaimesal or aspirin, an acetylsalicylic acid releasing pro drug.<br />

Hansen et al, 1998, studied the separation three metabolites such as salicyluric, and<br />

gentisic acid of acetylsalicylic acid in a non-aqueous capillary electrophoresis system<br />

with reversed electro osmotic flow.<br />

Amperometric biosensor for the salicylate determination in blood serum has been<br />

described by Rover et al., 2000.<br />

34


The quantification and separation of the anti-nerve agent pyridostgmine bromide, the<br />

analgesic drugs ASA and acetaminophen, and the stimulant caffeine in rat plasma and<br />

urine has been reported by Abu-Qare et al., 2001. The resulting chromatograms showed<br />

no interfering peaks from endogenous plasma or urine components.<br />

2.8. Quantification of Diclofenac Sodium in <strong>Ph</strong>armaceutical and<br />

Biological Samples<br />

Different analytical methods have been employed for the quantification of diclofenic<br />

sodium (DS), such as spectrophotometry (Matin et al., 2005), fluorimetry (Aranciba et<br />

al., 2000), FT-Raman spectroscopy (Mazurek & Szostak, 2006; 2008), potentiometry<br />

(Pimenta et al., 2002), chromatography (Lala et al. 2002), voltammetry (Yang et al.,<br />

2008) and polarography (Xu et al. 2004). Most of these methods face certain problems<br />

such as the use of additional reagents, complex formation, long time and hazardous<br />

matrices.<br />

A method has been devised by Carreira et al., 1995, for the analysis of diclofenac sodium<br />

in bulk using Eu 3+ ions as the Fluorescent probe. The method was developed around the<br />

hypersensitive property of the transitions of the fluorescent probe ion at 616 nm.<br />

Fernandez de Coardova et al., 1998, have reported spectrophotometric method which was<br />

a sensitive, easy and very selective for the quantification of diclofenac sodium in<br />

authentic pharmaceutical drugs.<br />

Gonzalez et al., 1999, developed a method for the simultaneous determination of<br />

diclofenac, betamethasone, and cyanocobalamin (Vitamin B12), by HPLC. Linearity,<br />

interday precision and accuracy for each active ingredient were analyzed.<br />

35


Klimes et al., 2001, have reported the HPLC methodology to select a suitable acceptor<br />

medium for permeation experiments and also determined the release of diclofenac and its<br />

in vitro permeation through the human skin.<br />

Lala et al., 2002, has been developed the method using high performance thin layer<br />

chromatographic for the determination of diclofenac sodium from serum. Standard<br />

diclofenac sodium was spotted on silica gel precoated plates using the mobile phase<br />

toluene:acetone:glacial acetic acid.<br />

Tubino & Rafael, 2006, reported a method for quantification diclofenac in<br />

pharmaceutical drugs by diffuse reflectance. The finding results were compared with the<br />

HPLC procedure recommended by the USP.<br />

Rodriguez et al., 2007, investigated a tubular bismuth film electrode, installed as part of a<br />

multisyringe FI system and utilized it as an amperometric detector for analysis of<br />

diclofenac sodium in drugs samples.<br />

Payán et al., 2009, described an extraction method using a polypropylene membrane<br />

supporting dihexyl ether for the analysis of some analgesic formulations in human urine<br />

samples by HPLC using a monolithic silica column such as ibuprofen, salicylic acid and<br />

diclofenac.<br />

Blanco-López et al., 2003, have developed a method for the determination of diclofenic<br />

sodium by voltammetric sensor based on the molecular recognition of the analyte by<br />

36


molecularly imprinted methacrylate ethyleneglycol dimethacrylate co-polymers. The best<br />

results were obtained in 0.025 M citrate solution (pH 6) containing 10% of acetonitrile.<br />

Fernández-Llano et al., 2007, reported the molecularly imprinted polymer for diclofenac,<br />

prepared thermal polymerization over silica beads using 2-(dimethylamino) ethyl-<br />

methacrylate as functional monomer. A selective solid-phase extraction of the drug from<br />

urine followed by its quantification by ADPV.<br />

Goyal et al., 2010, proposed the voltammetric determination of diclofenac using SWV at<br />

edge plane pyrolytic graphite (EPPG) sensor from real urine samples. Jin & Zhang, 2000<br />

have developed the for the detection of diclofenic sodium in urine sample by using<br />

capillary zone electrophoresis using carbon fiber microelectrode, at a constant potential.<br />

Xu et al., 2004 have investigated the polarographic response characteristics of diclofenac<br />

sodium in 0.25M HAc-NaAc (pH 5.0) supporting electrolyte in the absence and the<br />

presence of dissolved oxygen.<br />

Mazurek & Szostak, 2006, have studied the FT-Raman quantification of minophylline<br />

and diclofenac sodium. The efficiency of many spectra treatment protcols including<br />

multivariate partial least squares and classical univariate intensity ratio and principal<br />

component regression (PCR) methods was compared.<br />

Mazurek & Szostak, 2008, have reported the method for quantitative determination of<br />

diclofenac sodium in capsules and tablets by FT-Raman. PLS, PCR counter-propagation<br />

artificial neural networks (CP-ANN) strategies were also emlyed in this method for its<br />

fastness and convenient to the other pharmacopoeial methods. Wang et al., 2009, have<br />

37


investigated a method for quantitative analysis of diclofenac sodium powder on the basis<br />

of NIR spectroscopy.<br />

Hajjizadeh et al., 2007, have developed a sensitive and simple amperometric procedure<br />

for the analysis of mefenamic acid, diclofenac and indomethacin in bulk form and for the<br />

direct assay of tablets, using the NHMN electrode. The electrocatalytic oxidation of these<br />

drugs was examined on a nickel hydroxide-modified nickel electrode in alkaline solution.<br />

Another sensitive, simple, and time-saving amperometric approach by Heli et al., 2009,<br />

have investigated based on the electro-oxidation of several NSAIDs such as mefenamic<br />

acid, diclofenac and indomethacin on nanoparticles of Ni–curcumin-complex-modified<br />

glassy carbon electrode in alkaline solution. Atomic force microscopy and scanning<br />

electron microscopy were used for surface studies.<br />

Yilmaz, 2010, developed a procedure based on GC-MS for the determination of<br />

diclofenac in human plasma. This assay was successfully applied in Turkey to healthy<br />

volunteers after an oral administration of 50 mg diclofenac.<br />

García et al., 1998, have reported a rapid FI spectrophotometric method for the analysis<br />

of diclofenac sodium in pharmaceuticals and urine samples.<br />

Pimenta et al., 2002, have reported the two autonomous methods for the analysis of<br />

diclofenac simultaneously applied in an automated analytical system, based on the<br />

concept of sequential injection determination, providing real-time assessment of results<br />

quality. ISE based on cyclodextrin were used for the potentiometric detection.<br />

38


Pérez-Ruiz et al., 1997 reported the spectrophotometric determination the trace quantity<br />

of diclofenic were determined by liquid–liquid extraction. Furthermore New, rapid and<br />

accurate spectrophotometric method for the analysis of diclofenac sodium in drugs<br />

sample which is based the reaction of nitric acid (Conc. at 63% w/v) proposed by Matin<br />

et al., 2005.<br />

2.9. Quantification of Ibuprofen in <strong>Ph</strong>armaceutical and Biological<br />

Samples<br />

A rapid quantitative analysis of ibuprofen in plasma of human blood was carried by<br />

Jonkman et al., 1985, using a SFE on “Baker” C-18 disposable extraction column. The<br />

proposed method was a simple and sensitive.<br />

Stefan and Heitmeier & Blaschke, 1999, have investigated reliable screening method that<br />

allows determination of the drugs its metabolites in human urine after oral administration<br />

(Heitmeier, 1999).<br />

Damiani et al., 2001, have described the determination of ibuprofen in pharmaceutical<br />

creams, tablets and syrups without any interference of the excipients by<br />

spectrofluorimetric method. It involved emission at 288 nm and excitation at 263 nm. The<br />

linear range was 2–73 mgL −1 .<br />

Palabiyik et al., 2004, described the spectrophotometric methods for the simultaneous<br />

determination of pseudoephedrine hydrochloride and ibuprofen in their combination. The<br />

range 100–1300 µg ml −1 for pseudoephedrine hydrochlorides and 300–1300 µg ml −1 was<br />

found for ibuprofen. Hamoudová et al., 2006, have illustrated that the capillary zone<br />

electrophoresis with by using spectrophotometric for the determination of flurbiprofen<br />

39


and ibuprofen. A fused silica capillary with UV detection was used for separation which<br />

carried out at 232 nm. The method was successfully validated for the analysis of 10<br />

commercially available tablets, syrup, gel and cream. Hassan, 2008, presented three<br />

methods for simultaneous analysis of PC and ibuprofen without any earlier separation. 1 st<br />

derivative UV with zero-crossing measurement was the first method. Second depends on<br />

1 st derivative of the ratio-spectra by measurements. 3 rd method was depends on<br />

multivariate spectrophotometric calibration for the simultaneously analysis of<br />

components.<br />

Costi et al., 2008, have proposed the practically and quantitative solvent-free solid-phase<br />

extraction (SPE) of ibuprofen and naproxen from sewage samples. Khoshayand et al.,<br />

2008, have reported the simultaneous determination of caffeine paracetamol and<br />

ibuprofen in pharmaceuticals by chemometric approaches using UV spectrophotometry.<br />

Rapid, simple and accurate economical spectrophotometric method for simultaneous<br />

determination of PC and ibuprofen in combined soft gelatin capsule dosage form has<br />

been developed using two wavelengths at 224 and 248 nm simultaneously (Riddhi et al.,<br />

2010).<br />

Sochor et al., 1995, described the HPLC method for the analysis of ibuprofen in plasma<br />

and isolated erythrocytes. C-18 column were for the Practical and methanol-water<br />

(220:100, v/v) were used as mobile phase.<br />

40


A HP-TLC method for quantitative determination of ibuprofen in plasma was explained<br />

(Save et al., 1997). The limit of detection 50 ng was found for ibuprofen from human<br />

plasma.<br />

Kang et al., 1998, studied the quantitative aspects of HPLC with a column-switching<br />

system and CE for the determination of ibuprofen in plasma. Farrar et al., 2002,<br />

developed a fast and easy method of analysis the ibuprofen in small volumes of human<br />

plasma (50 µl) by HPLC. The calibration curve was linear and limit of quantitation of<br />

1.56 µg/ml.<br />

De Oliveira et al., 2005, described an easy, sensitive and fast off-line solid-phase<br />

microextraction process coupled with HPLC for the stereoselective analysis of the major<br />

metabolites of ibuprofen in human urine samples.<br />

Agatonovic-Kustrin et al., 2000, studied the enantiomeric purity of ibuprofen in a simple<br />

manner by diffuse reflectance infrared Fourier transform (DRIFT) spectroscopy with<br />

artificial neural networks (ANNs) methodology. Russeau et al., 2009, have used the<br />

ATR-FTIR and target factor analysis to explore the pharmaceutical gel containing<br />

ibuprofen in human skin. Results revealed that the data were effectively correlated<br />

between reference spectra of the components and factors from the data.<br />

Hergert & Escandar, 2003, have studied the complexation of ibuprofen in ß-cyclodextrin<br />

using spectrofluorimetry at both acid and alkaline pH successfully.<br />

Valderrama & Poppi, 2010, reported the spectrofluorimetric second order standard<br />

addition method for the determination of ibuprofen in human plasma and urine. The<br />

methodology was based on chiral recognition of ibuprofen by formation of complex with<br />

41


a β-cyclodextrin, in the presence of 1- butanol. The obtained results were in the molar<br />

fraction range from 50 to 80% of ibuprofen, providing absolute errors lowers than 4.0%<br />

for plasma and urine. Glówka, & Karazniewicz, 2005, developed a method for the<br />

quantitative determination of ibuprofen in biological matrices, human serum and urine<br />

using direct and stereospecific capillary zone electrophoresis (CZE). Bauza et al., 2001,<br />

used in situ chiral derivatisation to get diastereomeric amides of ibuprofen for their<br />

subsequent extraction with supercritical carbon dioxide. Urine sample containing<br />

ibuprofen were spiked to reveal the suitability of this method<br />

Sádecká et al., 2001, quantified ibuprofen and naproxen in tablets by capillary<br />

isotachophoresis. Linearity was from 40.0 to 200.0 mg L −1 of ibuprofen, with a<br />

coefficient of determination of 0.999.<br />

Tapan et al., 2010, have developed simple reproducible simultaneous equation method,<br />

requiring no prior separation, for the assessment of PC and ibuprofen in combined dosage<br />

form. Ibuprofen has absorbance maxima at 220 nm and paracetamol at 248.40 nm using<br />

ethanol as solvent. The method obeys Beer’s Law in concentration ranges employed for<br />

the estimation.<br />

Recently Issa et al., 2010, reported a simple, rapid and accurate method for the<br />

simultaneous spectrophotometric determination of ibuprofen and paracetamol in two<br />

components mixture and Cetofen tablets. Calibration graphs were linear in the range 2–32<br />

µg ml −1 (LOD 0.53 µg ml −1 ) and 2–24 µg ml −1 (LOD 0.57 µg ml −1 ) ibuprofen and<br />

paracetamol respectively.<br />

42


EXPERIMENTAL<br />

3.1. Material and Methods for Paracetamol using UV-Visible<br />

Spectrophotometry<br />

3.1.1. Reagents and Chemicals<br />

Chapter-03<br />

Acetonitrile, chloroform, 2-propanol, hydrochloric acid, ammonia, sodium hydroxide,<br />

paracetamol (PC), acetyl salicylic acid, ibuprofen, caffeine, diclofenic sodium, with<br />

>99% purity were obtained from Merck (Germany). Ascorbic acid, sodium sulfide,<br />

ammonium iron (III) sulfate, sulfuric acid and ethanol were from Fluka Chemicals.<br />

Methanol was obtained from a local sugar industry and triply distilled before use.<br />

Doubly distilled water was used throughout for final washings and preparations of all<br />

aqueous solutions. Aqueous PC stock solution was prepared by warming the solution<br />

for 10–20 min at a boiling water bath until complete dissolution of the analyte. The<br />

solution was then cooled to room temperature and made up with doubly distilled<br />

water.<br />

3.1.2. Instruments and Apparatus<br />

Lambda 2 UV/visible spectrometer of Perkin-Elmer company was used for the effect<br />

of all parameters and analysis of samples by recording the respective absorbance<br />

value at definite wavelength. FTIR spectra of the respective reagents and compounds<br />

were recorded by means of a Nicolet Avatar Model 330 FTIR of Thermo Electron<br />

Corporation. Temperature adjustment during experiments was carried out with<br />

controlled temperature Water Bath, Model, GMBH D-7633, Julabo HC5, Germany in<br />

which a netted tray was fitted for test tube holding in vertical position.<br />

43


3.1.3. Procedure for Determining PC<br />

PC was determined in dilute standard solution after taking it in the specific 1 cm<br />

quartz cell against a blank (doubly distilled water) at a λmax value of 243 nm by<br />

recording the respective signal or absorbance value using a controlled unit. A<br />

calibration range was set for PC by recording the spectra of various standard<br />

solutions. The locally available tablets containing PC were also analyzed by the same<br />

procedure after dissolution of five replicate portions (each tablet containing 0.01<br />

g/100 mL water) randomly selected from weighed three powdered tablets of each<br />

manufacturer company in the similar manner as accurate for preparing standard PC<br />

stock solution. During temperature related experiments, the temperature of PC and<br />

blank solutions were first regularized with controlled temperature water bath and then<br />

transferred to quartz cells very smartly but carefully for quick UV analysis. After<br />

several experiments it was found that there was a variation of ±1 ◦C temperature. The<br />

dilute samples analyzed were evaluated for their PC contents after fitting their average<br />

of replicate spectra in calibration plot and multiplying the result with dilution factor.<br />

3.1.4. Analysis of PC in Urine Samples<br />

Collection of urine samples from four volunteers was conducted in the same way as<br />

described elsewhere [Jelena et al., 2003]. Each individual was instructed not to use<br />

any medication before two weeks of urine collection. The urine samples were then<br />

collected in thoroughly washed and clean plastic bottles in the morning time from full<br />

bladder without any dose of PC. After this, each individual was instructed to eat the<br />

same diet he has used yesterday. Each of them was administered with a PC tablet of a<br />

specific brand containing 500 mg PC after 12 h of previous urine collection. The next<br />

day the urine samples were collected after 12 h in the same way as true for samples<br />

without PC. After same proper dilutions, the samples were processed for UV<br />

44


spectrophotometric determination of PC with the procedure described earlier by<br />

taking the diluted sample without PC as blank and PC containing as the actual sample.<br />

The difference gave the PC contents in the urine sample which was converted to<br />

actual value after adjusting for dilution factor.<br />

3.2. Material and Methods for Diclofenac Sodium using UV-Visible<br />

Spectrophotometry<br />

3.2.1 Apparatus<br />

The apparatus were used as same describe in section 3.1.2.<br />

3.2.2. Washing of Glassware<br />

All glassware was washed by soaking in 3 M HNO3 overnight followed by washing<br />

with detergent water. It was then thoroughly washed with tap water and finally rinsed<br />

at least 3 times with doubly distilled water. The glassware was then dried in an oven<br />

at 110 0 C.<br />

3.2.3. Reagents and Solutions<br />

All the reagents used in this study were of analytical grade ultra pure quality from<br />

Merck, Fluka and BDH, etc. Diclofenac sodium (DS) and other pharmaceutical<br />

standards were provided by Birds Chemotec Karachi, Pakistan. The identity of<br />

pharmaceutical standards was checked by Fourier Transform Infrared spectra and<br />

comparing these with the relevant data found in literature. Stock standard solution<br />

(w/v) of diclofenac sodium (1 mgmL -1 ) was prepared in 100 mL calibrated volumetric<br />

flask and diluted to the mark with doubly distilled water. Dilute working standards<br />

were prepared from time to time as per requirement. Solutions of other reagents were<br />

also prepared in doubly distilled water in the desired concentration.<br />

45


3.2.4. Procedure for Determining DS in Tablets<br />

Four brands of tablets containing DS from different manufacturers were purchased<br />

from local market and analyzed by using the current method. Ten tablets from each<br />

brand were finely powdered and mixed. An amount equal to the average weight of<br />

one tablet was collected randomly, transferred to a 100 mL volumetric flask,<br />

dissolved and made up with doubly distilled water. Dilute solutions were made from<br />

each sample and analyzed by UV-Vis Spectrometer. The concentration of DS per<br />

tablet was calculated with the help of equation for linear calibration curve of DS and<br />

multiplying with relevant dilution factor.<br />

3.2.5. Procedure for DS in Serum and Urine Samples<br />

The blood and urine samples were collected before the intake of DS (blank) at day 1<br />

and after taking DS at day 2. The individuals were instructed not to use any analgesics<br />

including DS one week before examination and use the same common diet at day 1<br />

and day 2. The DS was administered just after the collection of blank urine samples.<br />

The blood samples were collected after 2 hour of dosage while urine samples were<br />

collected after 12 hours of administration of 50 mg of tablet. Serum was obtained as<br />

supernatant from blank and DS containing clotted blood samples by centrifugation<br />

method. In order to freed the urine and serum sample from water insoluble impurities,<br />

1 mL of blank as well as DS containing sample were passed through a column DSC-<br />

18 (used in solid phase extraction), which was pre-washed with 2 mL of methanol.<br />

The recovered urine or serum sample was diluted to 10 mL with doubly distilled<br />

water and analyzed for DSF contents by UV-Visible spectrometer taking water as<br />

actual blank (reference) to record the spectra of blank and DS containing serum and<br />

urine samples. The difference of absorbance between blank and actual sample gave<br />

the concentration of DS after fitting the value in linear equation and multiplying the<br />

46


esult with dilution factor. This treatment was carried out due to the difficulty in<br />

finding out the absorbance in case of taking actual serum or urine sample as blank<br />

against DS containing serum or blood sample. Standard solution of DS was spiked to<br />

each serum or urine sample in order to confirm the peak of DS.<br />

3.3. Material and Methods for the Determination of Paracetamol using<br />

Differential Pulse Voltammetry<br />

3.3.1. Reagents and Solutions<br />

The stock solution of Paracetamol (N-acetyl-4-aminophenol, purity 98%, Aldrich,<br />

Germany) (c = 1×10 -3 mol.L -1 ) was prepared by means of dissolving 0.0076 g of the<br />

substance in 50 mL of de-ionized (DI) water. Further dilutions were prepared by<br />

required volume of the stock solution with DI water. All solutions were stored in the<br />

dark, at laboratory temperature and in glass vessels. Other chemicals such as boric<br />

acid, glacial acetic acid, phosphoric acid, sodium hydroxide, all % purity were<br />

purchased from Lachema Brno, Czech republic and potassium chloride was<br />

purchased from Lach-Ner, Czech republic. MilliQ plus system (Millipore, USA) was<br />

used for deionized water. The conductive carbon ink solution was arranged by<br />

combination 0.01 g of polystyrene (expanded – packaging box, Merck, Germany),<br />

0.09 g of carbon powder (crystalline graphite 2 µm, CR 2 Maziva Tyn, Czech<br />

Republic) in 0.5 mL dichlorethane (purity 99.5%, Merck, Germany). The mixture was<br />

thoroughly homogenized by agitation.<br />

3.3.2. Apparatus<br />

Eco-Tribo Polarograph coupled with POLAR PRO software version 5.1 (Polaro-<br />

Sensors, Prague) was employed for all voltammetric measurements. The instrumental<br />

software joined under the operational system Microsoft Windows XP (Microsoft<br />

Corp.). All measurements were carried out in a three-electrode system. Silver/silver<br />

47


chloride reference electrode ETP CZ R00308 (1 mol.L -1 KCl, Monokrystaly Turnov,<br />

Czech republic), platinum wire as an auxiliary electrode ETP CZ P01406, and CFE<br />

(based on AgSAE, no. 2-05-19 from Eco-trend Plus, Czech Republic, disk diameter<br />

0.5 mm, covered by carbon ink film) as a working electrode were used.<br />

The optimized parameters such as scan rate 20 mV.s -1 , the pulse amplitude 50 mV,<br />

sampling time of 20 ms beginning at 80 ms after the onset of the pulse were used.<br />

Better reproducibility of voltammetric measurements at CFE was assured by a<br />

suitable electrochemical regeneration [Fischer et al., 2007] of CFE. Optimal results<br />

were obtained for pH 4 with electrochemical regeneration by the application of<br />

periodical switching every 0.1 s between potentials –400 mV (Ereg1) and 1300 mV<br />

(Ereg2) in the given measured solution for 30 s (150 cycles).<br />

3.3.3. Procedures<br />

The voltammetric measurements were carried out by taking appropriate amount (1 µL<br />

– 1000 µL). Paracetamol solution in water was added into a 10 mL volumetric flask,<br />

which was filled up to the mark with corresponding Britton-Robinson buffer and then<br />

transferred into an electrochemical cell. Oxygen was removed from measured<br />

solutions by purging with nitrogen for 5 minutes.<br />

Current of peak was measured from the straight-line connecting minima on both sides<br />

of the peak. Current of background electrolyte was subtracted from current of peak.<br />

The calibration curves were measured in triplicate and evaluated by the least squares<br />

linear regression method. Limit of determination (LOD) was calculated using a<br />

10S/slope ratio, where S is the standard deviation of the mean value for 12 analyte<br />

determinations at the concentration corresponding to the lowest peak on the<br />

48


appropriate calibration straight line according to IUPAC recommendations (Inczedy et<br />

al., 1998). All the measurements were carried out at laboratory temperature.<br />

Determination of Paracetamol in pharmaceutical samples was carried out by standard<br />

addition method. Given tablet was dissolved in 100 mL of deonized water and 100 µL<br />

of this solution was diluted by Britton-Robinson buffer (pH 4.0) up to 10 mL and used<br />

for voltammetric measurement, and standard additions of Paracetamol stock solution.<br />

For testing of the quantitative investigation of paracetamol in samples of human urine<br />

the method of calibration curve was used. Calibration curves and series of<br />

determination for analyzed drug in model solutions of human urine were determined.<br />

3.4. Material and Methods for the Analysis of Ibuprofen using FT-IR<br />

Spectroscopy<br />

3.4.1. Reagents and Samples<br />

The stock solution of Ibuprofen purity 99%, (Merck, Germany) (c = 1000 ppm) was<br />

prepared by means of dissolving 0.1 g of the substance in 100 mL of chloroform<br />

99.98 % purity (Fisher scientific UK limited). Further dilutions were prepared by<br />

required volume of the stock solution with chloroform. All solutions were stored in<br />

the dark, at laboratory temperature and in glass vessels with tight cover. The different<br />

pharmaceutical tablet samples containing ibuprofen as an active component were<br />

purchased from pharmacy unit of Hyderabad, Pakistan.<br />

3.4.2. FT-IR Spectral Measurements<br />

For the obtaining of infrared spectra of standard, tablet and urine samples, FTIR<br />

spectrometer (Thermo Nicolet 5700) was employed fitted with detachable liquid cell<br />

(KBr) and DTGS detector was attached. OMNIC software version 7.3 was used to<br />

control the instrument. The mid-IR spectral range from 4000 cm -1 to 400 cm −1 was<br />

49


selected, total 16 scans with the resolution of 4 cm −1 . A new background spectrum of<br />

chloroform was taken before recording the spectra of all sample and standard.<br />

3.4.3. FT-IR Calibration<br />

Ten standard of Ibuprofen ranging from 10 to 100 ppm in chloroform were prepared<br />

for biological fluids and pharmaceutical samples and the samples were quantitatively<br />

determined by using Turbo Quant (TQ) analyst software.<br />

The already recorded spectra by Omnic software of ibuprofen standards were opened<br />

in TQ analyst program for the selection of particular region of carboxylic peak (1807-<br />

1461 cm -1 ). Some peak parameters such as peak height, peak width for all Ibuprofen<br />

standards were selected and these were calculated by TQ software which makes the<br />

use of FI-IR as powerful analytical technique for both qualitative and quantitative<br />

analysis also obtained excellent calibration curve between actual and predicted value.<br />

3.4.4. Sample Preparation Procedure<br />

This method requires only grinding of tablet samples followed by dissolution in<br />

chloroform for FTIR measurement. After weighing the tablet samples were grinded to<br />

fine powder in mortar to minimize the particle size. Quantitative analyses of solutions<br />

of ibuprofen in chloroform were performed in a cell with KBr windows and variable<br />

optic pathway (Wilks). The KBr windows were scanned from 4000 cm -1 to 400 cm -1<br />

on Thermo Nicolet 5700-FTIR spectrometer.<br />

3.4.5. Collection and Preparation of Urine Samples<br />

Urine samples were collected from healthy volunteers. 2 ml of urine sample passed<br />

through a column DSC-18 (used in solid phase extraction), which was pre-washed<br />

with 2 ml of methanol. Then sample was washed with 5 ml of 5% methanol followed<br />

50


y 1 ml chloroform. After washing the urine sample was spiked with known<br />

concentration of ibuprofen.<br />

3.5. Material and Methods for Investigation of Aspirin using Voltammetry<br />

3.5.1. Reagents<br />

The stock solution of acetylsalicylic acid and salicylic acid (Aldrich Chem. Co.)<br />

(c = 1×10 -1 mol.L -1, c = 1×10 -3 mol.L -1 ) were prepared respectively in de-ionized (DI)<br />

water. Further dilutions were prepared by required volume of the stock solution with<br />

DI water. All solutions were stored in the dark, at laboratory temperature and in glass<br />

vessels. Other chemicals boric acid, glacial acetic acid, phosphoric acid, sodium<br />

hydroxide, all p.a. purity were supplied by Lachema Brno, Czech republic and<br />

potassium chloride was supplied by Lachner Tovarni Neratovice Czech republic.<br />

MilliQ plus system was used for deionized water. The conductive carbon ink solution<br />

was arranged by combination 0.01 g of polystyrene, 0.09 g of carbon powder<br />

(crystalline graphite 2 µm, CR 2 Maziva Týn, Czech Republic) in 0.5 mL<br />

dichloreathane (purity 99.5% Merck). The mixture was thoroughly homogenized by<br />

agitation.<br />

3.5.2. Apparatus<br />

Eco-Tribo Polarograph coupled with POLAR PRO software version 5.1 (Polaro-<br />

Sensors, Prague) was employed for all voltammetric measurements. The software<br />

connected with computer system Microsoft Windows XP (Microsoft Corp.) and P-<br />

LAB a.s. Stuart Block heater model no (SBH 130 DC) was used for the hydrolysis of<br />

Acetysalicylic acid.<br />

51


3.5.3. Voltammetric Procedure<br />

All measurements were carried out in a three-electrode system. Silver/silver chloride<br />

reference electrode ETP CZ R00308 (1 mol.L -1 KCl, Monokrystaly Turnov, Czech<br />

republic), platinum wire as an auxiliary electrode ETP CZ P01406, and CFE (based<br />

on AgSAE, disk diameter 0.5 mm, covered by carbon ink film) No. 2-05-19 as a<br />

working electrode were used. The optimized parameters including the pulse amplitude<br />

50 mV, pulse width 80 ms and scan rate 20 mV.s -1 were used.<br />

Better reproducibility of voltammetric measurements at CFE was assured with a<br />

proper electrochemical activation of CFE at 2200 for 120 seconds. The Optimum<br />

results were achieved by the application of electrochemical regeneration potential<br />

between same peak potential at 750 mV.<br />

3.5.4. Indirect Determination of ASA<br />

After hydrolysis of ASA give DPV signal on the surface of CFE, according to<br />

increasing pH and temperature of Britton-Robinson buffer. 1 mL ASA (900 µL of<br />

Britton-Robinson buffer pH 12 and 100 µL of of ASA); (total concentration of ASA<br />

was 0.2-100 μmol -1 L) were hydrolyzed at 90 °C for 60 min and analyzed by DPV on<br />

the surface of CFE in the presence of 9ml Britton-Robinson buffer (pH= 2) up to total<br />

volume of 10 mL.<br />

3.5.5. Indirect Determination of ASA in <strong>Ph</strong>armaceutical Drugs and Urine<br />

Samples by DPV at CFE<br />

The above-mentioned method for indirect determination of ASA has been applied for<br />

analysis of pharmaceutical drugs. The tablets were homogenized and given mass were<br />

dissolved in water. We took 40 µl of this solution, added 60 µl water and added, into<br />

it Britton-Robinson buffer (900 µl, pH 12) for 60 min at 90 °C. After that, the<br />

52


obtained extract (1 ml) was added to the 9 ml of supporting electrolyte (Britton-<br />

Robinson, pH 2) and analyzed by DPV at CFE.<br />

For checking of quantitative investigation of ASA in urine samples of human the<br />

method of calibration curve was used.<br />

53


RESULTS AND DISCUSSION<br />

Chapter-04<br />

4.1. Simpler Spectrophotometric Assay of PC in Tablets and urine<br />

Samples<br />

Analysts do always try to process the analyte in a way which is suitable for its<br />

optimum analytical signal response. In case of PC determination by UV/visible<br />

spectrometry the standard or sample is hydrolyzed with alkali (Chunli and Baoxin,<br />

1995, Andres, et al., 2003) to convert it to p-aminophenol which is then reacted with a<br />

suitable ligand to get a signal at a specific wavelength. However, such treatment<br />

makes the process not only time consuming but costly as well. In view of the aim of<br />

the current study, the hydrolysis step was overcome by taking directly the photoactive<br />

properties of the PC in aqueous solution. Moreover, the process was made further<br />

economical and safe by avoiding the use of chemicals. Various optimization studies<br />

conducted during this work are presented below.<br />

4.1.1 Effect of Water Addition to PC (FTIR studies)<br />

Fig. 4.1.1. describes the behavior of PC after adding doubly distilled water. It is seen<br />

from the spectra that in case of original PC spectrum (A) the –OH and –NH stretching<br />

is dominant along with absorption by groups like amides. Increase in water addition<br />

(B) to (C) results in domination of –OH stretching over –NH stretching due to<br />

addition of more and more –OH ions from water to PC along with diminishing of<br />

spectra between 1700 to 800 cm −1 region. The spectra obtained at very low<br />

concentration of PC in aqueous solution are almost of the same shape as true for pure<br />

water. However, the FTIR spectra can be calibrated for higher concentration ranges as<br />

quoted previously by Mitsuko et al., 2002 who proposed calibration model for PC<br />

54


concentration at various temperature ranges based on the increment of spectral signals<br />

with increase in concentration of PC in water in the range of 1800–1100 cm −1 . Similar<br />

FTIR studies for PC in water at room temperature have also been conducted in<br />

another citation (Maiella, et al., 1998).<br />

Fig. 4.1.1 FTIR spectra of (A), pure PC (B), aqueous PC paste and (C) aqueous<br />

solution of 5 µgml -1 PC.<br />

In contrast to FTIR studies of water treated PC, the solid-state IR-LD spectral analysis<br />

of monoclinic and orthorhombic PC, polymorphs of aspirin and arginine-containing<br />

peptides have been described by (Ivanova 2005, Koleva, 2006 and Kolev, 2006),<br />

respectively. Another reference is the application of FTIR and Raman spectroscopic<br />

methods for identification and quantification of orthorhombic and monoclinic PC in<br />

powder mixes (Al-Zoubi, et al., 2002).<br />

4.1.2. Optimization of Time for Measurement and Stability of Analytical Signal<br />

Fig. 4.1.2. describes the effect of time on the absorbance value for a 5 µg ml -1 aqueous<br />

solution of PC during one hour duration at room temperature of 30 ± 1 0 C against a<br />

reagent (PC) blank.<br />

55


Absorbance<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0<br />

0 10 20 30 40 50 60<br />

Time (minutes)<br />

Fig. 4.1.2. Dependence of absorbance of PC on time<br />

The effect of time on absorbance of 5 µgml −1 aqueous solution of PC during 1 h<br />

duration at room temperature of 30±1 ◦ C against a reagent (PC) blank was studied. It<br />

was observed that absorbance remained constant throughout the whole period and<br />

thus independent of time. Further observation showed that absorption of the<br />

mentioned PC solution was constant for 72 h while it showed a 2.94% decrease after<br />

96 h. It means that one can analyze PC in standard or sample at his own choice. So the<br />

analysis is very much easy to determine quickly with little energy consumption. Such<br />

constant absorbance vs time has also been demonstrated elsewhere (Jalil and Tsan-<br />

Zon, 2001). As another comparison to current study, a stability time of 10 min has<br />

been reported (Chunli and Baoxin, 1995), who used Fe +3 ions in the presence of S −2<br />

ions for formation of methylene blue-like dye after reaction with p-aminophenol (the<br />

hydrolysis product of PC).<br />

4.1.3. Effect of Temperature<br />

The effect of temperature on absorbance of 5 µgml -1 PC at 243 nm was checked out in<br />

the range of 5–50 ◦ C with 5 ◦ C intervals between two readings. It was seen that the<br />

temperature at lower (5–15 ◦ C) and higher (40–50 ◦ C) range showed some<br />

enhancement of absorbance which corresponds to a 2.94% increase as compared to<br />

medium (20–35 ◦ C) range.<br />

56


Absorbance<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0 10 20 30 40 50<br />

Temperature ( 0 C)<br />

Fig. 4.1.3. Effect of temperature on absorbance of PC<br />

The higher absorbance value at lower temperature may be due to the possibility of<br />

adsorption of PC molecules which could be condensed on the inner wall of quartz<br />

cell. The higher absorbance at higher temperature may be due to the possibility of<br />

evaporation of some water molecules which could make the PC solution concentrated<br />

and hence adding to absorbance value. However, room temperature (30±1 ◦C) was<br />

taken as optimum temperature to avoid the possibility of false analytical signal due to<br />

adsorption at lower or evaporation of water at higher temperature. A temperature<br />

range of 20–60 ◦C has also been described for evaluation of PC by spectrophotometry<br />

(Andres, et al., 2000). They have also chosen room temperature for PC determination<br />

as other temperature values had no effect on its determination.<br />

4.1.4. Effect of Polar Solvents<br />

The effect of addition of 0.01 ml of different solvents on the absorbance of a standard<br />

solution of 5 µgml −1 PC at other optimized conditions is outlined in Table 4.1.1. It is<br />

seen that the addition of aliphatic alcohols in minute quantity has no effect at all.<br />

57


Table 4.1.1. Effect of different polar solvents on absorbance of PC.<br />

Solvent Absorbance Effect (%)<br />

Methanol 0.34 0.00<br />

Ethanol 0.34 0.00<br />

2-propanol 0.34 0.00<br />

N-dimethyl formamide 0.32 -5.88<br />

Acetonitrile 0.32 -5.88<br />

Chloroform 0.35 +2.94<br />

It may be due to increased miscibility and structural similarity of these alcohols with<br />

water. In case of dimethyl formamide and acetonitrile, the negative effect may be due<br />

to greater structural differences of these solvents with water. In case of chloroform,<br />

the little bit higher absorbance value is because of increased concentration of PC in<br />

water due to immiscibility of former in the later. However, the effect of solvent on PC<br />

determination is rarely studied in literature but some authors (Marcelo and Ronei,<br />

2004) have used a 20:80 (v/v) ethanol/water as better solvent mixture for<br />

determination of PC, caffeine and acetyl salicylic acid.<br />

4.1.5. Interference by various Analgesic Drugs<br />

The interference of various analgesic drugs to PC in various proportions was checked<br />

to investigate the possibility of its determination in the presence of other drugs<br />

because some companies use the combination of two or more analgesics for more<br />

efficient response. Table 4.1.2 describes the interference caused by various analgesics<br />

with various ratios present in a solution containing 5 µgml −1 PC. It is evident from the<br />

table that in a 1:1 ratio of drug to PC, there is little or no interference while in a 5:1<br />

ratio, caffeine, diclofenic sodium and ascorbic acid show >±5.0% interference during<br />

PC determination. In a 10:1 ratio the caffeine and ascorbic acid are showing minimum<br />

58


interference as compared to their previous combinations. However, the last<br />

combination is rarely taken in analgesic formulations. So we can say that except<br />

caffeine and ascorbic acid, all other drugs are considered as producing no or little<br />

effect in analysis of PC up to certain possible limits. Similar interference studies have<br />

also been carried out by other workers (Chunli and Baoxin, 1995, Wirat and<br />

Saisunee., 2006). According to later reference, ascorbic acid has been included among<br />

the major interferers while caffeine has been declared as non-interfering one.<br />

Moreover, in the same citation (Wirat and Saisunee., 2006) acetyl salicylic acid has<br />

also been included in the major interferers but in that particular case the conditions<br />

and solution parameters were quite different than ours (Table 4.1.2).<br />

Table 4.1.2. % interference by various analgesics in different ratios on 5 µg ml -1 PC<br />

Ratio to PC<br />

Caffeine Diclofenic<br />

sodium<br />

% interference by drug<br />

Acetyl<br />

salicylic<br />

acid<br />

Ibuprofen Ascorbic<br />

acid<br />

1:1 +2.94 0.00 0.00 -2.94 -2.94<br />

5:1 +8.82 +5.88 0.00 0.00 +11.76<br />

10:1 0.00 +17.64 -14.71 0.00 -2.94<br />

59


Table. 4.1.3. Effect of strong and weak acids and bases on PC determination<br />

Acid / base (1M) Volume of acid<br />

/ base (ml)<br />

% effect pH of PC<br />

solution<br />

λmax<br />

(mid point)<br />

(nm)<br />

Hydrochloric acid 0.1 -2.94 2.13 243<br />

0.5 -2.94 1.72 243<br />

1 -2.94 1.35 243<br />

Acetic acid 0.1 -2.94 3.65 243<br />

0.5 0.00 3.29 243<br />

1 +2.94 3.16 243<br />

NaOH 0.1 +2.94 11.43 257<br />

0.5 0.00 12.28 257<br />

1 +5.88 12.50 257<br />

Ammonia 0.1 0.00 10.40 254<br />

4.1.6. Effect of Acidic and Basic Solutions<br />

0.5 0.00 10.70 255<br />

1 0.00 10.85 256<br />

The effect of adding various volumes of 1Mof various strong and weak acids and<br />

bases on the absorbance and wavelength shift of 5 µgml −1 PC solution was<br />

investigated and the results are given in the following table (Table 4.1.3). It is<br />

observed from the data that the addition of various quantity of 1MNH3 has no effect at<br />

all on the absorbance of PC. HCl and CH3COOH have positive or negative<br />

interference within the acceptable limit of ±5.0%. The only positive effect of +5.88 is<br />

shown after the addition of 1ml of 1M NaOH. It is seen from Fig. 4.1.4 that<br />

dependence of absorbance on concentration of PC is not much affected by variation in<br />

pH in this case and we can say that it is independent of pH. This quality helps in the<br />

analysis of PC in various types of acidic or basic solutions.<br />

60


Fig. 4.1.4. Shift of wavelength with pH: (A) 3.3 and (B) 12. 3 for 5 µgml -1 PC<br />

solution.<br />

4.1.7. Calibration Range<br />

The dependence of various concentrations of PC standard solution on absorbance in a<br />

measurable working range was evaluated at optimized parameters.<br />

Fig. 4.1.5. Calibration range of absorbance vs. concentration for PC from 0.3 to 20<br />

µgml -1 .<br />

61


Fig. 4.1.5 shows linear calibration range for standard PC solutions from 0.3 to 20<br />

µgml −1 . The lower detection limit for this was 0.1 µgml -1 with linear regression<br />

coefficient of 0.9999. The relative standard deviation for a 5 µgml −1 PC (n = 11) was<br />

found out to be 1.6%. The calibration range obtained by this method is better than that<br />

given by Erk et al., 2001, who reported a range of 2–30 µgml −1 for PC determination<br />

by ratio spectra derivative spectrophotometry. The lower detection limit of 0.1 µgml −1<br />

in our case is also closer to that of the later (0.097 µgml −1 ). The detection limit of<br />

current method is better than that reported by Wirat et al., 2006. Who described lower<br />

detection limit of 0.2µgml -1 for PC determination. The linearity of the current<br />

calibration plot is also better than that reported by other workers who described a<br />

linear regression value of 0.9974 for determining monoclinic form of PC (Ivanova,<br />

2005) using IR-LD spectroscopy. The r value of 0.9999 for the current method also<br />

proves the better linearity of our calibration plot in comparison to that described by<br />

Al-Zoubi et al 2002 who obtained the r value of 0.9965 and 0.9954 for eight<br />

calibration points each in case of monoclinic PC by FTIR and FT-Raman<br />

spectroscopy, respectively. Moreover, the lower standard deviation and lower<br />

detection limit values of the current method prove the advantage of its better<br />

repeatability and greater sensitivity over all these solid state methods mentioned<br />

above.<br />

4.1.8. Analysis of Tablets<br />

Table 4.1.4. shows the results of various locally manufactured tablets containing PC<br />

with mentioned and determined concentration of the analyte. Each true result was<br />

obtained after multiplying the actual result with dilution factor 50. The results<br />

obtained by the currently developed method are very close to those reported earlier<br />

(Chunli and Baoxin, 1995), which prove its validity for determination of PC by this<br />

62


method. Moreover, the standard deviation values by the current method are better than<br />

later.<br />

Table 4.1.4. Determination of PC in tables of various companies by proposed method<br />

Chemical<br />

formulation<br />

Mentioned<br />

concentration<br />

(mg unit -1 )<br />

Found concentration a<br />

(mg unit -1 )<br />

By proposed method Reported method<br />

[Chunli and Baoxin, 1995]<br />

Paracetamol 125 121 ± 2.2 121 ± 2.0<br />

Disprol 500 494 ± 2.2 491 ± 3.4<br />

Rascodal 500 497 ± 2.7 499 ± 4.8<br />

Calpol 500 485 ± 3.5 481 ± 3.8<br />

Panadol 500 466 ± 4.2 468 ± 4.4<br />

a , average value; ±, standard deviation (n=5)<br />

Table 4.1.5. Comparative analyses of PC in urine samples.<br />

Paracetamol a<br />

Urine sample By current method By reported method<br />

[Jelena, et al., 2003]<br />

(µg ml -1 ) % age (µg ml -1 ) %age<br />

1 464 ± 13 92.8 459 ±18 91.8<br />

2 475 ± 14 95.0 472 ± 12 94.4<br />

3 434 ± 07 86.6 435 ± 16 87.0<br />

4 427 ± 05 84.5 430 ± 09 86.0<br />

N=11, other abbreviations, as for table 4.<br />

4.1.9. Analysis of Urine Samples<br />

PC determination in four urine samples of four volunteers was conducted by<br />

recording absorbance of 1000 times diluted samples of urine without PC and with PC<br />

63


against double distilled water as blank in each case. The results obtained were<br />

interpreted after adjustment for dilution factor. The data are presented in Table 4.1.5.<br />

For determining PC in urine by comparative method (Jelena, et al., 2003), the dilution<br />

factor of 100 was used for urine sample and the data obtained after proper adjustment.<br />

It is seen from the table that the results obtained with the current method are very<br />

close to those obtained with the reported method. The results show that ≤95%<br />

paracetamol is excreted in urine after 12 h. Similar studies with 95% average<br />

excretion of PC in urine after 12 h have been reported earlier (Jelena, et al., 2003),<br />

which strengthens the confirmation of our results. UV spectra of diluted urine samples<br />

with and without PC are presented in Fig. 4.1.6. An average difference of absorbance<br />


4.2. Simpler and Faster Spectrophotometric Determination of<br />

Diclofenac Sodium (DS) in Tablets, Serum and Urine Samples<br />

The absorption spectra of DS in aqueous medium have been described by several<br />

workers (Matin et al. 2005; Ferreyra and Ortiz 2002). However up to the best of our<br />

knowledge, no attempt has been made so far to utilize the aqueous medium as one of<br />

the most reliable matrices for determining DS with excellent working linear range,<br />

sufficiently low detection limits and its application to many types of samples. So,<br />

various parameters were studied for their effect upon the UV-Visible<br />

spectrophotometric determination of DS.<br />

4.2.1. Influence of Time<br />

The effect of time on the absorbance behavior of 5 µg ml -1 aqueous solution of DS at<br />

276 nm was studied in the range of 1–90 minutes at room temperature of 25 ± 1 0 C<br />

using double distilled water as a blank solution (Fig. 4.2.1). It was observed that the<br />

absorbance remained constant throughout the whole period and thus independent of<br />

time.<br />

A<br />

0.18<br />

0.16<br />

0.14<br />

0.12<br />

0.1<br />

0 10 20 30 40 50 60 70 80 90<br />

Time (min)<br />

Fig. 4.2.1. Time effect on absorbance of Diclofenac Sodium<br />

65


Time effect has been described by other workers (Matin et al. 2005; Sirajuddin et al.<br />

2007) as well. The constant absorbance with time confirms the stability of the analyte<br />

and faster analysis of DS in aqueous solution.<br />

4.2.2. Effect of Temperature<br />

The effect of temperature on absorbance of 5µg ml -1 DS at 276 nm was observed in<br />

the range of 5–60 0 C (Fig. 4.2.2). It was seen that lower temperature range of 5–25 0 C<br />

showed maximum absorbance, with a little bit decreased value for the range of 30–45<br />

0 C was observed followed by a further decrease thereafter.<br />

A<br />

0.16<br />

0.15<br />

0.14<br />

0.13<br />

0.12<br />

5 15 25 35 45 55<br />

Temperature 0 C<br />

Fig. 4.2.2. Temperature effect on UV absorbance of 5 µg ml -1 DS solution.<br />

The possible reason of higher absorbance value at lower temperature may be due to<br />

the adsorption of DS and/ water molecules on the wall of quartz cell and thus<br />

absorbing a little bit higher amount of UV light (Sirajuddin et al. 2007). The lower<br />

absorbance at higher temperature may be due to the instability of DS molecules.<br />

66


However the room temperature (25 ± 1 0 C) was taken as optimum temperature in<br />

order to make the process simple by avoiding additional steps. Temperature studies<br />

have also been reported earlier (Matin et al. 2005) regarding the assay of DS.<br />

4.2.3. Interference by Various Analgesic Drugs<br />

The interference of various analgesic drugs at the absorbance on 5µg ml -1 DS solution<br />

in various proportions was checked to investigate the possibility of its determination<br />

in the presence of other drugs (Table 4.2.1).<br />

Table 4.2.1. % interference by various analgesics in different ratios on 5 µg ml -1 DS<br />

Ratio<br />

to DFS<br />

1:1<br />

5:1<br />

10:1<br />

(%) Interference<br />

Caffeine Paracetamol Aspirin Ibuprofen Ascorbic<br />

acid<br />

+5.47<br />

+49.72<br />

0.00<br />

+39.72<br />

+13.69<br />

-12.32<br />

-18.49<br />

-11.64<br />

+13.01<br />

+ 1.36<br />

-6.16<br />

-10.27<br />

-7.53<br />

-58.90<br />

-62.32<br />

In a 1:1 ratio caffeine and Ibuprofen show little interference while PC and aspirin are<br />

the major positive and negative interferers respectively. The positive interference by<br />

PC may be due to its somewhat structural similarity with or interaction with DS. The<br />

negative value shown by aspirin may be due interaction of central N atom of DS with<br />

the –COO group of aspirin thus hindering its presence to some extent as actual<br />

diclofenac. Other higher values of interference for a 5:1 ratio showing +49.72 %<br />

interference in case of caffeine may be due to indistinguishable wavelength (275 nm)<br />

(Ferreyra and Ortiz 2002) to that of DS (276 nm) which adds up to increase the total<br />

absorbance of DS. No interference at 10:1 ratio of caffeine to DS may be due to rate<br />

67


of interaction of DS with UV light in a first order manner where the higher<br />

concentration of second analyte is usually constant. The highest negative interference<br />

value for 5:1 and 10:1 ratio of ascorbic acid:DS may be due to the interaction of –<br />

COO groups of DS with attached -OH groups of ascorbic acid through H-bonding<br />

which then hinders the actual absorbing activity of DS. As the lower ratio (1:1, 1:2 or<br />

2:1) is usually taken in most of combined drugs, hence we can say that at usual<br />

combination level the mentioned drugs are showing interferences in acceptable<br />

values. However, the presence of PC or ascorbic acid will interfere with the true<br />

analytical value of DS.<br />

This has been observed that the amount of caffeine is quite high in the biological<br />

fluids such as urine, serum or whole blood because various natural foods contain<br />

sufficient amount of caffeine which is ultimately transferred in these fluids. So, no<br />

interference by the highest combination of caffeine and DS in 10:1 ratio is a good<br />

indicator for application of this method in the presence of high amount of caffeine<br />

especially in biological samples.<br />

4.2.4. Effect of Acidic and Alkaline Conditions<br />

Spectrophotometric study of 5 5µg ml -1 DS solution was carried out in various<br />

concentrations of strong and weak acids and bases. UV spectra of a 5 µg ml -1 DS<br />

solution are given (Fig. 4.2.3) to describe the shift in wavelength and absorbance of<br />

DS at extreme conditions of acidity and alkalinity.<br />

68


A<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0.0<br />

200 225 250 275 300 325 350<br />

Fig. 4.2.3. UV spectra of DS at acidic pH, 3.56 (lower) and basic pH, 11.68 (higher).<br />

Moreover, a detailed sketch of such effects upon the addition of different strong and<br />

weak acids and bases was also observed (Table 4.2.2). According to the data, more<br />

negative interference is true at lower pH values of DS solution. However as the pH is<br />

increased, the negative interference is decreased and reaches to acceptable limit. The<br />

value of λmax shifted from 273 nm to 276 nm by entering from acidic into basic<br />

conditions.<br />

nm<br />

69


Table 4.2.2. Effect of strong and weak acids and bases on DFS determination<br />

Acid / base (1M)<br />

Hydrochloric<br />

acid<br />

Acetic acid<br />

Ammonia<br />

NaOH<br />

Volume of acid<br />

/ base (ml)<br />

% effect<br />

pH of DS<br />

solution<br />

λmax<br />

(mid point)<br />

(nm)<br />

0.1 -10.67 2.44 273<br />

0.5 -14.44 2.07 273<br />

1.0 -18.31 1.73 273<br />

0.1 -5.97 3.78 273<br />

0.5 -8.32 3.56 273<br />

1.0 -12.06 3.24 273<br />

0.1 0.00 9.85 276<br />

0.5 -2.23 10.30 276<br />

1.0 -2.98 10.65 276<br />

0.1 -1.0 10.93 276<br />

0.5 -1.86 11.68 276<br />

1.0 -2.23 12.34 276<br />

Lower absorbance value of DS is linked with 2 factors. First is the solubility which<br />

depends upon pH and it is reported that solubility of DS is decreased in acidic<br />

solution. Secondly, DS is subjected to intramolecular cyclization at acidic pH and<br />

hence inactivated (Palomo et al. 1999). In basic conditions a reverse of cyclization<br />

restores the actual molecule along with maximum efficiency. However, a different<br />

situation is presented elsewhere (Matin et al. 2005) where acidic medium (HNO3)<br />

results in the formation of yellowish nitrated derivative of DS which absorbs heavily.<br />

According to our opinion in a protons rich (highly acidic) medium all the Na + ions are<br />

not easy to freed the diclofenac ions available for maximum absorption because the<br />

available protons (H + ions) try to repel the formation of free Na + ions due to similar<br />

charges. So when the number of H + ions decreases, the number of free diclofenac ions<br />

is increased accordingly that results in increased absorption. In case of basic solution,<br />

70


the Na + ions are accepted by the negative OH - ions and hence sufficient diclofenac<br />

ions are available for increased absorption. However the very little negative effect in<br />

basic solution is attributed to base hydrolysis of very limited number of diclofenac<br />

ions into smaller metabolites.<br />

4.2.5. Calibration Plot<br />

UV spectra were recorded at 276 nm for standard solutions of DS with different<br />

concentrations in the range of 0.1–30 µg ml -1 (Fig. 4.2.4.).<br />

A<br />

0.8<br />

0.7<br />

0.6<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0.0<br />

200 225 250 275 300 325 350<br />

Fig. 4.2.4. Calibration plot of absorbance verses concentration for DS solutions from<br />

below to above as 0.1, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25 and 30 µg<br />

ml -1 . Linear equation obtained from calibration plot is represented as; Y=<br />

0.0269X+0.0041 with regression coefficient of 0.9998 and detection limit<br />

of 0.01 µg ml -1 at 276 nm.<br />

A relative standard deviation of 0.33 % was observed for a solution of 5 µg ml -1 DS<br />

(n=11) which describes an excellent reproducibility and repeatability of the method.<br />

4.2.6. Comparison with Other Reported Spectroscopic Methods<br />

A comparison of linear calibration range and detection limits of the current method<br />

with those of some other spectroscopic methods was also described (Table 4.2.3).<br />

nm<br />

71


Table 4.2.3. Comparison of current method with other spectroscopic methods for<br />

determination of DS<br />

Method Reference Linear range LOD<br />

Spectrofluorimetry Damiani et al.,<br />

1999<br />

FI spectroscopy Garcia et al.,<br />

1998<br />

0.2 – 5.0 µg ml -1 0.2 µg ml -1<br />

0.2 – 8 µg ml -1 0.023 µg ml -1<br />

UV-Vis. Spectrometry Matin et al. 2005 1-30 µg ml -1<br />

0.46 µg ml -1<br />

UV-Vis. Spectrometry Botello J.C. and<br />

Perez-Caballero.,<br />

1995<br />

0.8 –6.4 µg ml -1 0.37 µg ml -1<br />

UV-Vis. Spectrometry Mitic et al., 2007 1.59-38.18 µg ml -1 1.29 µg ml -1<br />

UV-Vis. Spectrometry Current method 0.1–30 µg ml -1 0.01 µg ml -1<br />

It is quite clear that despite using various complexing agents, still the working ranges<br />

and detection limits of the reported methods (Matin et al. 2005; Damian et al. 1999;<br />

Garcia et al. 1998; Botello et al. 1995; Mitic et al. 2007) could not compete with that<br />

of the currently developed method. The addition of other reagents for complex<br />

formation makes the other methods time consuming, complicated and expensive. Due<br />

to the lack of the mentioned problems, better sensitivity, broader linear range and<br />

environmental friendly nature, the newly investigated method has a clear edge over<br />

described methods. Moreover, the method is also better than some other reported<br />

methods described in section 1, which possess nearly similar problems as true in case<br />

of reported spectroscopic methods.<br />

4.2.7. Analysis of Tablets<br />

A representative UV spectrum of DS in a randomly selected sample of tablets<br />

(Diclofen) diluted to 5 µg ml -1 DS according to the mentioned concentration is<br />

described (Fig. 4.2.5). The clarity of the signal proves no interference from the matrix.<br />

72


A<br />

0.5<br />

0.4<br />

0.3<br />

0.2<br />

0.1<br />

0.0<br />

200 225 250 275 300 325 350<br />

Fig. 4.2.5. Representative UV-spectrum of expected 5 µg ml -1 DS in (Diclofen) tablet.<br />

The average results of various locally manufactured tablets containing DS with<br />

mentioned and determined concentration for 5 replicate runs were recorded (Table<br />

4.2.4). Each actual result was obtained after multiplying the determined concentration<br />

with dilution factor of 100.<br />

Table 4.2.4. Determination of DS in tablets of various companies by proposed<br />

method<br />

Chemical<br />

formulation<br />

Fenac<br />

Dichloran<br />

Voltral<br />

Ardifenac<br />

Diclofenac<br />

Mentioned<br />

concentration<br />

(mg unit -1 )<br />

50<br />

50<br />

50<br />

50<br />

50<br />

a , average value; ±, standard deviation (n=5)<br />

nm<br />

Actual Concentration a Recovery<br />

(%)<br />

mg unit -1<br />

51.153 ± 0.005<br />

50.646 ± 0.005<br />

49.388 ± 0.008<br />

50.923 ± 0.004<br />

49.837 ± 0.003<br />

102.31± 0.011<br />

101.21 ± 0.011<br />

98.68 ± 0.016<br />

101.85 ± 0.008<br />

99.87 ± 0.006<br />

73


The closeness of mentioned and calculated concentrations of DS in the collected<br />

tablets samples proves the validity of the method and lack of interference from the<br />

excipients.<br />

4.2.8. Analysis of Urine and Serum Samples<br />

10 fold diluted urine or serum sample was processed according to the procedure<br />

mentioned in experimental section. Representative spectra of each of urine and serum<br />

sample are demonstrated respectively (Fig. 4.2.6 and Fig. 4.2.7). For each spectral<br />

observation, the diluted blank and DS containing urine or serum sample of the same<br />

individual was processed.<br />

A<br />

2.0<br />

1.6<br />

1.2<br />

0.8<br />

0.4<br />

0.0<br />

200 225 250 275 300 325 350 375 400<br />

nm<br />

Fig. 4.2.6. UV spectra showing blank (black), DS containing (blue) and DS spiked<br />

(red) urine.<br />

74


A<br />

0.30<br />

0.25<br />

0.20<br />

0.15<br />

0.10<br />

0.05<br />

0.00<br />

200 225 250 275 300 325 350<br />

Fig. 4.2.7. UV spectra showing blank (black), DS containing (blue) and DS spiked<br />

(red) serum.<br />

In case of blank urine sample a hump is seen which reflects the presence of sufficient<br />

amount of caffeine because it has nearly similar λmax (275 nm) Ferreyra and Ortiz,<br />

2002, as DS (276 nm). In all samples, the absorbance of blank was considered as zero<br />

for DS. A clear signal represented by blue line indicates the presence and absorption<br />

spectrum of DS in the urine sample of the individual. The value is however blue<br />

shifted due to the acidic pH and presence of some other ingredients in case of urine<br />

samples. In case of serum sample the lower spectrum shows the presence of caffeine<br />

at lower concentration indicating that very little of it is retained by the serum while<br />

most is removed in urine. The results also show that DS containing serum sample<br />

from the individual administered with 50 mg tablet has a higher absorbance value as<br />

compared to his blank serum sample showing the presence of DS because caffeine is<br />

constant for both samples. The spiking of each diluted urine and serum sample was<br />

nm<br />

75


performed with 40 µl of DS solution in order to confirm the peak signal and hence the<br />

presence of DS in the serum sample.<br />

The results of DS in 4 urine and 4 serum samples by the currently developed method<br />

were observed (Table 4.2.5). Each sample of urine and serum with specific number<br />

(e.g. urine 1 and serum 1) was collected from same individual at different times as<br />

described in experimental section.<br />

Table 4.2.5. Concentration of determined DS and its relation with ingested DS by<br />

oral administration<br />

Sample type DS found (µg ml -1 ) a Actual DS (µg ml -1 ) a DS % of ingested<br />

tablet<br />

Urine 1 3.49 ± 0.010 34.9 ± 0.10 69.8<br />

Urine 2 2.93 ± 0.0120 29.3 ± 0.12 58.6<br />

Urine 3 2.34 ± 0.0160 23.4 ± 0.16 46.8<br />

Urine 4 2.56 ± 0.020 25.6 ± 0.20 51.2<br />

Serum 1 1.14 ± 0.004 11.4 ± 0.04 11.8<br />

Serum 2 1.09 ± 0.004 10.9 ± 0.04 21.8<br />

Serum 3 1.22 ± 0.005 12.2 ± 0.05 24.4<br />

Serum 4 1.15 ± 0.006 11.5 ± 0.06 23.0<br />

a<br />

, average of three replicates<br />

DS in each sample was determined with the help of linear equation and multiplied by<br />

dilution factor 10 in order to get the actual concentration of the ingested DS<br />

transferred to respective urine or serum sample. The data show that the urine and<br />

serum contents of DS have the range of 46.8–62.0 % and 10.09–12.2 % respectively<br />

for a 50 mg ingested tablet. The remaining DS may be present in plasma and/ or<br />

converted to inactive metabolites. As there is no satisfactory data available for DS<br />

contents in these fluids hence we rely upon the currently investigated data. Further<br />

studies in this regard could throw sufficient light upon actual kinetics of this drug and<br />

its fate in the body fluids.<br />

76


4.3. Differential Pulse Voltammetric Determination of PC in Tablet<br />

and Urine Samples at Carbon Film Electrode<br />

4.3.1. Influence of pH on PC at DC and DP Voltammetry.<br />

Firstly, the electrochemical responses of PC on CFE in Britton-Robinson buffer in pH<br />

range from 2-12 were investigated by DC and DP voltammetry. As can be seen in<br />

Figure 4.3.1.<br />

2<br />

I, µA<br />

1<br />

0<br />

A 600<br />

E , mV 1/2<br />

400<br />

10 9<br />

11<br />

8<br />

12<br />

6<br />

7<br />

4<br />

5<br />

200<br />

2 7 12<br />

pH<br />

3 2<br />

200 500<br />

E, mV<br />

800<br />

4<br />

I, µA<br />

2<br />

0<br />

B<br />

9<br />

10<br />

11<br />

12<br />

7<br />

8<br />

6<br />

5<br />

1<br />

4<br />

600<br />

E, mV<br />

400<br />

200<br />

2<br />

3<br />

2 7 pH 12<br />

200 500 800<br />

E, mV<br />

Fig. 4.3.1. DC voltammograms (A) and DP voltammograms (B) of Paracetamol (c =<br />

100 μmol L -1 ) at CFE in Britton-Robinson buffer pH 2 to 12 (numbers<br />

above curves correspond to given pH) without electrode regeneration.<br />

Inset is corresponding dependence of peak potential on the pH.<br />

The influence of different pH from 2-12 in B-R buffer are summarized in (table. 4.3.1)<br />

which shows different peak currents at different peak potentials of paracetamol at DC<br />

and DP voltammetry.<br />

77


Table 4.3.1. The influence of pH on DCV and DPV of Paracetamol, c = 100 μmol L -1<br />

pH<br />

DC Voltammetry DP Voltammetry<br />

Ip, nA Ep, mV Ip, nA Ep, mV<br />

2 817 597 1633 584<br />

3 905 553 1633 541<br />

4 1360 508 2524 464<br />

5 1170 463 2285 410<br />

6 736 419 771 367<br />

7 784 374 7123 317<br />

8 748 329 598 297<br />

9 1141 285 598 238<br />

10 896 240 700 212<br />

11 822 195 889 178<br />

12 344 151 305 159<br />

PC gives one well defined anodic wave / peak which is similar to that observed for an<br />

unmodified glassy carbon electrode Miner et. al, 1981, Van Benschoten et al. 1983,<br />

investigated the electrochemical oxidation of PC using cyclic voltammetry. The first<br />

reaction step is an electrochemical oxidation involving two electrons and two protons<br />

to generate N-acetyl-p-quinoneimine. All subsequent reaction steps are non-<br />

electrochemical, but pH-dependent processes (slope around –50 mV per pH unit or –<br />

43 mV per pH unit for DCV or DPV, respectively). For oxidations at pH values<br />

higher than 6, the final product is a benzoquinone Skeika, et. al. 2008. The best<br />

developed wave and also peak was obtained in BR buffer at pH 4 in aqueous medium.<br />

This medium was further used for measuring of calibration dependences. DPV was<br />

used for further measurements due to its higher sensitivity and easy evaluation.<br />

4.3.2. Optimization of Parameters and Calibration Curve<br />

Repeated measurements revealed passivation of the electrode, probably by products of<br />

the electrode reaction, resulting in decreasing peaks moving toward more negative<br />

potentials. Effect of passivation of the electrode surface was reduced by<br />

electrochemical regeneration of electrode surface with settings of regeneration<br />

78


potentials to values Ereg1 = –400 mV and Ereg2 = 1300 mV (values were optimized by<br />

series of trials) as shown in Fig.4.3.2.<br />

3000<br />

I, nA<br />

1500<br />

3000<br />

I p , nA<br />

1500<br />

0<br />

0 25 N 50<br />

0<br />

200 500 800<br />

E, mV<br />

Fig. 4.3.2. Repetitive measurements of 100 μmol L -1 Paracetamol using DPV at CFE<br />

in Britton-Robinson buffer pH 4.0 with regeneration potential Ereg1 = –400<br />

mV and Ereg2 = 1300 mV Inset is corresponding dependence of peak<br />

current on the number of scans.<br />

Linear calibration curves were obtained using optimal regeneration potentials in the<br />

concentration ranges from 0.02 to100 µmol L -1 . These parameters are summarized in<br />

Table 4.3.2.<br />

Table 4.3.2. Parameters of the calibration straight lines for the determination of<br />

Paracetamol in Britton-Robinson buffer pH 4.0 using DPV at CFE<br />

with regeneration potential Ereg1 = –400 mV, and Ereg2 = 1300 mV.<br />

Concentration<br />

range<br />

[μmol L -1 ]<br />

20 – 100<br />

2 – 10<br />

0.2 – 1<br />

0.02 – 0.1<br />

Slope<br />

[nA.μmol L -1 ]<br />

29.7<br />

48.3<br />

48.7<br />

58.5<br />

Intercept<br />

[nA]<br />

49.5<br />

15.8<br />

1.15<br />

2.28<br />

R<br />

0.9976<br />

0.9981<br />

0.9987<br />

0.9982<br />

LOD<br />

[μmol L -1 ]<br />

–<br />

–<br />

–<br />

0.034<br />

79


The voltammograms connected with the concentration ranges from 20-100 μmol L -1 ,<br />

2-10 μmol L -1 , 0.2-1.0 μmol L -1 and 0.02-0.1 μmol L -1 with correlation coefficients<br />

(R) of 0.9976, 0.9981, 0.9987 and 0.9982 respectively. This shows that the developed<br />

method is very sensitive for salicylic acid determination as depicted in Fig. 4.3.3, Fig.<br />

4.3.4, Fig. 4.3.5. and Fig. 4.3.6. respectively, for illustration.<br />

3000<br />

I, nA<br />

1500<br />

6<br />

5<br />

4<br />

3<br />

2<br />

3000<br />

I , nA p<br />

1500<br />

0<br />

0 50 c, µM 100<br />

1<br />

0<br />

200 500 E, mV 800<br />

Fig. 4.3.3. Differential pulse voltammograms of Paracetamol at CFE in Britton-<br />

Robinson buffer pH 4.0, c(PC): (1) 0, (2) 20 (3) 40, (4) 60, (5) 80, (6) 100<br />

μmol L -1 . Regeneration potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding calibration<br />

dependence.<br />

80


600<br />

I, nA<br />

400<br />

200<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

I , nA p<br />

250<br />

0<br />

0 5 c, µM 10<br />

200 500 800<br />

E, mV<br />

500<br />

Fig. 4.3.4. Differential pulse voltammograms of Paracetamol at CFE in Britton-<br />

Robinson buffer pH 4.0, c(PC): (1) 0, (2) 2 (3) 4, (4) 6, (5) 8, (6) 10 μmol<br />

L -1 . Regeneration potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding calibration<br />

dependence.<br />

200<br />

I, nA<br />

150<br />

6<br />

5<br />

4<br />

3<br />

2<br />

50<br />

I p , nA<br />

0<br />

0.0 0.5<br />

c, µM<br />

1.0<br />

100<br />

200<br />

1<br />

500<br />

E, mV<br />

800<br />

25<br />

Fig. 4.3.5. Differential pulse voltammograms of Paracetamol at CFE in Britton-<br />

Robinson buffer pH 4.0, c(PC): (1) 0, (2) 0,2 (3) 0,4, (4) 0,6, (5) 0,8, (6) 1<br />

μmol L -1 . Regeneration potential Ereg1 = –400 mV and Ereg2 = 1300 mV.<br />

Background current = 1.94 nA. Inset is corresponding calibration<br />

dependence.<br />

81


200<br />

I, nA<br />

150<br />

6<br />

I , nA p<br />

5<br />

5<br />

4<br />

3 2<br />

0<br />

0.00 0.05<br />

c, µM<br />

0.10<br />

100<br />

200 500 800<br />

E, mV<br />

10<br />

1<br />

Fig. 4.3.6. Differential pulse voltammograms of Paracetamol at CFE in Britton-<br />

Robinson buffer pH 4.0, c (PC): (1) 0, (2) 0,02 (3) 0,04, (4) 0,06, (5) 0,08,<br />

(6) 0,1 μmol L -1 . Regeneration potential Ereg1 = –400 mV and Ereg2 = 1300<br />

mV. Background current = 1.94 nA. Inset is corresponding calibration<br />

dependence.<br />

Adsorptive stripping voltammetry was tested for increasing the sensitivity of this<br />

method but no accumulation effect was observed in range accumulation potential<br />

from –300 mV to +300 mV and accumulation time up to 500 s.<br />

4.3.3. Analysis of <strong>Ph</strong>armaceutical Drugs<br />

Commercial pharmaceutical samples (tablets) containing PC were analyzed in order<br />

to evaluate the validity of this proposed method. Recovery experiments were carried<br />

out to evaluate matrix effects after standard-solution additions yielded a good average<br />

recovery (Table 4.3.3).<br />

82


Table 4.3.3. The amount of Paracetamol determined by DPV in tablets of<br />

commercial drugs with declare contents 500 mg of Paracetamol.<br />

Name of tablet Company name<br />

Paralen<br />

Panadol<br />

Paracetamol<br />

Coldrex<br />

Zentiva. Cz<br />

Glaxosmithkline<br />

Glaxosmithkline<br />

Glaxosmithkline<br />

Contents of Paracetamol<br />

[mg]<br />

494<br />

496<br />

503<br />

493<br />

Recovery<br />

[%]<br />

98.8 ± 3<br />

99.1 ± 2<br />

100.7 ± 2<br />

98.5 ± 4<br />

Indicating that there were no important matrix interferences for the samples analyzed<br />

by the proposed DPV method, and had the corresponding results to reference<br />

spectrometric determination electrochemically active ascorbic acid (vitamin C) is<br />

often combined as a medicine with PC based drugs. As documented in Fig. 4.3.7. no<br />

significant interference effect was observed in the developed DPV method up to<br />

tenfold concentration of Ascorbic acid in comparison to concentration of PC.<br />

200<br />

I, nA<br />

100<br />

0<br />

110<br />

90<br />

70<br />

50<br />

30<br />

20<br />

10<br />

0<br />

100<br />

80<br />

60<br />

40<br />

200<br />

I , nA p<br />

100<br />

0<br />

0 50 100<br />

Ascorbic Acid<br />

c, µM<br />

200 500 E, mV 800<br />

Fig. 4.3.7. Differential pulse voltammograms of Paracetamol 10 μmol L -1 with<br />

Ascorbic acid from 10 to 100 μmol L -1 (concentrations are written<br />

above curves in plot) at CFE in Britton-Robinson buffer pH 4.0,<br />

regeneration potential Ereg1 = –400 mV, and Ereg2 = 1300 mV. Inset is<br />

corresponding dependence of peak current of PC on concentration of<br />

Ascorbic acid.<br />

83


4.3.4. Analysis of Urine Samples<br />

For testing the possibility of determination of PC in human urine samples the method<br />

of calibration curve was used. Calibration curves and ranges of determination for<br />

analyzed drug in model solutions of human urine gave linear response in the<br />

concentration ranges of 2 – 10 (Fig. 4.3.8.A) and 20 – 100 mol.l -1 (Fig. 4.3.8.B).<br />

240<br />

I, nA<br />

160<br />

80<br />

A<br />

10<br />

8<br />

6<br />

4<br />

2<br />

1<br />

150<br />

I p , nA<br />

0<br />

0 5 10<br />

c, µM<br />

200 500<br />

E, mV<br />

800<br />

75<br />

1600<br />

I, nA<br />

800<br />

B<br />

20<br />

100<br />

80<br />

60<br />

40<br />

1400<br />

I p , nA<br />

700<br />

0<br />

0 50 100<br />

c, µM<br />

0<br />

200<br />

0<br />

500<br />

E, mV<br />

800<br />

Fig. 4.3.8. Differential pulse voltammograms of 0.1 ml urine with model sample of<br />

Paracetamol (2 – 10 µmol L -1 (A), and 20 – 100 µmol L -1 (B),<br />

concentration is written next to curves in plots) sample at CFE in<br />

Britton-Robinson buffer pH 4.0, regeneration potentials Ereg1 = –400<br />

mV, and Ereg2 = 1300 mV. Inset is corresponding calibration dependence.<br />

The calibration curves show linear response over the whole range of concentration<br />

used in the assay procedure. The parameters associated are summarized in Table<br />

4.3.4. Further parameters of the calibration straight line are seen in Table 4.3.4.<br />

84


Table 4.3.4. Parameters of the calibration straight lines for the determination of<br />

model samples of Paracetamol in urine, media of Britton-Robinson<br />

buffer pH 4.0 using DPV at CFE with regeneration potentials Ereg1 = –<br />

400 mV and Ereg2 = 1300 mV.<br />

Concetration range<br />

[μmol -1 .l]<br />

20 – 100<br />

2 – 10<br />

Slope<br />

[nA.μmol -1 .l]<br />

12.91<br />

14.17<br />

Intercept<br />

[nA]<br />

31.66<br />

–7.61<br />

R LOD<br />

[μ mol -1 .l]<br />

0.9975<br />

0.9994<br />

–<br />

0.48<br />

85


4.4. Quantification of Ibuprofen in <strong>Ph</strong>armaceuticals and Biological<br />

Samples by Transmission FTIR Spectroscopy.<br />

The main object of the current study was to develop a new rapid method by FTIR<br />

spectroscopy which is equally sensitive for standards, pharmaceutical and biological<br />

samples in liquid form containing ibuprofen under similar conditions that can be<br />

frequently used for the routine quality control in pharmaceutical industries and for<br />

quantification in diagnostic laboratories as no method already exists. The existing<br />

methods using high cost instruments which although provide good sensitivity but at<br />

the same time they suffer from the draw backs of time consumption as well as<br />

intensive labor involved. The current method uses the approach to quantify ibuprofen<br />

in pharmaceuticals as well as biological sample such as urine by FTIR Spectroscopy<br />

under optimized parameters. PLS multivariate calibration model was applied here in<br />

order to correlate the measurements collected between different variables on the same<br />

observations in multi-component mixtures as PLS works on the principle of<br />

computing large amount of information which is common between the different<br />

variables with maximal covariance.<br />

4.4.1. Analysis of <strong>Ph</strong>armaceutical Samples<br />

For the quantification of ibuprofen in pharmaceutical samples, calibration was<br />

prepared in the range of 10-100 ppm on FTIR under optimized parameters with very<br />

good linearity R 2 =0.998 as shown in (Fig. 4.4.1) by excellent response of the FTIR<br />

with the increasing concentration.<br />

86


Fig. 4.4.1. Calibration plot in the range of 10-100 ppm for biological samples<br />

The calibration results clearly depict the significant sensitivity and selectivity for the<br />

analyte under examination hence proving the method to be useful. The results<br />

expressed in Table 4.4.1 were highly reproducible with negligible variation as taken<br />

for five replicate measurements.<br />

Table 4.4.1. Results for the ibuprofen concentration found in the tablet samples<br />

Sample<br />

Ibuprofen labeled<br />

mg/Tablet<br />

Ibuprofen found<br />

mg/Tablet<br />

Sample 01 400 412 ± 1.21<br />

Sample 02 400 397 ± 0.71<br />

Sample 03 400 408 ± 1.73<br />

Sample 04 400 403 ± 1.14<br />

Sample 05 400 389 ± 0.85<br />

Sample 06 400 403 ± 1.52<br />

87


There was very good conformity between obtained results and the values labeled on<br />

the formulations, which determines the suitability of the proposed procedure for the<br />

determination of ibuprofen in pharmaceutical sample and are according to the limits<br />

of pharmacopoeia. To check the validity of the newly developed method the known<br />

amounts of ibuprofen was added to the commercial formulation as in Fig. 4.4.2.<br />

Spike 3<br />

Spike 2<br />

Spike 1<br />

Tablet sample<br />

Fig. 4.4.2. <strong>Ph</strong>armaceutical Tablet sample and spikes of 30 ppm, 50 ppm and 70 ppm<br />

Ibuprofen standard.<br />

The recovery was measured by comparing the concentration of pharmaceutical<br />

sample before spiking with that obtained from the spiked samples. After the addition<br />

we observed that the amount of standard added was almost fully recovered with the %<br />

recovery of (98.53, 99.66, 102.02) and were within acceptable limits according to<br />

AOAC guidelines for single laboratory validation of chemical methods for dietary<br />

supplements and botanicals [AOAC, 2002] as given in Table 4.4.2. The coefficient of<br />

variation (CV) values was also small showing that the results were acceptable.<br />

88


Table 4.4.2. Recovery result of ibuprofen from tablet samples after spiking with<br />

known concentrations of standard<br />

(A)<br />

ppm<br />

(B)<br />

ppm<br />

By FT-IR<br />

(C) Recovery a CV b<br />

(%) (%)<br />

1 30 20 49.56. ± 1.25 98.53 1.6<br />

2 50 20 69.83 ± 0.8 99.66 1.3<br />

3 70 20 91.42 ± 1.4 102.02 1.5<br />

(A) Exogenous addition<br />

(B) Before addition<br />

(C) After addition<br />

a Recovery (%)=(C–B)/A x100.<br />

Acceptable recovery (%)<br />

[AOAC, 2002]<br />

90-108<br />

b Coefficient of variation was obtained from the mean of five replicate tests.<br />

4.4.2. Analysis of Urine Samples<br />

For the analysis of ibuprofen in urine samples the same method worked effectively as<br />

the results were precise and reproducible listed in Table 4.4.3. The same calibration<br />

range was used as it also covered the lower range of the analyte specie in urine<br />

sample.<br />

89


Table 4.4.3. Results for the ibuprofen concentration found in urine samples<br />

Sample<br />

Ibuprofen<br />

concentration found<br />

(ppm)<br />

Standard Deviation<br />

Sample 01 11.32 0.81<br />

Sample 02 11.64 0.34<br />

Sample 03 10.75 0.73<br />

Sample 04 11.03 0.24<br />

Sample 05 11.94 0.85<br />

The spectra of extracted urine as blank was collected on FTIR, then a selected urine<br />

sample were spiked with 10 and 20 ppm respectively (these spectra are shown in Fig.<br />

4.4.3.) to check the response and accuracy of the method.<br />

Fig. 4.4.3. Sample urine and 2 spikes of 10 and 20 ppm ibuprofen standard.<br />

90


The results of blank urine sample and spiking are given in Table 4.4.3 which show<br />

very good recovery of the standard added to the sample as recovery is ranging from<br />

98.6 to 100.15 % with very small variation which is clear indication of the reliability<br />

of the method for biological fluid.<br />

Table 4.4.4. Recovery results of ibuprofen in urine samples after spiking with known<br />

concentration of standard<br />

(A) ppm<br />

(B)<br />

ppm<br />

By FT-IR<br />

(C) Recovery a CV b<br />

(%) (%)<br />

1 10 11 20.86 ± 1.3 98.6 2.1<br />

2 20 11 31.03 ± 0.8 100.15 1.4<br />

(A) Exogenous addition<br />

(B) Before addition<br />

(C) After addition<br />

a Recovery (%)=(C–B)/A x100.<br />

b Coefficient of variation was obtained from the mean of five replicate tests.<br />

All the measurements were taken in five replicates to ensure reproducibility and this<br />

can be explained by very low values of standard deviation. Fig. 4.4.5. shows group<br />

spectra of the ibuprofen standards acquired for the calibration to analyze ibuprofen in<br />

urine and pharmaceutical sample analyzed using this method.<br />

91


Fig. 4.4.5. Group Spectra of ibuprofen Standards<br />

From the spectra we can understand that there is very negligible interference from the<br />

other matrix present in the sample while analyzing the analyte of our interest. The<br />

residual mean standard error of calibration (RMSEC) values of 1.06 was achieved<br />

after comparison of actual and computed concentration for all the standards which<br />

explain the accuracy of the method with very good precision.<br />

4.4.3. Limits of Detection and Quantitation<br />

The analysis at the lowest concentration which produced substantial signal was<br />

repeated eleven times and calculated by the following formula:<br />

LOD = 3 × SD × C / M<br />

Where SD is the standard deviation; C is the concentration of analyte and M is the<br />

mean peak area. While LOQ was determined by the same way with following<br />

equation: LOQ = 10 × SD × C / M<br />

The lowest concentration of Ibuprofen to be detected through this method was found<br />

to be 0.77 ppm and quantification limit was found to be 2.54 ppm for IBP<br />

determination.<br />

92


3000<br />

I, nA<br />

1500<br />

4.5. Differential Pulse Voltammetric Determination of Salicylic acid<br />

and Acetylsalicylic acid in Tablet and Urine Samples at Carbon<br />

Film Electrode<br />

4.5.1. Influence of pH on Salicylic Acid in DC and DP Voltammetry<br />

Fig. 4.5.1A describes the influence of pH from 2-12 of BR buffer on Ip salicylic acid<br />

using DC voltammetry. The results showed that at the maximum ions mobility at pH<br />

2 of BR buffer, the peak height is increased for salicylic acid while others are<br />

decreased due to over extent of ions of salicylic acid from pH 3-10 and others pH of<br />

BR buffer did not show peaks of salicylic acid at pH 11-12 due to over the surface of<br />

CFE electrode. So pH 2 of BR buffer was selected for further study. Fig. 4.5.1B<br />

shows the effect of pH of BR buffer from 2-12 using DP voltammetry to compare the<br />

sensitivity of salicylic acid for both techniques.<br />

900<br />

E , mV 1/2<br />

600<br />

2 4 6 8 10 12<br />

pH<br />

12<br />

11<br />

8<br />

4<br />

10<br />

9<br />

5<br />

6<br />

0<br />

200 700 E, mV 1200<br />

3<br />

2<br />

7<br />

3000<br />

I, nA<br />

1500<br />

0<br />

1200<br />

E, mV<br />

600<br />

0<br />

2 4 6 8 10 12<br />

pH<br />

A B<br />

12<br />

200 700<br />

E, mV<br />

1200<br />

4<br />

8<br />

5<br />

10 11<br />

6<br />

9 7<br />

Fig. 4.5.1. A and B. DC & DP voltammograms of Salicylic Acid (c = 100 μmol L -1 )<br />

at CFE in Britton-Robinson buffer pH 2 to 12 (numbers in above curves<br />

correspond to given pH). Inset is corresponding dependence of peak<br />

potential on the pH.<br />

We concluded that the response of salicylic acid with enhanced peak current was at<br />

pH 2 with smooth background current. Due to sensitivity of DP voltammograms, we<br />

selected DP voltammetry for further studies. The influence of different pH from 2-12<br />

2<br />

93<br />

3


in B-R buffer are summarized in (table. 4.5.1) which shows different peak currents at<br />

different peak potentials of Salicylic acid at DC and DP voltammetry.<br />

Table 4.5.1. The influence of pH on DCV and DPV of Salicylic Acid (c = 100 μmol L -1 )<br />

pH<br />

DC Voltammetry DP Voltammetry<br />

Ip, nA Ep, mV Ip, nA Ep, mV<br />

2 2097 1074 2793 1065<br />

3 934 1036 1185 1028<br />

4 1078 993 1944 990<br />

5 820 963. 1060 944<br />

6 713 909. 866 899<br />

7 731 888 746 871<br />

8 1423 875 1302 864<br />

9 838 881 736 817<br />

10 895 825 736 817<br />

11 721 862 372 864<br />

12 168 580 83 521<br />

For the sake of comparison between DC and DP voltammetric determination of<br />

salicylic acid at optimal conditions was made (Supalkova, et al., 2006). All the results<br />

were obtained at pH 2 of BR buffer. The results indicate that better sensitivity can be<br />

obtained at DP voltammetry in comparison to DC voltammetry.<br />

4.5.2. Reproducibility of Salicylic Acid<br />

The peak current for stability 100 µmol L -1 of salicylic acid was studied. Figure 4.5.2<br />

displays voltammogram from series of 45 successive measurements with different<br />

activation potentials at -2000 mV, 1500 mV and 2200 mV. In these three activated<br />

potentials the best stability peak current was seen at activation potential 2200 mV. It<br />

means that activation potential 2200 mV reveals excellent reproducibility of the<br />

salicylic acid at pH 2 of BR buffer at CFE.<br />

94


I, nA<br />

3000<br />

1500<br />

0<br />

0 4 8<br />

Fig. 4.5.2. Repetitive measurements of 100 μmol L -1 Salicylic Acid using DPV at<br />

CFE in Britton- Robinson Buffer pH 2.0 with activation potential (1) –<br />

2000,(2) 1500 and (3) 2200mV<br />

4.5.3. Linear Calibration Curves of Salicylic Acid<br />

Under optimized experimental conditions, calibration plots were performed for<br />

salicylic acid using activation potential 2200 mV at CFE. Both high and low<br />

concentration ranges in Britton-Robinson buffer pH 2 were obtained. The peak<br />

current increased linearly with the increasing concentrations of salicylic acid in the<br />

concentration ranges of 20-100 μmol L -1 , 2-10 μmol L -1 and 0.2-1.0 μmol L -1 with<br />

correlation coefficients ( R) of 0.9993, 0.9994 and 0.9996 respectively . This shows<br />

that the developed method is very sensitive for salicylic acid determination as<br />

depicted in Fig. 4.5.3, Fig. 4.5.4 and Fig. 4.5.5. respectively.<br />

N<br />

1<br />

2<br />

3<br />

95


2400<br />

I, nA<br />

1600<br />

800<br />

1500<br />

I p , nA<br />

750<br />

0<br />

0 50 c, µM 100<br />

750 1000 E, mV 1250<br />

Fig. 4.5.3. Differential pulse voltammograms of Salicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, c(SA): (1) 0, (2) 20 (3) 40, (4) 60, (5) 8,0 (6)<br />

100 μmol L -1 . Activation of potential = 2200 mV for 120 sec. Inset is<br />

corresponding calibration dependence.<br />

750<br />

I, nA<br />

500<br />

250<br />

200<br />

I p , nA<br />

100<br />

0<br />

0 5 c, µM 10<br />

750 1000 E, mV 1250<br />

Fig. 4.5.4. Differential pulse voltammograms of Salicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, c(SA): (1) 0, (2) 2 (3) 4, (4) 6, (5) 8, (6) 10 μmol<br />

L -1 . Activation of potential=2200mV for 120 sec Inset is corresponding<br />

calibration dependence.<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

96


80<br />

I, nA<br />

60<br />

40<br />

20<br />

I , nA p<br />

10<br />

0<br />

0.0 0.5<br />

c, µM<br />

1.0<br />

6<br />

5<br />

4<br />

3<br />

2<br />

800 1000 E, mV 1200<br />

Fig. 4.5.5. Differential pulse voltammograms of Salicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, c (SA): (1) 0, (2) 0.2 (3) 0.4, (4) 0.6, (5) 0.8, (6)<br />

1 μmol L -1 . Activation of potential = 2200 mV for 120 sec. Inset is<br />

corresponding calibration dependence.<br />

The observed splitting of the peak can be connected with different mechanism of the<br />

oxidation of salicylic acid. The limit of detection was obtained 0.21 µM of salicylic<br />

acid. Relatively big intercept complicated the use of the calibration curve but it does<br />

not make it impossible (see table 4.5.2.).<br />

Table 4.5.2. Parameters of the calibration straight lines for the determination of<br />

Salicylic Acid in a Britton-Robinson buffer pH 2.0 using DPV at<br />

CFE with activation potential 2200 mV<br />

Concentration range<br />

[μ mol L -1 ]<br />

20 – 100<br />

2 – 10<br />

0.2-1<br />

Slope<br />

[nA μmol L -1 ]<br />

13.43<br />

16.73<br />

18.52<br />

4.5.4. Hydrolysis of Acetylsalicylic Acid<br />

Intercept<br />

[nA]<br />

35.77<br />

9.58<br />

0.84<br />

1<br />

R<br />

0.9993<br />

0.9994<br />

0.9996<br />

L O D<br />

[μ mol L -1 ]<br />

–<br />

–<br />

0.21<br />

In figure 4.5.6. DP voltammogram shows the hydrolysis of acetylsalicylic acid into<br />

salicylic acid at CFE of pH 2 in B-R buffer. The peak current shows the same peak<br />

97


current of salicylic acid and acetylsalicylic acid (Aspirin) at 2 minutes with activation<br />

potential 2200 mV for 120 sec.<br />

900<br />

I, nA<br />

600<br />

4<br />

3<br />

2<br />

300<br />

750 1000 E, mV 1250<br />

Fig. 4.5.6. Differential pulse voltammograms of 10 μmol L -1 hydrolyzed Acetylsalicylic<br />

Acid at CFE in Britton-Robinson buffer pH 2.0, (1) Electrolyte, (2) 10 μmol<br />

L -1 without hydrolysis (3) 10 μmol L -1 Salicylic Acid (4) 10 μmol L -1 after<br />

hydrolysis Acetylsalicylic acid. Activation potential=2200mV for 120 sec<br />

4.5.5. Linear Calibration Curves of Hydrolyzed Acetylsalicylic Acid at CFE<br />

The calibration dependences of hydrolyzed acetylsalicylic acid for DPV at CFE were<br />

measured in concentration ranges, 20-100 μmol L -1 , 2-10 μmol L -1 and 0.2-1.0 μmol<br />

L -1 depicted in Fig 4.5.7, Fig 4.5.8. and Fig 4.5.9. in Britton-Robinson buffer pH 2<br />

with pulse amplitude 50 mV, pulse width 80 ms and scan rate 20 mV s -1 with<br />

correlation corresponding coefficients (R) of 0.9996, 0.9991 and 0.9996 respectively.<br />

1<br />

98


2000<br />

I, nA<br />

1000<br />

1500<br />

I p , nA<br />

750<br />

0<br />

0 50 c, µM 100<br />

1<br />

0<br />

750 1000 E, mV 1250<br />

Fig. 4.5.7. DP voltammograms of hydrolyzed Acetylsalicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, c(ASA): (1) 0, (2) 20 (3) 40, (4) 60, (5) 80, (6)<br />

100 μmol L -1 . Activation potential = 2200 mV for 120 sec. Inset is<br />

corresponding calibration dependence.<br />

300<br />

I, nA<br />

200<br />

100<br />

200<br />

I p , nA<br />

100<br />

0<br />

0 5 c, µM 10<br />

750 1000 E, mV 1250<br />

Fig. 4.5.8. DP voltammograms of hydrolyzed Acetylsalicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, c (ASA): (1) 0, (2) 2 (3) 4, (4) 6, (5) 8, (6) 10<br />

μmol L -1 . Activation potential = 2200 mV for 120 sec. Inset is<br />

corresponding calibration dependence.<br />

6<br />

5<br />

4<br />

3<br />

2<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

99


120<br />

I, nA<br />

80<br />

20<br />

I , nA p<br />

10<br />

0<br />

0.0 0.5 1.0<br />

c, µM<br />

40<br />

750 1000 E, mV 1250<br />

Fig. 4.5.9. DP voltammograms of hydrolyzed Acetylsalicylic Acid at CFE in Britton-<br />

Robinson buffer pH 2.0, c(ASA): (1) 0, (2) 0.2 (3) 0.4, (4) 0.6, (5) 0.8, (6)<br />

1 μmol L -1 . Activation potential = 2200 mV for 120 sec. Inset is<br />

corresponding calibration dependence.<br />

The limit of detection obtained was 0.15µM for acetylsalicylic acid. This shows that<br />

the developed method is very sensitive for hydrolyzed acetylsalicylic acid. The<br />

optimized parameters obtained for linear calibration curves are summarized in table<br />

4.5.3.<br />

Table 4.5.3. Parameters of the calibration straight lines for the determination of<br />

hydrolyzed Acetylsalicylic Acid in a Britton-Robinson buffer pH 2.0<br />

using DPV at CFE with activation potential 2200 mV<br />

Concentration range<br />

[μmol L -1 ]<br />

20 – 100<br />

2 – 10<br />

0.2-1<br />

Slope<br />

[nA μmol L -1 ]<br />

13.93<br />

18.20<br />

20.17<br />

Intercept<br />

[nA]<br />

17.2<br />

3.11<br />

0.43<br />

6<br />

4<br />

3<br />

2<br />

5<br />

1<br />

R<br />

0.9996<br />

0.9991<br />

0.9996<br />

4.5.6. Determination of Salicylic Acid in Different Drugs<br />

L O D<br />

[μmol L -1 ]<br />

–<br />

–<br />

0.15<br />

The developed method was tested for salicylic acid determination in 4 µM Duofilm<br />

and Saloxyl drugs. The different concentrations of salicylic acid were spiked upto<br />

100


10µM for the determination of salicylic acid at CFE. Good linear calibration curves<br />

were found as given in figure 4.5.10.<br />

240<br />

I, nA<br />

160<br />

80<br />

750 1000<br />

E, mV<br />

1250<br />

Fig. 4.5.10. Differential pulse voltammograms of 4 μmol L -1 Duofilm sample with<br />

spikes of salicylic acid standard at CFE in Britton-Robinson buffer<br />

pH2.0, c (SA): (1) electrolyte, (2) expected 4 μmol L -1 salicylic acid in<br />

sample of Duofilm (3) 6 μmol L -1 spike, (4) 8 μmol L -1 spike and (5) 10<br />

μmol L -1 spike. Activation potential 2200 mV.<br />

The results confirmed the applicability of the method. The parameters of calibration<br />

curves in same medium are thus obtained and summarized in table 4.5.4.<br />

Table 4.5.4. The amount of Salicylic Acid determined by DPV in tablets<br />

S. no Name of Tablet<br />

1<br />

2<br />

Duofilm<br />

Saloxyl<br />

5<br />

4<br />

3<br />

2<br />

Company Name<br />

Stiefel<br />

1<br />

Herbacos-bofarma<br />

Tablets<br />

(%)<br />

16.7<br />

4.5.7. Determination of Acetylsalicylic Acid from Different Drugs<br />

10<br />

Recovery ± stdev<br />

from 5 measur. (%)<br />

103 ± 3<br />

105 ± 1<br />

The developed method was tested for 4 µM Aspirin, Acypyrin, Acypyrin + C,<br />

Disprin, Acefein and Acefein + C drugs. The different concentration of hydrolyzed<br />

101


acetylsalicylic acid were spiked upto 10 µM for the determination of hydrolyzed<br />

acetylsalicylic at CFE. Good linear calibration curves were found as given in figure<br />

4.5.11.<br />

300<br />

I, nA<br />

200<br />

100<br />

750 1000<br />

E, mV<br />

1250<br />

Fig. 4.5.11. Differential pulse voltammograms of 4 μmol L -1 Aspirin sample with<br />

spikes of hydrolyzed acetylsalicylic acid standard at CFE in Britton-<br />

Robinson buffer pH 2.0, c(SA): (1) electrolyte, (2) expected 4 μmol L -1<br />

salicylic acid in sample of Aspirin (3) 6 μmol L -1 spike, (4) 8 μmol L -1<br />

spike and (5) 10 μmol L -1 spike. Activation potential 2200 mV.<br />

The results confirmed the applicability of the method. The parameters of calibration<br />

curves in same medium are summarized in table 4.5.5.<br />

Table 4.5.5. The amount of hydrolyzed Acetylsalicylic Acid determined by DPV in<br />

Tablets<br />

S. no Name of Tablet<br />

1<br />

2<br />

3<br />

4<br />

5<br />

Aspirin<br />

Acylpyrin<br />

Acylpyrin + C<br />

Disprin<br />

Acifein<br />

Company Name<br />

Bayer<br />

Herbacos-bofarma<br />

Herbacos-bofarma<br />

Reckitt Benckiser<br />

Herbacos-bofarma<br />

5<br />

4<br />

3<br />

2<br />

1<br />

Tablets<br />

(mg)<br />

500<br />

500<br />

320<br />

300<br />

250<br />

Recovery ± stdev<br />

from 5 measur. (%)<br />

106 ± 2<br />

106 ± 2<br />

95 ± 1<br />

101 ± 3<br />

100 ± 2<br />

102


4.5.8. Determination of Salicylic Acid in Urine Samples at CFE<br />

The proposed method was also applied to test urine samples for the determination of<br />

salicylic acid at CFE. The linear calibration curves were obtained after the spiked<br />

0.1ml urine samples in ranges from 2 µM to 100 µM concentration of salicylic acid<br />

(see fig. 4.5.12 and fig 4.5.13 respectively).<br />

750<br />

I, nA<br />

500<br />

250<br />

600<br />

I , nA p<br />

400<br />

200<br />

0 50c, µM 100<br />

750 1000 E, mV 1250<br />

Fig. 4.5.12. Differential pulse voltammograms of 0.1 ml urine sample at CFE in<br />

Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2) 20 (3) 40, (4) 60,<br />

(5) 80, (6) 100 μmol L -1 . Activation potential 2200 mV. Inset is<br />

corresponding calibration dependence.<br />

180<br />

I, nA<br />

120<br />

80<br />

I p , nA<br />

40<br />

0<br />

0 5 c, µM 10<br />

60<br />

750 1000<br />

E, mV<br />

1250<br />

Fig. 4.5.13. Differential pulse voltammograms of 0.1 ml urine sample at CFE in<br />

Britton-Robinson buffer pH 2.0, c(SA): (1) 0, (2) 2 (3) 4, (4) 6, (5) 8,<br />

(6) 10 μmol L -1 . Activation potential 2200 mV. Inset is<br />

corresponding calibration dependence.<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

103


These results show the applicability of the method using DP voltammetry at CFE. The<br />

coefficient of regression (R) and limit of detection are obtained from different<br />

calibration curves are summarized in below table 4.5.6.<br />

Table 4.5.6. Parameters of the calibration straight lines for the determination of<br />

Salicylic Acid in 0.1 ml urine samples in a Britton-Robinson buffer<br />

pH 2.0 using DPV at CFE with activation potential 2200 mV<br />

Concentration range<br />

[μ mol -1 L]<br />

20 – 100<br />

2 – 10<br />

Slope<br />

[nA μmol L -1 ]<br />

5.41<br />

8.12<br />

Intercept<br />

[nA]<br />

39.21<br />

–9.03<br />

R<br />

0.9886<br />

0.9976<br />

4.5.9. Determination of Acetylsalicylic Acid in Urine at CFE<br />

L O D<br />

[μ mol -1 L]<br />

–<br />

0.64<br />

Fig. 4.5.14 shows the determination of hydrolyzed acetylsalicylic acid in urine<br />

samples at CFE at pH value of 2 in BR buffer. The concentration of spiked<br />

acetylsalicylic acid in 0.1 ml urine samples was from 20-100µM. The lower<br />

concentration results of acetylsalicylic acid from 2-10µM is given in fig 4.5.15.<br />

104


450<br />

I, nA<br />

300<br />

150<br />

300<br />

I p , nA<br />

150<br />

0<br />

0 50c, µM 100<br />

750 1000<br />

E, mV<br />

1250<br />

Fig. 4.5.14. Differential pulse voltammograms of 0.1 ml urine sample at CFE in<br />

Britton-Robinson buffer pH 2.0, c(ASA): (1) 0, (2) 20 (3) 40, (4) 60,<br />

(5) 80, (6) 100 μmol L -1 . Activation potential 2200 mV.<br />

Inset is corresponding calibration dependence.<br />

90<br />

I, nA<br />

60<br />

30<br />

40<br />

I , nA p<br />

20<br />

0<br />

0 5 c, µM 10<br />

750 1000<br />

E, mV<br />

1250<br />

Fig. 4.5.15. Differential pulse voltammograms of 0.1 ml urine sample at CFE in<br />

Britton-Robinson buffer pH 2.0, c(ASA): (1) 0, (2) 2 (3) 4, (4) 6, (5) 8,<br />

(6) 10 μmol L -1 . Activation potential 2200 mV. Inset is corresponding<br />

calibration dependence.<br />

These results show the applicability of the method using DP voltammetry at CFE. The<br />

coefficient of regression (R) and limit of detection are obtained from different<br />

calibration curves and summarized in table 4.5.7.<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

6<br />

5<br />

4<br />

3<br />

2<br />

1<br />

105


Table 4.5.7. Parameters of the calibration straight lines for the determination of<br />

hydrolyzed Acetylsalicylic Acid in 0.1 ml urine sample in a Britton-<br />

Robinson buffer pH 2.0 using DPV at CFE with activation potential<br />

2200 mV<br />

Concentration range<br />

[μmol L -1 ]<br />

20 – 100<br />

2 – 10<br />

4.5.10. Interference Study<br />

Slope<br />

[nA μmol L -1 ]<br />

3.06<br />

4.51<br />

Intercept<br />

[nA]<br />

17.99<br />

– 3.92<br />

R<br />

0.9997<br />

0.9998<br />

L O D<br />

[μmol L -1 ]<br />

–<br />

0.71<br />

No interference was observed in hydrolyzed acetylsalicylic acid (see fig 4.5.16.) The<br />

concentration from 5 µM to 100 µM ascorbic acid added in 5 µM hydrolyzed<br />

acetylsalicylic acid at CFE was observed. The DP voltammogram of ascorbic acid<br />

show peak potential around 700 mV, while that of acetylsalicylic acid at 1100 to 1200<br />

mV. It means ascorbic acid does not interfere in the presence of hydrolyzed<br />

acetylsalicylic acid.<br />

450<br />

I, nA<br />

300<br />

150<br />

I p , nA<br />

100<br />

50<br />

0<br />

0 50c, µM 100<br />

200 700<br />

E, mV<br />

1200<br />

Fig. 4.5.16. Differential pulse voltammograms of hydrolyzed Acetylsalicylic acid 5<br />

μmol L -1 with ascorbic acid 5-100 μmol L -1 at CFE in Britton-Robinson<br />

buffer pH 2.0. Activation potential 2200 mV.<br />

106


4.6. Conclusions<br />

Various analytical methods were developed for the determination of pharmaceuticals<br />

such as paracetamol, diclofenac sodium, aspirin and brufen based on simplicity,<br />

sensitivity, selectivity, rapidity, low cost as well as environmental safety. For this purpose<br />

voltammetric and spectroscopic techniques were employed. The first proposed method<br />

based on uv-visible spectrophotometric technique for paracetamol determination<br />

possesses many advantages over other analytical methods due to its rapidity, lower cost,<br />

environmental safety and better sensitivity. The method can be successfully employed for<br />

paracetamol quantification in all types of pharmaceutical preparations and liquid samples,<br />

such as urine, serum, gastric juice, etc.<br />

In another approach method was developed for determining diclofenac sodium which is<br />

more superior to reported spectrophotometric methods due to its better sensitivity,<br />

simplicity, stability, economy, environmental safety, broader linear working and lower<br />

detection limits. Application of the developed method for quantification of diclofenac<br />

sodium in tablets with good recovery and lower standard deviation proved its suitability<br />

for analysis of diclofenac sodium in other pharmaceutical preparations. Its successful use<br />

in the determination of diclofenac sodium in urine and serum samples of patients makes<br />

this method as biomarker for identification and hence diagnosis of some diseases<br />

recognized by elevated or decreased level of diclofenac sodium. It may also be concluded<br />

that voltammetric determination of paracetamol using CFE is a suitable for the<br />

micromolar and submicromolar concentrations. The proposed methodology was<br />

successfully applied to the determination of paracetamol in four types of commercial<br />

107


drugs and for model human urine samples and satisfactory results were obtained.<br />

Preparation of the sample was easy and the method is less time-consuming and<br />

economical. Therefore, this quick and simple analytical procedure is suitable for practical<br />

applications. This method may be considered in lot of cases as a suitable alternative to<br />

existing more time-consuming and expensive chromatographic methods.<br />

The main goals achieved through this method include analytical simplicity, remarkable<br />

efficiency, better selectivity and lower cost for the quantification of Ibuprofen. Finally<br />

FTIR spectroscopic method has been developed using KBr windows for the<br />

determination of ibuprofen. A PLS model was successfully developed with the help of<br />

TQ analyst software. The developed method was easy to handle, cheapest and selective<br />

for the quantification of ibuprofen in pharmaceutical drugs and biological fluids i.e. urine<br />

samples.<br />

A suitable voltammetric method was developed using AgA-CFE for the determination of<br />

micromolar concentrations of Acetylsalicylic acid (aspirin) and salicylic acid. The limit<br />

of determination of Aspirin using DPV at AgA-CFE is lower due to efficient<br />

electrochemical activation; the AgA-CFE gives better reproducibility with excellent<br />

signal stability given by less passivation of electrode surface. Furthermore, fairly good<br />

reproducibility of surface renovation has been observed.<br />

108


References<br />

Abu, Q. A. W., Abou, D. M. B. (2001). A validated HPLC method for the determination of<br />

pyridostigmine bromide, acetaminophen, acetylsalicylic acid and caffeine in rat plasma<br />

and urine. J. <strong>Ph</strong>arm. Biomed. Anal. 26(5-6), 939-947.<br />

Afshari, J. T., Liu, T.-Z. (2001). Rapid spectrophotometric method for the quantitation of<br />

acetaminophen in serum. Anal. Chim. Acta 443(1), 165-169.<br />

Agatonovic, K. S., Beresford, R., Razzak, M. (2000). Determination of enantiomeric composition<br />

of ibuprofen in solid state mixtures of the two by DRIFT spectroscopy. Anal. Chim. Acta<br />

417(1), 31-39.<br />

Al-Obaidy, S. S., Li Wan Po, A., McKiernan, P. J., Glasgow, J. F. T., Millership, J. (1995). Assay<br />

of paracetamol and its metabolites in urine, plasma and saliva of children with chroni<br />

liver disease. J. <strong>Ph</strong>arm. Biomed. Anal. 13(8), 1033-1039.<br />

Altair, M. B., Oliveira, H. P. M., Atvars, T. D. Z., Dias, I. L. T., Neto, G. O., Zagatto, E. A. G.,<br />

Kubota, L. T. (2005). Direct determination of paracetamol in powdered pharmaceutical<br />

samples by fluorescence spectroscopy. Anal. Chim. Acta 539(1-2), 257-261.<br />

Al-Zoubi, N., Koundourellis, J. E., Malamataris, S. (2002). FT-IR and Raman spectroscopic<br />

methods for identification and quantitation of orthorhombic and monoclinic paracetamol<br />

in powder mixes. J. <strong>Ph</strong>arm. Biomed. Anal. 29, 459-467.<br />

Amann, G., Gübitz, G., Frei, R. W., Santi, W. (1980). Spectrofluorimetric determination of<br />

pharmaceutical compounds with the cerium(IV)--cerium(III) system. Anal. Chim. Acta<br />

116(1), 119-125.<br />

Ameer, B., Greenblatt, J. D., Divoll, M., Abernethy, R. D., Shargel, L. (1981). High-performance<br />

chromatographic determination of acetaminophen in plasma, single-dose<br />

pharmacokinetic studies. J. Chromatogr. 226, 224–230.<br />

American Academy of Pediatrics Committee on Drugs, (2001). Acetaminophen toxicity in<br />

children. Pediatrics 108, 1020–1024.<br />

AOAC (2002) International guidelines for single laboratory validation of chemical methods for<br />

dietary supplements and botanicals, Association of Official Analytical Chemists,<br />

Arlington, VA.<br />

Arancibia, J. A., Boldrini, M. A., Escandar, G. M. (2000). Spectrofluorimetric determination of<br />

diclofenac in the presence of [alpha]-cyclodextrin. Talanta 52(2), 261-268.<br />

Arancibia, J., Olivieri, A., Escandar, G. (2002). First- and second-order multivariate calibration<br />

applied to biological samples, determination of anti-inflammatories in serum and urine.<br />

Anal. Bioanal.Chem. 374(3), 451-459.<br />

Awtry, E. H., Loscalzo, J. (2000). Aspirin. Circulation 101(10), 1206-1218.<br />

109


Ayora, C. M. J., Pascual, R. M. I., Ruiz, M. A., Fernández-de, C. M. L., Molina, D. A. (2000).<br />

Fast determination of paracetamol by using a very simple photometric flow-through<br />

sensing device. J. <strong>Ph</strong>arm. Biomed. Anal. 22(1), 59-66.<br />

Bahram, B. (2008). Diclofenac sodium 0.1% ophthalmic solution, update on pharmacodynamics,<br />

clinical interest and safety profile. Expt. Rev. Ophth. 3(2), 139-148.<br />

Baranowska, I., Marta, K. (2009). The preliminary studies of electrochemical behavior of<br />

paracetamol and its metabolites on glassy carbon electrode by voltammetric methods.<br />

Electroanalysis 21(10), 1194 – 1199.<br />

Bari, S. B., Kaskhedikar, S. G. (1998). Simultaneous estimation of meclozine HCl and nicotinic<br />

acid from combined dosage form. Ind. J. <strong>Ph</strong>arm. Sci. 60(2), 111-113.<br />

Bauza, R., Ríos, A., Valcárcel, M. (2001). Supercritical fluid extraction with in situ chiral<br />

derivatization for the enantiospecific determination of ibuprofen in urine samples. Anal.<br />

Chim. Acta 450(1-2), 1-11.<br />

Benschoten, J. J. V., Lewis, J. Y., Heineman, W. R., Roston, D. A., Kissinger, P. T. (1983).<br />

Cyclic Voltammetry Experiment. J. Chem. Ed. 60, 772-776.<br />

Bermejo, A. M., Lopez, R. M., Fernandez, P., Cruz, A. (1991). Application of second-derivative<br />

spectroscopy to the simultaneous identification and determination of plasma salicylate<br />

and paracetamol. Anal. Lett. 24, 1147–1157.<br />

Beun, G. D., Leunissen, K. M., Van, B., Vriesman, P. J., Van Hooff, J. P., Grave, W. (1987).<br />

Isolated minimal change nephropathy associated with diclofenac. Br. Med. J. 295(6591),<br />

182-183.<br />

Bidaut, R. M., Gabriel, S. E. (2001). Adverse gastrointestinal effects of NSAIDs, consequences<br />

and costs. Clin. Gastroenterol. 15(5), 739-753.<br />

Blanco, L. M., Lobo, C. M. J., Miranda, O. A., Tuñón B. P. (2003). Voltammetric response of<br />

diclofenac-molecularly imprinted film modified carbon electrodes. Anal. Bioanal.Chem.<br />

377(2), 257-261.<br />

Bojidarka, B. I. (2005). Monoclinic and orthorhombic polymorphs of paracetamol solid state<br />

linear dichroic infrared spectral analysis. J. Mol. Struct. 738, 233-238.<br />

Bojidarka, B. K. (2006), Polymorphs of aspirin solid state IR-LD spectroscopic and quantitative<br />

determination in solid mixtures. J. Mol. Struct. 4, 23-27.<br />

Boopathi, M., Won, M. S., Shim, Y. B. (2004). A sensor for acetaminophen in a blood medium<br />

using a Cu(II)-conducting polymer complex modified electrode. Anal. Chim. Acta 512(2),<br />

191-197.<br />

Bosch, M. E., Sánchez, A. J. R., Rojas, F. S., Ojeda, C. B. (2006). Determination of paracetamol,<br />

Historical evolution. J. <strong>Ph</strong>arm. Biomed. Anal. 42(3), 291-321.<br />

110


Bouhsain, Z., Garrigues, S., Morales-Rubio, A., de la Guardia, M. (1996). Flow injection<br />

spectrophotometric determination of paracetamol in pharmaceuticals by means of on-line<br />

microwave-assisted hydrolysis and reaction with 8-hydroxyquinoline (8-quinolinol).<br />

Anal. Chim. Acta 330(1), 59-69.<br />

Brandt, K. (2003). Paracetamol in the treatment of osteoarthritis pain. Drugs 63(2), 23-41.<br />

British <strong>Ph</strong>armacopoeia (BP, 1980). HMSO, London p. 733.<br />

Bruna, C. L.¸ Roberta A. M., Romeu C., Rocha, F., Luiz, H. M., Orlando F. F. (2009).<br />

Simultaneous voltammetric determination of paracetamol and caffeine in pharmaceutical<br />

formulations using a boron-doped diamond electrode. Talanta 78, 748–752.<br />

Campanella, L., Tomassetti, M., Visco, G. (1990). Chemometric study and analytical enzymatic<br />

methods for diagnosis of cholesterol gallstones. J. <strong>Ph</strong>arm. Biomed. Anal. 8(3), 229-234.<br />

Campanero, M. A., Calahorra, B., GarcIa-Quétglas, E., López-Ocáriz, A., Honorato, J. (1999).<br />

Rapid liquid chromatographic assay for the determination of acetaminophen in plasma<br />

after propacetamol administration, application to pharmacokinetic studies. J. <strong>Ph</strong>arm.<br />

Biomed. Anal. 20(1-2), 327-334.<br />

Carola, F. F., Cristina, S. O. (2002). Simultaneous spectrophotometric determination of<br />

phenilpropanolamine HCL, caffeine and diazepam in tablets. J. <strong>Ph</strong>arm. Biomed. Anal.<br />

29(5),811-818.<br />

Carreira, L. A., Rizk, M., El-Shabrawy, Y., Zakhari, N. A., Toubar, S. S. (1995). Europium(III)<br />

ion probe spectrofluorometric determination of diclofenac sodium. J. <strong>Ph</strong>arm. Biomed.<br />

Anal. 13(11), 1331-1337.<br />

Cashman, J. N. (1996). The Mechanisms of action of NSAIDs in analgesia. Drugs. 52, 13-23.<br />

Catarino, R. I. L., Conceição, A. C. L., Garcia, M. B. Q., Gonçalves, M. L. S., Lima, J. L.<br />

F. C., Correia dos Santos, M. M. (2003). Flow amperometric determination of<br />

pharmaceuticals with on-line electrode surface renewal. J. <strong>Ph</strong>arm. Biomed. Anal. 33(4),<br />

571-580.<br />

Cervini, P., Cavalheiro, Ã. d. T. G. (2008). Determination of paracetamol at a graphitepolyurethane<br />

composite electrode as an amperometric flow detector. J. Braz. Chem. Soc.<br />

19, 836-841.<br />

Chenghang, W. C. L., Fang, C. W. (2006). Covalent Modification of Glassy Carbon Electrode<br />

with L-Cysteine for the Determination of Acetaminophen. Microchim. Acta 155, 365-<br />

371.<br />

Christian, G. D. (2004). Analytical Chemistry, John Wiley Sons, New York, p. 113.<br />

Chu, Q., Jiang, L., Tian, X., Ye, J. (2008). Rapid determination of acetaminophen and paminophenol<br />

in pharmaceutical formulations using miniaturized capillary electrophoresis<br />

with amperometric detection. Anal. Chim. Acta 606(2), 246-251.<br />

111


Clara, D. P., Di, P. R., Perrotta, C., Mazzon, E., Cattaneo, D., Trabucchi, E., Cuzzocrea, S.,<br />

Clementi, E. (2009). Ibuprofen arginine generates nitric oxide and has enhanced antiinflammatory<br />

effects. <strong>Ph</strong>armacol. Res. 60(4), 221-228.<br />

Clements, D., Williams, G. T., Rhodes, J. (1990). Colitis associated with ibuprofen. Biol. Med. J.<br />

301, 987.<br />

Codognoto, L., Machado, S. A. S., Avaca, L. A. (2003). Selective oxidation of pentachlorophenol<br />

on diamond electrodes. J. Appl. Electrochem. 33(10), 951-957.<br />

Colomina, P., Garcia, S. (1989). Agranulocytosis caused by diclofenac. Ann. <strong>Ph</strong>armacother.<br />

23(6), 507.<br />

Committee on Drug (2001). Acetaminophen Toxicity in Children. Pediatrics, 108(4), 1020-1024.<br />

Connors, K. A., Amidon, G. L., Kennon, L. (1979). Chem. Stab. <strong>Ph</strong>arm. Wiley, New York, p.<br />

151.<br />

Costi, E. M., Goryacheva, I., Sicilia, M. D., Rubio, S., Pérez-Bendito, D. (2008). Supramolecular<br />

solid-phase extraction of ibuprofen and naproxen from sewage based on the formation of<br />

mixed supramolecular aggregates prior to their liquid chromatographic/photometric<br />

determination. J. Chromatogr. 1210(1), 1-7.<br />

Criado, A., Cárdenas, S., Gallego, M., Valcárcel, M. (2000). Continuous flow spectrophotometric<br />

determination of paracetamol in pharmaceuticals following continuous microwave<br />

assisted alkaline hydrolysis. Talanta 53(2), 417-423.<br />

Criado, A., Cardenas, S., Gallego, M., Valcarcel, M. (2000). Fast urinary screening for<br />

paracetamol using on-line microwave assisted hydrolysis and spectrophotometric<br />

detection. Analyst 125(6), 1179-1183.<br />

Damiani, P. C., Bearzotti, M., Cabezón, M. A. (2001). Spectrofluorometric determination of<br />

ibuprofen in pharmaceutical formulations. J. <strong>Ph</strong>arm. Biomed. Anal. 25(3-4), 679-683.<br />

Damiani, P. C., Ribone, M. Ã., Olivieri, A. C. (1995). Rapid Determination of Paracetamol in<br />

Blood Serum Samples by First-Derivative UV Absorption Spectroscopy. Anal. Lett.<br />

28(12), 2219 - 2226.<br />

Dargan P. I, Jones A, L. (2003). Management of paracetamol poisoning. Trends <strong>Ph</strong>armacol Sci.<br />

24(4), 154-7.<br />

Dasgupta, A., Kinnaman, G. (1993). Microwave-induced rapid hydrolysis of acetaminophen and<br />

its conjugates in urine for emergency toxicological screen. Clin. Chem. 39(11), 2349-<br />

2350.<br />

Davey, L., Naidoo, D. (1993). Urinary screen for acetaminophen (paracetamol) in the presence of<br />

N- acetylcysteine. Clin. Chem. 39(11), 2348-2349.<br />

De-Carvalho, R. M., Freire, R. S., Rath, S., Kubota, L. T. (2004). Effects of EDTA on signal<br />

stability during electrochemical detection of acetaminophen. J. <strong>Ph</strong>arm. Biomed. Anal.<br />

34(5), 871-878.<br />

112


De-Los, A., Oliva, M., Olsina, R. A., Masi, A. N. (2005). Selective spectrofluorimetric method<br />

for paracetamol determination through coumarinic compound formation. Talanta 66(1),<br />

229-235.<br />

De Oliveira, A. R. M., De Santana, F. J. M., Bonato, P. S. (2005). Stereoselective determination<br />

of the major ibuprofen metabolites in human urine by off-line coupling solid-phase<br />

microextraction and high-performance liquid chromatography. Anal. Chim. Acta 538(1-<br />

2), 25-34.<br />

Dellinger, T. M. Livingston, H. M. (1998). Aspirin burn of the oral cavity. Ann <strong>Ph</strong>armacother.<br />

32(10), 1107.<br />

Dinç, E., Yücesoy, C., Onur, F. (2002). Simultaneous spectrophotometric determination of<br />

mefenamic acid and paracetamol in a pharmaceutical preparation using ratio spectra<br />

derivative spectrophotometry and chemometric methods. J. <strong>Ph</strong>arm. Biomed. Anal. 28(6),<br />

1091-1100.<br />

Dirckx, K., Cabri, P., Merien, A., Galajdova, L., Gerris, J., Dhont, M., De Sutter, P. (2009). Does<br />

low-dose aspirin improve pregnancy rate in IVF/ICSI? A randomized double-blind<br />

placebo controlled trial. Hum. Reprod. 24(4), 856-860.<br />

Dom, Ã. R, A., Medina, R. l. L., P, M. H. (2000). Bioequivalence study of paracetamol tablets, in<br />

vitro-in vivo correlation. Drug Dev. Ind. <strong>Ph</strong>arm. 26(8), 821-828.<br />

Donald, M. R., Aspley, S., Daniels, S. E., Bandy, D. P. (2010). Comparison of the analgesic<br />

efficacy of concurrent ibuprofen and paracetamol with ibuprofen or paracetamol alone in<br />

the management of moderate to severe acute postoperative dental pain in adolescents and<br />

adults, A randomized, double-blind, placebo-controlled, parallel-group, single-dose, twocenter,<br />

modified factorial study. Clin. Ther. 32(5), 882-895.<br />

Dreser, H. (1899). <strong>Ph</strong>armakologisches über aspirin (acetylsalicylsäure). Pfluger’s Arch. 76, 306-<br />

319.<br />

Dubach, U. C., Rosner, B., Starmer, T. (1991). An epidemiologic study of abuse of analgesic<br />

drugs. N. Engl. J. Med. 324(3), 155-160.<br />

Easwaramoorthy, D., Yu, Y.-C., Huang, H.-J. (2001). Chemiluminescence detection of<br />

paracetamol by a luminol-permanganate based reaction. Anal. Chim. Acta 439(1), 95-<br />

100.<br />

Eccles, M., Freemantle, N., Mason, J., the North of England Aspirin Guideline Development, G.<br />

(1998). North of England evidence based guideline development project, guideline on the<br />

use of aspirin as secondary prophylaxis for vascular disease in primary care. Biol. Med. J.<br />

316, 1303-1309.<br />

Edwardson, P. A. D., Nichols, J. D., Sugden, K. (1989). Application of a modified calorimetric<br />

enzyme assay to monitor plasma paracetamol levels following single oral doses to nonpatient<br />

volunteers. J. <strong>Ph</strong>arm. Biomed. Anal. 7(3), 287-293.<br />

113


El Haj, B. M., Al Ainri, A. M., Hassan, M. H., Bin Khadem, R. K., Marzouq, M. S. (1999). The<br />

GC/MS analysis of some commonly used non-steriodal anti-inflammatory drugs<br />

(NSAIDs) in pharmaceutical dosage forms and in urine. Forensic Sci. Int. 105(3), 141-<br />

153.<br />

Elen, R. S., Roberta, A. M., Romeu, C. R., Orlando, F. (2009). Square-wave voltammetric<br />

determination of acetylsalicylic acid in pharmaceutical formulations using a boron-doped<br />

diamond electrode without the need of previous alkaline hydrolysis step. J. Braz. Chem.<br />

Soc. 20, 360-366.<br />

Elwood, P. C. (2001) Aspirin, past, present and future. Clin. Med. 1, 132–37.<br />

Erdal, D., Kökdil, G., Onur, F. (2001). Derivative ratio spectra-zero crossing spectrophotometry<br />

and LC method applied to the quantitative determination of paracetamol,<br />

propyphenazone and caffeine in ternary mixtures. J. <strong>Ph</strong>arm. Biomed. Anal. 26(5-6), 769-<br />

778.<br />

Erica, F. B. F., Sartori, E. R. O., Medeiros, R. A., Rocha-Filho, R. C., Fatibello-Filho, O. (2010).<br />

Differential pulse voltammetric determination of sildenafil citrate (viagraâ) in<br />

pharmaceutical formulations using a boron-doped diamond electrode. Anal. Lett. 43(6),<br />

1046-1054.<br />

Erk, N., Özkan, Y., Banoglu, E., Özkan, S. A., Sentürk, Z. (2001). Simultaneous determination of<br />

paracetamol and methocarbamol in tablets by ratio spectra derivative spectrophotometry<br />

and LC. J. <strong>Ph</strong>arm. Biomed. Anal. 24(3), 469-475.<br />

Evans, D., Hart, J. P., Rees, G. (1991). Voltammetric behaviour of salicylic acid at a glassy<br />

carbon electrode and its determination in serum using liquid chromatography with<br />

amperometric detection. Analyst 116(8), 803-806.<br />

Fang, Y. Z., Long, D., Ye, J. (1997). Study of acetaminophen by parallel incident<br />

spectroelectrochemistry. Anal. Chim. Acta. 342(1), 13-21.<br />

Fanjul, B. P., Lamas, A. P. J., Hernández, S. D., Costa, G. A. (2009). Electrochemical study and<br />

flow injection analysis of paracetamol in pharmaceutical formulations based on screenprinted<br />

electrodes and carbon nanotubes. Anal. Chim. Acta. 638(2), 133-138.<br />

Farquhar, H., Stewart, A., Mitchell, E., Crane, J., Eyers, S., Weatherall, M., Beasley, R.<br />

(2009). The role of paracetamol in the pathogenesis of asthma. Clin. Exp. Allergy 40(1), 32-<br />

41.<br />

Farrar, H., Letzig, L., Gill, M. (2002). Validation of a liquid chromatographic method for the<br />

determination of ibuprofen in human plasma. J. Chromatogr. 780(2), 341-348.<br />

Felix, F. S., Brett, C. M. A., Angnes, L. (2007). Carbon film resistor electrode for amperometric<br />

determination of acetaminophen in pharmaceutical formulations. J. <strong>Ph</strong>arm. Biomed.<br />

Anal. 43(5), 1622-1627.<br />

Fernandez, C. M. L., Ortega, B. P., Molina, D. A. (1998). Sensitive and selective determination of<br />

diclofenac sodium in pharmaceutical preparations by solid phase ultraviolet<br />

absorptiometry. Anal. Chim. Acta 369(3), 263-268.<br />

114


Fernández, L. L., Blanco, L. M. C., Lobo, C. M. J., Miranda O. A. J., Tuñón, B. P. (2007).<br />

Determination of diclofenac in urine samples by molecularly-imprinted solid-phase<br />

extraction and adsorptive differential pulse voltammetry. Electroanalysis 19(15), 1555-<br />

1561.<br />

Fernando, A. H. N., Temple, R. M., Thomas, S., Lee, H. A. (1994). Renal failure after topical use<br />

of NSAIDs. Biol. Med. J, 308, 533.<br />

Fischer J., Vanourkova L., Danhel A., Vyskocil V., Cizek K., Barek J., Peckova K., Yosypchuk<br />

B., Navratil T., (2007). Voltammetric detrmination of nitrophenols at a silver solid<br />

amalgam electrode. int. j. electrochem. Sci. 2, 226.<br />

Franeta, J. T., Agbaba, D., Eric, S., Pavkov, S., Aleksic, M., Vladimirov, S. (2002). HPLC assay<br />

of acetylsalicylic acid, paracetamol, caffeine and phenobarbital in tablets. Il Farmaco<br />

57(9), 709-713.<br />

Freeland, G. R., Northington, R. S., Hedrich D. A., Walker B. R. (1988). Hepatic safety of two<br />

analgesics used over the counter, ibuprofen and aspirin. Clin. <strong>Ph</strong>armacol. Ther. 43, 473-<br />

479.<br />

Fujiwara, M., Chow, P. S., Ma, D. L., Braatz, R. D. (2002). Paracetamol crystallization using<br />

laser backscattering and ATR-FTIR spectroscopy, metastability, agglomeration, and<br />

control. Cryst. Growth Des. 2(5), 363-370.<br />

Fung, Y. S., Luk, S. F. (1989). Determination of salicylic acid in pharmaceutical formulations and<br />

foods by differential-pulse voltammetry using a glassy carbon electrode. Analyst 114(8),<br />

943-945.<br />

Fuster, V.; Dyken, M. L.; Vokonas, P. S.; Hennekens, C. (1993). Aspirin as a therapeutic agent in<br />

cardiovascula disease. Circulation 87, 659-675.<br />

García G. E. B., Ruitenberg, A., Madretsma, G. S., Hintzen, R. Q. (2003). Hyponatraemic coma<br />

induced by desmopressin and ibuprofen in a woman with von Willebrand's disease.<br />

Haemophilia 9(2), 232-234.<br />

García, M. S., Albero, M. I., Sánchez, P. C., Molina, J. (1998). Flow-injection spectrophotometric<br />

determination of diclofenac sodium in pharmaceuticals and urine samples. J. <strong>Ph</strong>arm.<br />

Biomed. Anal. 17(2), 267-273.<br />

Garrido, J. M. P. J.; Lima, J. L. F. C.; Matos, C. D. (2000). Flow injection determination of<br />

acetylsalicylic acid in pharmaceutical preparations with an amperometric detector.<br />

Collect. Czech. Chem. Commun. 65, 954-962.<br />

George, S., Rahi, A. H. (1995). Thrombocytopenia associated with diclofenac therapy. Am. J.<br />

Health Syst. <strong>Ph</strong>arm. 52(4), 420-421.<br />

Gikas, E., Parissi P. M., Kazanis, M., Vavagianis, A. (2003). BrMOZ<strong>Ph</strong>C, A novel coumarin type<br />

reagent for the fluorescent derivatisation of carboxylic acids. Anal. Chim. Acta 489(2),<br />

153-163.<br />

115


Gimenes, D. T., Santos, W. T. P. d., Munoz, R. A. A., Richter, E. M. (2010). Internal standard in<br />

flow injection analysis with amperometric detection. Electrochem. Commun. 12(2), 216-<br />

218.<br />

Glombitza, B. W., Schmidt, P. C. (1994). Comparison of three new spectrophotometric methods<br />

for simultaneous determination of aspirin and salicyclic acid in tablets without separation<br />

of pharmaceutical excipients. J. <strong>Ph</strong>arm. Sci. 83(5), 751-757.<br />

Glówka, F. K., Karazniewicz, M. (2005). High performance capillary electrophoresis method for<br />

determination of ibuprofen enantiomers in human serum and urine. Anal. Chim. Acta<br />

540(1), 95-102.<br />

Goicoechea, A. G., De-Alda, M. J. L. p., Vila-Jato, J. L. (1995). A Validated high-performance<br />

liquid chromatographic method for the determination of paracetamol and its major<br />

metabolites in urine. J. Liq. Chromatogr. 18(16), 3257 - 3268.<br />

Golabi, S. M., Irannejad, L. (2005). Preparation and electrochemical study of fisetin modified<br />

glassy carbon electrode. Application to the determination of NADH and ascorbic acid.<br />

Electroanalysis 17(11), 985-996.<br />

Gonzalez, L., Yuln, G., Volonté, M. G. (1999). Determination of cyanocobalamin,<br />

betamethasone, and diclofenac sodium in pharmaceutical formulations, by high<br />

performance liquid chromatography. J. <strong>Ph</strong>arm. Biomed. Anal. 20(3), 487-492.<br />

Goodman, G. (2007). The pharmacological basis of therapeutics, Ninth ed., McGraw-Hill,<br />

Mexico.<br />

Gorton, L. (1995). Carbon paste electrodes modified with enzymes, tissues, and cells.<br />

Electroanalysis 7(1), 23-45.<br />

Goyal, R. N., Singh, S. P. (2006). Voltammetric determination of paracetamol at C60-modified<br />

glassy carbon electrode. Electrochim. Acta 51(15), 3008-3012.<br />

Goyal, R. N., Chatterjee, S., Agrawal, B. (2010). Electrochemical investigations of diclofenac at<br />

edge plane pyrolytic graphite electrode and its determination in human urine. Sens.<br />

Actuators B 145(2), 743-748.<br />

Goyal, R. N., Gupta, V. K., Oyama, M., Bachheti, N. (2005). Differential pulse voltammetric<br />

determination of paracetamol at nanogold modified indium tin oxide electrode.<br />

Electrochem. Commun. 7(8), 803-807.<br />

Gryfe, C. I., Rubenzahl, S. (1976). Agranulocytosis and aplastic anemia possibly due to<br />

ibuprofen. Can. Med. Assoc. J. 114, 877.<br />

Guidry, J. B., Ogburn, C. L., Griffin, F. M. (1979). Fatal autoimmune hemolytic anemia<br />

associated with ibuprofen. J. Am. Med. Assoc. 242(1), 68-69.<br />

Gupta, S., Tarkkila, P. (1998). Complications of opioids and nonsteroidal antiinflammatories in<br />

chronic nonmalignant pain. Tech. Reg. Anesth. Pain Manage. 2(3), 112-118.<br />

116


Hajjizadeh, M., Jabbari, A., Heli, H., Moosavi M. A. A., Haghgoo, S. (2007). Electrocatalytic<br />

oxidation of some anti-inflammatory drugs on a nickel hydroxide-modified nickel<br />

electrode. Electrochim. Acta 53(4), 1766-1774.<br />

Hammerschmidt, D. E. (1998). About the cover illustration, The first commercial bottle of<br />

aspirin, March 6, 1899. J. Lab. Clin. Med. 132(6), 556.<br />

Hammond, P. M., Scawen, M. D., Atkinson, T., Campbell, R. S., Price, C. P. (1984).<br />

Development of an enzyme-based assay for acetaminophen. Anal. Biochem. 143(1), 152-<br />

157.<br />

Hamoudová, R., Pospísilová, M. (2006). Determination of ibuprofen and flurbiprofen in<br />

pharmaceuticals by capillary zone electrophoresis. J. <strong>Ph</strong>arm. Biomed. Anal. 41(4), 1463-<br />

1467.<br />

Hansen, S. H., Jensen, M. E., Bjørnsdottir, I. (1998). Assay of acetylsalicylic acid and three of its<br />

metabolites in human plasma and urine using non-aqueous capillary electrophoresis with<br />

reversed electroosmotic flow. J. <strong>Ph</strong>arm. Biomed. Anal. 17(6-7), 1155-1160.<br />

Hardman, J. G, Limbird L. E., Goodman, A. G. (1996) Goodman Gilman’s. The <strong>Ph</strong>armacological<br />

Basis of Therapeutics, ninth ed., McGraw-Hill, Mexico, D.F., p. 669.<br />

Hardwick, J. C. H., Van Santen, M., van den Brink, G. R., van Deventer, S. J. H., Peppelenbosch,<br />

M. P. (2004). DNA array analysis of the effects of aspirin on colon cancer cells,<br />

involvement of Rac1. Carcinog. 25(7), 1293-1298.<br />

Hassan, S. M., Walash, M. I., El-Sayed, S. M., Abou A. O. (1981). Colorimetric determination of<br />

certain phenol derivatives in pharmaceutical preparations. J. Assoc. Off. Anal. Chem. 64,<br />

1442–1453.<br />

Hassan, W. S. (2008). Determination of ibuprofen and paracetamol in binary mixture using<br />

cemometric-assisted spectrophotometric methods. Am. J. Appl. Sci. 5(8), 1005-1012.<br />

Hawkey, C. J. (1999) COX-2 inhibitors. The Lancet 353, 307–14.<br />

Hayllar, J., Bjarnason, I. (1995). NSAIDs, Cox-2 inhibitors, and the gut. The Lancet, 346, 521-<br />

522.<br />

Heitmeier, S., Blaschke, G. (1999). Direct determination of paracetamol and its metabolites in<br />

urine and serum by capillary electrophoresis with ultraviolet and mass spectrometric<br />

detection. J. Chromatogr. Biomed. Sci. Appl. 721(1), 93-108.<br />

Heli, H., Jabbari, A., Majdi, S., Mahjoub, M., Moosavi-Movahedi, A., Sheibani, S. (2009).<br />

Electrooxidation and determination of some non-steroidal anti-inflammatory drugs on<br />

nanoparticles of Ni–curcumin-complex-modified electrode. J. Solid State Electrochem.<br />

13(12), 1951-1958.<br />

Hennekens, C. H., Dyken, M. L., Fuster, V. (1997). Aspirin as a therapeutic agent in<br />

cardiovascular disease, a statement for healthcare professionals from the american heart<br />

association. Circulation 96(8), 2751-2753.<br />

117


Hergert, L. A., Escandar, G. M. (2003). Spectrofluorimetric study of the [beta]-cyclodextrinibuprofen<br />

complex and determination of ibuprofen in pharmaceutical preparations and<br />

serum. Talanta 60(2-3), 235-246.<br />

Hindson, B. J., Barnett, N. W. (2001). Analytical applications of acidic potassium permanganate<br />

as a chemiluminescence reagent. Anal. Chim. Acta 445(1), 1-19.<br />

Hoppmann, R. A., Peden, J. G., Ober, S. K. (1991). Central nervous system side effects of<br />

nonsteroidal anti-inflammatory drugs, aseptic meningitis, psychosis, and cognitive<br />

dysfunction. Arch. Int. Med. 151(7), 1309-1313.<br />

Horn, A. C., Jarrett, S. W. (1997). Ibuprofen-induced aseptic meningitis in rheumatoid arthritis.<br />

Ann <strong>Ph</strong>armacother, 31(9), 1009-1011.<br />

Houshmand, M., Jabbari, A., Heli, H., Hajjizadeh, M., Moosavi-Movahedi, A. (2008).<br />

Electrocatalytic oxidation of aspirin and acetaminophen on a cobalt hydroxide<br />

nanoparticles modified glassy carbon electrode. J. Solid State Electrochem. 12(9), 1117-<br />

1128.<br />

Inczedy, J., Lengyel, T., Ure, A. M., (1998) Compendium of Analytical Nomenclature (Definitive<br />

Rules, Blackwell Science, Santa Fe, NM, USA.<br />

Ishida, K., Sakazume, M., Ikegami, H., Doi, K. (1997). Enhanced nephrotoxicity of<br />

acetaminophen in fructose-induced hypertriglyceridemic rats, effect of partial<br />

hepatectomy. Exp. Toxicol. Pathol. 49(5), 321-327.<br />

Ishida, K., Ikegami, H., Doi, K. (1997). Enhanced nephrotoxicity of acetaminophen in fructoseinduced<br />

hypertriglyceridemic rats, contribution of oxidation and deacetylation of<br />

acetaminophen to an enhancement of nephrotoxicity. Exp. Toxicol. Pathol. 49, 313-319.<br />

Issa, Y. M., Zayed, S. I. M., Habib, I. H. I. (2010). Simultaneous determination of ibuprofen and<br />

paracetamol using derivatives of the ratio spectra method. Arabian J. Chem. (Available<br />

online).<br />

Jacoby, H. I. (2000). Gastrointestinal Agents. John Wiley Sons, Inc.<br />

Jafarian, M., Mahjani, M. G., Heli, H., Gobal, F., Heydarpoor, M. (2003). Electrocatalytic<br />

oxidation of methane at nickel hydroxide modified nickel electrode in alkaline solution.<br />

Electrochem. Commun. 5(2), 184-188.<br />

Jain, S. (1994). Ibuprofen-induced thrombocytopenia. Br. J. Clin. Pract. 48, 51.<br />

Jelena, P., Karljiković, R. K., Durić, Z., Jovanović, M., Ibrić, S. (2003). Development of the<br />

second-order derivative UV spectrophotometric method for direct determination of<br />

paracetamol in urine intended for biopharmaceutical characterisation of drug products.<br />

Biopharm. Drug Dispos. 24(7), 309-314.<br />

Jia, L., Zhang, X., Li, Q., Wang, S. (2007). Determination of acetaminophen by square wavevoltammetry<br />

at a gold electrode modified by 4-amino-2-mercaptopyrimidine selfassembled<br />

monolayers. J. Anal. Chem. 62(3), 266-269.<br />

118


Jin, W., Zhang, J. (2000). Determination of diclofenac sodium by capillary zone electrophoresis<br />

with electrochemical detection. J. Chromatogr. 868(1), 101-107.<br />

Jonkman, J. H. G., Schoenmaker, R., Holtkamp, A. H., Hempenius, J. (1985). Determination of<br />

ibuprofen in human plasma by solid phase extraction and reversed-phase highperformance<br />

liquid chromatography. J. <strong>Ph</strong>arm. Biomed. Anal. 3(5), 433-438.<br />

Jouzeau, J. Y., Terlain, B., Abid, A. (1997). Cyclooxygenase isoenzymes, how recent findings<br />

affect thinking about nonsteroidal anti-inflammatory drugs. Drugs 53, 563–582.<br />

Julio, C. B., Guadalupe, P. C. (1995). Spectrophotometric determination of diclofenac sodium<br />

withmethylene-blue. Talanta 42(1), 105-108.<br />

Justiniani, F. R. (1986). Over-the-counter ibuprofen and nephrotic syndrome. Ann. Intern. Med.<br />

105(2), 303.<br />

Kachoosangi, R. T., Wildgoose, G. G., Compton, R. G. (2008). Sensitive adsorptive stripping<br />

voltammetric determination of paracetamol at multiwalled carbon nanotube modified<br />

basal plane pyrolytic graphite electrode. Anal. Chim. Acta 618(1), 54-60.<br />

Kalcher, K., Schach, K., Švancara, I., Vytras, K., Alemu, H. (1998). Recent progresses in the<br />

development of electrochemical carbon paste sensors. Sci. Pap. Univ. Pardubice, Ser. A<br />

3, 41-56.<br />

Kalcher, K., Wang, J., Kauffmann, J. M., Svancara, I., Vytras, K., Neuhold, C., Zhongping, Y.<br />

(1995). Sensors based on carbon paste in electrochemical analysis, a review with<br />

particular emphasis on the period 1990-1993. Ann. Intern. Med. 7, 5-22.<br />

Kalcher, K. (1990). Chemically modified carbon paste electrodes in voltammetric analysis.<br />

Electroanalysis 2(6), 419-433.<br />

Kang, S. H., Chang, S. Y., Do, K.-C., Chi, S.-C., Doo Soo, C. (1998). High-performance liquid<br />

chromatography with a column-switching system and capillary electrophoresis for the<br />

determination of ibuprofen in plasma. J. Chromatogr. B 712(1-2), 153-160.<br />

Kelloff, G. J., Schilsky, R. L., Alberts, D. S., Day, R. W., Guyton, K. Z., Pearce, H. L., Peck, J.<br />

C., <strong>Ph</strong>illips, R., Sigman, C. C. (2004). Colorectal adenomas, A prototype for the use of<br />

surrogate end points in the development of cancer prevention drugs. Colorectal<br />

Adenomas. Clin. Cancer Res. 10(11), 3908-3918.<br />

Khazan, U., Toth, M., Mutgi, A. (1990). Diclofenac sodium and bruising. Ann. Intern. Med. 112,<br />

472–473.<br />

Khoshayand, M. R., Abdollahi, H., Shariatpanahi, M., Saadatfard, A., Mohammadi, A. (2008).<br />

Simultaneous spectrophotometric determination of paracetamol, ibuprofen and caffeine<br />

in pharmaceuticals by chemometric methods. Spectrochim. Acta Part A 70(3), 491-499.<br />

Kibbey, C. E., Park, S. B., DeAdwyler, G., Meyerhoff, M. E. (1992). Further studies on the<br />

potentiometric salicylate response of polymeric membranes doped with tin(IV)tetraphenylporphyrins.<br />

J. Electroanal. Chem. 335(1-2), 135-149.<br />

119


Kim, H. L., Kovacs, M. J. (1995). Diclofenac-associated thrombocytopenia and neutropenia. Ann.<br />

<strong>Ph</strong>armacother. 713–715.<br />

Klimes, J., Sochor, J., Dolezal, P., Körner, J. (2001). HPLC evaluation of diclofenac in<br />

transdermal therapeutic preparations. Int. J. <strong>Ph</strong>armaceutics 217(1-2), 153-160.<br />

Knochen, M., Giglio, J., Reis, B. F. (2003). Flow-injection spectrophotometric determination of<br />

paracetamol in tablets and oral solutions. J. <strong>Ph</strong>arm. Biomed. Anal. 33(2), 191-197.<br />

Kocao, Ä., Ÿlu, S., Karan, A., Berkan, T., BaÅŸdemir, G. (1997). Acute acetaminophen<br />

nephrotoxicity and urinary gamma-glutamyl transferase activity in rats. Drug Metabol.<br />

Drug Interact.14(1), 47-54.<br />

Koleva, T. (2006). Solid-state IR–LD spectroscopic and theoretical analysis of argininecontaining<br />

peptides. Biopolymers 83, 39-45.<br />

Koukli, I. I., Calokerinos, A. C., Hadjiioannou, T. P. (1989). Continuous-flow<br />

chemiluminescence determination of acetaminophen by reduction of cerium(IV). Analyst<br />

114(6), 711-714.<br />

Koupparis, M. A., Anagnostopoulou, P. I. (1988). Automated flow injection determination of<br />

salicylates using Trinder reaction for clinical analysis, assays and dissolution studies of<br />

formulations. J. <strong>Ph</strong>arm. Biomed. Anal. 6(1), 35-46.<br />

Kowal, A., Port, S. N., Nichols, R. J. (1997). Nickel hydroxide electrocatalysts for alcohol<br />

oxidation reactions, An evaluation by infrared spectroscopy and electrochemical<br />

methods. Catal. Today, 38(4), 483-492.<br />

Kubota, L. T., Fernandes, J. C. B., RoverJr, L., Neto, G. d. O. (1999). Determination of<br />

acetylsalicylic acid by FIA-potentiometric system in drugs after on-line hydrolysis.<br />

Talanta 50(3), 661-667.<br />

Kupferwasser, L. I., Yeaman, M. R., Nast, C. C., Kupferwasser, D., Xiong, Y.-Q., Palma, M.,<br />

Cheung, A. L., Bayer, A. S. (2003). Salicylic acid attenuates virulence in endovascular<br />

infections by targeting global regulatory pathways in Staphylococcus aureus. J. Clin.<br />

Invest. 112(2), 222-233.<br />

Lala, L. G., D'Mello, P. M., Naik, S. R. (2002). HPTLC determination of diclofenac sodium from<br />

serum. J. <strong>Ph</strong>arm. Biomed. Anal. 29(3), 539-544.<br />

Lau, G. S. N., Critchley, J. A. J. H. (1994). The estimation of paracetamol and its major<br />

metabolites in both plasma and urine by a single high-performance liquid<br />

chromatography assay. J. <strong>Ph</strong>arm. Biomed. Anal. 12(12), 1563-1572.<br />

Lewis, J. H. Hepatic toxicity of nonsteroidal anti-inflammatory drugs. (1984). Clin. <strong>Ph</strong>arm. 3,<br />

128–38.<br />

Lo, L. Y., Bye, A. (1979). Rapid determination of paracetamol in plasma by reversed-phase highperformance<br />

liquid chromatography. J. Chromatogr. 173(1), 198-201.<br />

120


Lobo, M. J., Miranda, A. J., Tuñón, P. (1997). Amperometric biosensors based on NAD(P)dependent<br />

dehydrogenase enzymes. Electroanalysis 9(3), 191-202.<br />

Loh, H. C., Ahmad, M., Taib, M. N. (2005). A novel salicylic acid optical fibre probe fabrication.<br />

Sens. Actuators B. 107(1), 59-63.<br />

Lohmann, W. Karst U. (2006). Simulation of the detoxification of paracetamol using on-line<br />

electrochemistry/liquid chromatography/mass spectrometry. Anal. Bioanal.Chem. 386,<br />

1701-1708.<br />

López, A., Linares, M., Sánchez, H., Blanquer, A. (1995). Autoimmune hemolytic anemia<br />

induced by diclofenac. Ann <strong>Ph</strong>armacother. 29, 787.<br />

López, C. G., Ostra, M., Ubide, C. (2002). The acetylsalicylic acid-bromine system for<br />

multicomponent kinetic determinations. Anal. Bioanal.Chem. 374(5), 915-922.<br />

Lou, H., Yuan, H., Ruan, Z., Jiang, B. (2010). Simultaneous determination of paracetamol,<br />

pseudoephedrine, dextrophan and chlorpheniramine in human plasma by liquid<br />

chromatography-tandem mass spectrometry. J. Chromatogr. 878(7-8), 682-688.<br />

Love, L. J. C., Zibas, S., Noroski, J., Arunyanart, M. (1985). Direct injection of untreated serum<br />

using nonionic and ionic micellar liquid chromatography for determination of drugs. J.<br />

<strong>Ph</strong>arm. Biomed. Anal. 3(6), 511-521.<br />

Lu, T. L., Tsai, Y. C. (2010). Electrocatalytic oxidation of acetylsalicylic acid at multiwalled<br />

carbon nanotube-alumina-coated silica nanocomposite modified glassy carbon electrodes.<br />

Sens. Acuators B. 148(2), 590-594.<br />

Majdi, S., Jabbari, A., Heli, H. (2007). A study of the electrocatalytic oxidation of aspirin on a<br />

nickel hydroxide-modified nickel electrode. J. Solid State Electrochem. 11, 601-607.<br />

Mamus, S. W., Burton, J. D., Groat, J. D., Schulte, D. A., Lobell, M., Zanjani, E. D. (1986).<br />

Ibuprofen-Associated Pure White-Cell Aplasia. N. Engl. J. Med. 314(10), 624-625.<br />

Manson, J. E., Stampfer, M. J., Colditz, G. A., Willett, W. C., Rosner, B., Speizer, F. E.,<br />

Hennekens, C. H. (1991). A prospective study of aspirin use and primary prevention of<br />

cardiovascular disease in women. J. Am. Med. Assoc. 266(4), 521-527.<br />

Marnett, L. J. (1992). Aspirin and the potential role of prostaglandins in colon cancer. Cancer<br />

Research 52(20), 5575-5589.<br />

Martin, F. L., McLean, A. E. M. (1998). Comparison of paracetamol-induced hepatotoxicity in<br />

the rat in vivo with progression of cell injury in vitro in rat liver slices. Drug and Chem.<br />

Toxicol. 21(4), 477-494.<br />

Martinez, C. J., Gomez, B. C. (1990). Flow-injection spectrofluorimetric determination of<br />

paracetamol. Anal. Chim. Acta 231, 259-264.<br />

121


Martos, N. R., Daaz, A. M., Navalan, A., Capitain, V. L. F. (2001). Spectrofluorimetric<br />

determination of acetylsalicylic acid and codeine mixtures in pharmaceuticals. Anal. Lett.<br />

34(4), 579-595.<br />

Matias, F. A. A., Vila, M. M. D. C., Tubino, M. (2004). Quantitative reflectance spot test for the<br />

determination of acetylsalicylic acid in pharmaceutical preparations. J. Braz. Chem. Soc.<br />

15, 327-330.<br />

Matin, A. A., Farajzadeh, M. A., Jouyban, A. (2005). A simple spectrophotometric method for<br />

determination of sodium diclofenac in pharmaceutical formulations. Il Farmaco 60(10),<br />

855-858.<br />

Mattok, G. L., McGilveray, I. J., Mainville, C. A. (1971). Acetaminophen III, Dissolution studies<br />

of commercial tablets of acetaminophen and comparison with in vivo absorption<br />

parameters. J. <strong>Ph</strong>arm. Sci. 60(4), 561-564.<br />

Mazurek, S., Szostak, R. (2006). Quantitative determination of diclofenac sodium and<br />

aminophylline in injection solutions by FT-Raman spectroscopy. J. <strong>Ph</strong>arm. Biomed.<br />

Anal. 40(5), 1235-1242.<br />

Mazurek, S., Szostak, R. (2008). Quantitative determination of diclofenac sodium in solid dosage<br />

forms by FT-Raman spectroscopy. J. <strong>Ph</strong>arm. Biomed. Anal. 48(3), 814-821.<br />

McGregor, A. H., More, L. J., Simpson, K. J., Harrison, D. J. (2003). Liver death and<br />

regeneration in paracetamol toxicity. Hum. Exp. Toxicol. 22(4), 221-227.<br />

Merckle, P., Kovar, K. A. (1998). Assay of effervescent tablets by near-infrared spectroscopy in<br />

transmittance and reflectance mode, acetylsalicylic acid in mono and combination<br />

formulations. J. <strong>Ph</strong>arm. Biomed. Anal. 17(3), 365-374.<br />

Miles, F. K., Kamath, R., Dorney, S. F., Gaskin, K. J., O'Loughlin, E. V. (1999). Accidental<br />

paracetamol overdosing and fulminant hepatic failure in children. Med. J. Aust. 1. 171(9),<br />

472-475.<br />

Miner, D. J., Rice, J. R., Riggin, R. M., Kissinger, P. T. (1981). Voltammetry of acetaminophen<br />

[paracetamol] and its metabolites. Anal. Chem. 53(14), 2258-2263.<br />

Moore, T. J., Joseph, M. J., Allen, B. W., Coury, L. A. (1995). Enzymically amplified<br />

voltammetric sensor for microliter sample volumes of salicylate. Ana.l Chem. 67(11),<br />

1896-1902.<br />

Moreira, A. B., Dias, I. L. T., Neto, G. O., Zagatto, E. A. G., Kubota, L. T. (2004). Solid-phase<br />

fluorescence spectroscopy for the determination of acetylsalicylic acid in powdered<br />

pharmaceutical samples. Anal. Chim. Acta. 523(1), 49-52.<br />

Moreira, A. B., Oliveira, H. P. M., Atvars, T. D. Z., Dias, I. L. T., Neto, G. O., Zagatto, E. A. G.,<br />

Kubota, L. T. (2005). Direct determination of paracetamol in powdered pharmaceutical<br />

samples by fluorescence spectroscopy. Anal. Chim. Acta. 539(1-2), 257-261.<br />

122


Mugford, C. A., Tarloff, J. B. (1997). The contribution of oxidation and deacetylation to<br />

acetaminophen nephrotoxicity in female Sprague-Dawley rats. Toxicol. Letters 93(1), 15-<br />

22.<br />

Muralidharan, B., Gopu, G., Vedhi, C., Manisankar, P. (2008). Voltammetric determination of<br />

analgesics using a montmorillonite modified electrode. App. Clay Science 42(1-2), 206-<br />

213.<br />

Murfin, J. W., Wragg, J. S. (1972). A colorimetric method for the determination of phenacetin<br />

and paracetamol. Part II. A manual procedure for the determination of phenacetin or<br />

paracetamol in formulations. Analyst 97(1157), 670-675.<br />

Murillo, J. A., Garcia, L. F. (1996). Application of first derivative fluorescence spectrometry to<br />

the simultaneous determination of paracetamol and salicylamide in pharmaceuticals.<br />

Anal. Lett., 29(3), 423-438.<br />

Nada, F. A., Maher, F. E. (2009). Poly (3-methylthiophene)/palladium sub-micro-modified sensor<br />

electrode. Part II, Voltammetric and EIS studies, and analysis of catecholamine<br />

neurotransmitters, ascorbic acid and acetaminophen. Talanta 79. 639–647.<br />

Nagasawa, H. T., Shoeman, D. W., Cohen, J. F., Rathbun, W. B. (1996). Protection against<br />

acetaminophen-induced hepatotoxicity by L-CySSME and its N-acetyl and ethyl ester<br />

derivatives. J. Biochem. Toxicol. 11(6), 289-295.<br />

Nakamura, H., Tamura, Z. (1980). Fluorimetric determination of secondary amines based on their<br />

reaction with fluorescamine. Anal. Chem. 52(13), 2087-2092.<br />

Nanda, A., Ola, M., Bhaskar, R., Sharma, C. K., Nayak, S. (2010). Formulation and evaluation of<br />

an effervescent, gastroretentive drug-delivery system. (Peer reviewed) <strong>Ph</strong>armaceut.<br />

Technol.10(34) 60-71.<br />

Nikles, C. J., Yelland, M., Del Mar, C., Wilkinson, D. (2005). The role of paracetamol in chronic<br />

pain, an evidence-based approach. American J. Therapeutics 12(1), 80-91.<br />

Nogowska, M., Muszalska, I., Zajac, M. (1999). Simultaneous spectrophotometric determination<br />

of acetylsalicylic acid, paracetamol and caffeine in pharmaceutical preparations. Chem<br />

Anal. 44, 1041.<br />

Osteryoung, J. G., Osteryoung, R. A. (1985). Square wave voltammetry. Anal. Chem .57(1),<br />

101A-110A.<br />

Palabiyik, I. M., Dinç, E., Onur, F. (2004). Simultaneous spectrophotometric determination of<br />

pseudoephedrine hydrochloride and ibuprofen in a pharmaceutical preparation using ratio<br />

spectra derivative spectrophotometry and multivariate calibration techniques. J. <strong>Ph</strong>arm.<br />

Biomed. Anal. 34(3), 473-483.<br />

Palomo, M. E., Ballesteros, M. P., Frutos, P. (1999). Analysis of diclofenac sodium and<br />

derivatives. J. <strong>Ph</strong>arm. Biomed. Anal. 21 (1) 83-94.<br />

Parojčić, J., Karljiković R. K., Durić, Z., Jovanović, M., Ibrić, S. (2003). Development of the<br />

second-order derivative UV spectrophotometric method for direct determination of<br />

123


paracetamol in urine intended for biopharmaceutical characterization of drug products.<br />

Biopharm. Drug Dispos. 24(7), 309-314.<br />

Parviz, N., Faezeh, D., Mohammad, R. G., Parandis, D. (2009). sprosium Nanowire Modified<br />

Carbon Paste Electrode for the Simultaneous Determination of Naproxen and<br />

Paracetamol, Application in <strong>Ph</strong>armaceutical Formulation and Biological Fluid. Int. J.<br />

Electrochem. Sci. 4, 373 – 1386.<br />

Pasekova, H., Sales, M. G., Montenegro, M. C., Araújo, A. N., Polásek, M. (2001).<br />

Potentiometric determination of acetylsalicylic acid by sequential injection analysis (SIA)<br />

using a tubular salicylate-selective electrode. J. <strong>Ph</strong>arm. Biomed. Anal. 24(5-6), 1027-<br />

1036.<br />

Patel, F. The fatal paracetamol dosage-how low can you go? (1992), Med Sci Law 32,303–310.<br />

Patricia, C., Damiani, Mariela B., Miguel, A., Cabezón, Alejandro C. O. (1999).<br />

Spectrofluorometric determination of diclofenac in tablets and ointments. J. <strong>Ph</strong>arm.<br />

Biomed.Anal.203,587-590.<br />

Patrono, C. (1994). Aspirin as an antiplatelet drug. New Engl. J. Med. 330, 1287-1294.<br />

Payán, M. R., López, M. Á. B., Fernández-Torres, R., Bernal, J. L. P., Mochón, M. C. (2009).<br />

HPLC determination of ibuprofen, diclofenac and salicylic acid using hollow fiber-based<br />

liquid phase microextraction (HF-LPME). Anal. Chim. Acta. 653(2), 184-190.<br />

Pedrosa, V. A., Lowinsohn, D., Bertotti, M. (2006). FIA Determination of Paracetamol in<br />

<strong>Ph</strong>armaceutical Drugs by Using Gold Electrodes Modified with a 3-Mercaptopropionic<br />

Acid Monolayer. Electroanalysis 18(9), 931-934.<br />

Pedrosa, V. D. A., Codognoto, L. C., Avaca, L. A. (2003). Determinação voltamétrica de<br />

4-clorofenol sobre o eletrodo de diamante dopado com boro utilizando a voltametria de<br />

onda quadrada. QuÃmica Nova, 26, 844-849.<br />

Pegon, Y., Vallon, J. J. (1981). Extraction du paracetamol par relargage de solvant et application<br />

au dosage par chromatographie en phase gazeuse dans le plasma , enA rapid extraction<br />

technique for the determination of paracetamol in plasma by gas chromatography. Anal.<br />

Chim. Acta. 130(2), 405-408.<br />

Pereira, A. V., Aniceto, C., Fatibello, F. O. (1998). Flow injection spectrophotometric<br />

determination of acetylsalicylic acid in tablets after on-line microwave-assisted alkaline<br />

hydrolysis. Analyst 123, 1011–1015.<br />

Pérez, R. T., MartInez, L. C., Sanz, A., San Miguel, M. T. (1997). Flow extraction<br />

spectrophotometric method for the determination of diclofenac sodium in pharmaceutical<br />

preparations. J. <strong>Ph</strong>arm. Biomed. Anal. 16(2), 249-254.<br />

Perneger, T. V., Whelton, P. K., Klag, M. J. (1994). Risk of kidney failure associated with the use<br />

of acetaminophen, aspirin, and nonsteroidal antiinflammatory drugs. N. Engl. J. Med.<br />

331(25), 1675-1679.<br />

124


Pimenta, A. M., Araújo, A. N., Montenegro, M. C. B. S. M. (2002). Simultaneous potentiometric<br />

and fluorimetric determination of diclofenac in a sequential injection analysis system.<br />

Anal. Chim. Acta. 470(2), 185-194.<br />

Pirker, R., Huck, C. W., Popp, M., Bonn, G. K. (2004). Simultaneous determination of gentisic,<br />

salicyluric and salicylic acid in human plasma using solid-phase extraction, liquid<br />

chromatography and electrospray ionization mass spectrometry. J. Chromatogr. 809(2),<br />

257-264.<br />

Piro, B., Do, V.-A., Le, L. A., Hedayatullah, M., <strong>Ph</strong>am, M. C. (2000). Electrosynthesis of a new<br />

enzyme-modified electrode for the amperometric detection of glucose. J. Electroanal.<br />

Chem. 486(2), 133-140.<br />

Pirola, R., Bareggi, S. R., De Benedittis, G. (1998). Determination of acetylsalicylic acid and<br />

salicylic acid in skin and plasma by high-performance liquid chromatography. J.<br />

Chromatogr. B 705(2), 309-315.<br />

Pournaghi, M. H., Sabzi, R. E. (2004). Electrocatalytic oxidation of sulfite on a cobalt<br />

pentacyanonitrosylferrate film modified glassy carbon electrode. Electroanalysis 16(10),<br />

860-865.<br />

Prasek, J., Hubalek, J., Adamek, M., Jasek, O., Zajickova, L. (2007). Nanopatterned working<br />

electrode with carbon nanotubes improving electrochemical sensors. J. Nanoeng.<br />

Nanosyst. 221(3), 115-119.<br />

Prescott, L. F. (2001). Paracetamol overdose. In Paracetamol (Acetaminophen) – A Critical<br />

Bibliographic Review. 2nd Edition 527 – 624. London, Taylor Francis Ltd.<br />

Price, A. J., Obeid, D. (1989). Spontaneous non-gastrointestinal bleeding associated with<br />

diclofenac. Lancet. 2, 23-30.<br />

Pulgarín, J. A. M., Bermejo, L. F. G. (1996). Flow-injection stopped-flow spectrofluorimetric<br />

kinetic determination of paracetamol based on its oxidation reaction by<br />

hexacyanoferrate(III). Anal. Chim. Acta 333(1-2), 59-69.<br />

Purcell, P., Henry, D., Melville, G. (1991). Diclofenac hepatitis. Gut, 32(11), 1381-1385.<br />

Quintino, M. S. M., Corbo, D., Bertotti, M., Angnes, L. (2002). Amperometric determination of<br />

acetylsalicylic acid in drugs by batch injection analysis at a copper electrode in alkaline<br />

solutions. Talanta 58(5), 943-949.<br />

Quintino, M. S. M., Angnes, L. (2004). Bia-amperometric quantification of salbutamol in<br />

pharmaceutical products. Talanta 62(2), 231-236.<br />

Ragno, G., Ioele, G., Risoli, A. (2004). Multivariate calibration techniques applied to the<br />

spectrophotometric analysis of one-to-four component systems. Anal. Chim. Acta 512(1),<br />

173-180.<br />

Ravi, S., Keat, A. C., Keat, E. C. (1986). Colitis caused by non-steroidal anti-inflammatory drugs.<br />

Postgrad. Med. J. 62(730), 773-776.<br />

125


Ray, J. E., Stove, J., Williams, K. M. (1987). A rapid urinary screen for acetaminophen modified<br />

to avoid false- negative results. Clin Chem, 33(5), 718.<br />

Richardson, C., Emery, P. (1996). The clinical implications of inhibition of the inducible form of<br />

cyclo-oxygenase. Drug Safety 15, 249–60.<br />

Riddhi, G., Rajashree, M., Pankaj, S. (2010). Development and validation of spectrophotometric<br />

methods for simultaneous estimation of ibuprofen and paracetamol in soft gelatin capsule<br />

by simultaneous equation method. Inter. J. Chem.Tech. Res. 2, 1881-1885.<br />

Schootstra, R., Yska, J. P. (2008). Compounding medication for digestive decontamination,<br />

pharmaceutical aspects. Selective Digestive Tract Decontamination in Intensive Care<br />

Medicine, a Practical Guide to Controlling Infection (73-87), Springer Milan.<br />

Robards, K., Worsfold, P. J. (1992). Analytical applications of liquid-phase chemiluminescence.<br />

Anal. Chim. Acta 266(2), 147-173.<br />

Rodriguez, J. A., Barrado, E., Castrillejo, Y., Santos, J. R., Lima, J. L. F. C. (2007). Validation of<br />

a tubular bismuth film amperometric detector, Determination of diclofenac sodium by<br />

multisyringe flow injection analysis. J. <strong>Ph</strong>arm. Biomed. Anal. 45(1), 47-53.<br />

Rover, J. L., De-oliveira, N. G., Roberto, F. J., Tatsuo, K. L. (2000). Determination of salicylate<br />

in blood serum using an amperometric biosensor based on salicylate hydroxylase<br />

immobilized in a polypyrrole-glutaraldehyde matrix. Talanta 51(3), 547-557.<br />

Rover, L., Garcia, C. A. B., Neto, G. d. O., Kubota, L. T., Galembeck, F. (1998). Acetylsalicylic<br />

acid determination in pharmaceutical samples by FIA-potentiometry using a salicylatesensitive<br />

tubular electrode with an ethylene-vinyl acetate membrane. Anal. Chim. Acta.<br />

366(1-3), 103-109.<br />

Ruengsitagoon, W., Liawruangrath, S., Townshend, A. (2006). Flow injection<br />

chemiluminescence determination of paracetamol. Talanta 69(4), 976-983.<br />

Ruiz, M. A., Fernández-de, C. M. L., Ortega, B. P., Molina, D. A. (2001). Flow-through UV<br />

spectrophotometric sensor for determination of (acetyl)salicylic acid in pharmaceutical<br />

preparations. Int. J. <strong>Ph</strong>armaceutics 216(1-2), 95-104.<br />

Russeau, W., Mitchell, J., Tetteh, J., Lane, M. E., Hadgraft, J. (2009). Investigation of the<br />

permeation of model formulations and a commercial ibuprofen formulation in Carbosil®<br />

and human skin using ATR-FTIR and multivariate spectral analysis. Int. J. <strong>Ph</strong>arm.<br />

374(1-2), 17-25.<br />

Ryley, N. G, (1989). Diclofenac associated hepatitis. Gut. 30, A708.<br />

Sádecká, J., Cakrt, M., Hercegová, A., Polonský, J., Skacáni, I. (2001). Determination of<br />

ibuprofen and naproxen in tablets. J. <strong>Ph</strong>arm. Biomed. Anal. 25(5-6), 881-891.<br />

Sadik, O. A., Wallace, G. G. (1993). Pulse damperometric detection of proteins using antibody<br />

containing conducting polymers. Anal. Chim. Acta 279(2), 209-212.<br />

126


Safavi, A., Maleki, N., Moradlou, O. (2008). A selective and sensitive method for simultaneous<br />

determination of traces of paracetamol and p-aminophenol in pharmaceuticals using<br />

carbon ionic liquid electrode. Electroanalysis 20(19), 2158-2162.<br />

Sandulescu, R., Mirel, S., Oprean, R. (2000). The development of spectrophotometric and<br />

electroanalytical methods for ascorbic acid and acetaminophen and their applications in<br />

the analysis of effervescent dosage forms. J. <strong>Ph</strong>arm. Biomed. Anal. 23(1), 77-87.<br />

Santhosh Nair, S., John, S. A., Sagara, T. (2009). Simultaneous determination of paracetamol and<br />

ascorbic acid using tetraoctylammonium bromide capped gold nanoparticles immobilized<br />

on 1,6-hexanedithiol modified Au electrode. Electrochim. Acta 54(27), 6837-6843.<br />

Santini, O., J. E. de Oliveira, H. R., Pezza, L. (2006). A new potentiometric ibuprofenate ion<br />

sensor immobilized in a graphite matrix for determination of ibuprofen in tablets,<br />

Microchem. J. 84, 44–49.<br />

Santos, W. D. R., P. R. Lima, C. R. Tarley, L. T. Kubota, (2007). A catalytically active<br />

molecularly imprinted polymer that mimics peroxidase based on hemin, application to<br />

the determination of p –aminophenol. Anal. and Bioanal. Chem. 389, 1919.<br />

Santos, W. T. P., Dos, E. G. N., de Almeida, H. E. A., Ferreira, D. T., Gimenes, E. M. R. (2008).<br />

Simultaneous flow injection analysis of paracetamol and ascorbic acid with multiple<br />

pulse amperometric detection. Electroanalysis 20, 1878-1883.<br />

Saraswathyamma, B., Grzybowska, I., Orlewska, C., Radecki, J., Dehaen, W., Kumar, K. G.,<br />

Radecka, H. (2008). Electroactive dipyrromethene-Cu(II) monolayers deposited onto<br />

gold electrodes for voltammetric determination of paracetamol. Electroanalysis 20(21),<br />

2317-2323.<br />

Šatínský, D., Neto, I., Solich, P., Sklenářová, H., Conceição, M., Montenegro, B. S. M., Araújo,<br />

A. N. (2004). Sequential injection chromatographic determination of paracetamol,<br />

caffeine, and acetylsalicylic acid in pharmaceutical tablets. J. Separation Sci. 27(7-8),<br />

529-536.<br />

Save, T. K., Parmar, D. V., Devarajan, P. V. (1997). High-performance thin-layer<br />

chromatographic determination of ibuprofen in plasma. J. Chromatogr. B. 690(1-2), 315-<br />

319.<br />

Schirmer, R.E., (1991). Modern Methods of <strong>Ph</strong>armaceutical Analysis, vol. I, Second ed., CRS<br />

Press, New York, 213.<br />

Seitz, W., Knox, R., Van, D. C. (1981). Chemiluminescence and bioluminescence analysis,<br />

fundamentals and biomedical applications. Rev. Anal. Chem. 13,1-58.<br />

Sena, M. M., Poppi, R. J. (2004). N-way PLS applied to simultaneous spectrophotometric<br />

determination of acetylsalicylic acid, paracetamol and caffeine. J. <strong>Ph</strong>arm. Biomed. Anal.<br />

34(1), 27-34.<br />

Sena, M. M., Fernandes, J. C. B., Rover, L., Poppi, R. J., Kubota, L. T. (2000). Application of<br />

two- and three-way chemometric methods in the study of acetylsalicylic acid and<br />

127


ascorbic acid mixtures using ultraviolet spectrophotometry. Anal. Chim. Acta 409(1-2),<br />

159-170.<br />

Shah, M., Balaraman, R. (1999). Effect of phenobarbitone, carbamazepine and sodium valproate<br />

on the elimination kinetics of acetaminophen. Ind. J. <strong>Ph</strong>armacol. 31(4), 311-314.<br />

Shaheen, S. O., Newson, R. B., Sherriff, A., Henderson, A. J., Heron, J. E., Burney, P. G. J.,<br />

Golding, J., the Alspac Study, T. (2002). Paracetamol use in pregnancy and wheezing in<br />

early childhood. Thorax 57(11), 958-963.<br />

Shahrokhian, S. Asadian, E. (2010) . Simultaneous voltammetric determination of ascorbic acid,<br />

acetaminophen and isoniazid using thionine immobilized multi-walled carbon nanotube<br />

modified carbon paste electrode. Electrochim. Acta 55, 666–672.<br />

Shen, S. C., Ko, C. H., Hsu, K. C., Chen, Y. C. (2004). 3-OH flavone inhibition of epidermal<br />

growth factor-induced proliferaton through blocking prostaglandin E2 production. Int. J.<br />

Cancer 108(4), 502-510.<br />

Sirajuddin, Khaskheli, A. R., Shah, A., Bhanger, M. I., Niaz, A., Mahesar, S. (2007). Simpler<br />

spectrophotometric assay of paracetamol in tablets and urine samples. Spectrochim. Acta,<br />

Part A. 68(3), 747-751.<br />

Skeika, T., Faria, M. F. D., Nagata, N., Pessoa, C. A. (2008). Simultaneous voltammetric<br />

determination of dypirone and paracetamol with carbon paste electrode and multivariate<br />

calibration methodology. J. Braz. Chem. Soc. 19, 762-768.<br />

Sneader, W. (2000). The discovery of aspirin, a reappraisal. Biol. Med. J, 321(7276), 1591-1594.<br />

Snežana, M., Gordana, M., Aleksandra, P., Snežana, T., Emilija, P. (2007). Determination of<br />

diclofenac sodium in commercial pharmaceutical formulations and human control serum<br />

using a kinetic-spectrophotometric method. Chem. <strong>Ph</strong>arm. Bull. 55, 1423-1426.<br />

Sochor, J., Klimes, J., Sedlácek, J., Zahradnícek, M. (1995). Determination of ibuprofen in<br />

erythrocytes and plasma by high performance liquid chromatography. J. <strong>Ph</strong>arm. Biomed.<br />

Anal. 13(7), 899-903.<br />

Sotiropoulus, J. B., Deutsch, T., Plakogiannis, F. M. (1981). Comparative bioavailability of three<br />

commercial acetaminophen tablets. J. <strong>Ph</strong>arm. Sci. 70(4), 422-425.<br />

Souza, D. d., Machado, S. A. S., Avaca, L. A. (2003). Voltametria de onda quadrada. Primeira<br />

parte, aspectos teóricos. QuÃmica Nova, 26, 81-89.<br />

Stolker, A. M., Niesing, W., Fuchs, R., Vreeken, R., Niessen, W. A., Brinkman, U. T. (2004).<br />

Liquid chromatography with triple-quadrupole and quadrupole-time-of-flight mass<br />

spectrometry for the determination of micro-constituents, A comparison. Anal.<br />

Bioanal.Chem. 378(7), 1754-1761.<br />

Stone, E. (1763). An account of the success of the bark of the willow tree in the cure of agues.<br />

<strong>Ph</strong>ilos. Trans. R. Soc. Lond., 53, 195-200.<br />

128


Su, W.-Y., Cheng, S.-H. (2010). Electrochemical oxidation and sensitive determination of<br />

acetaminophen in pharmaceuticals at poly(3,4-ethylenedioxythiophene)-modified screenprinted<br />

electrodes. Electroanalysis, 22(6), 707-714.<br />

Sun, S. F., Lui, G. R., Wang, Y. H. (2006). Simultaneous determination of acetaminophen,<br />

caffeine, and chlorphenamine maleate in paracetamol and chlorphenamine maleate<br />

granules. Chromatographia 64(11-12), 719-724.<br />

Sun, Y., Takaba, K., Kido, H., Nakashima, M. N., Nakashima, K. (2003). Simultaneous<br />

determination of arylpropionic acidic non-steroidal anti-inflammatory drugs in<br />

pharmaceutical formulations and human plasma by HPLC with UV detection. J. <strong>Ph</strong>arm.<br />

Biomed. Anal. 30(5), 1611-1619.<br />

Supalkova, V., Petrek, J., Havel, L., Krizkova, S., Petrlova, J., Adam, V., Potesil, D., Babula, P.,<br />

Beklova, M., Horna, A., Kizek, R. (2006). Electrochemical sensors for detection of<br />

acetylsalicylic acid. Sensors 6(11), 1483-1497.<br />

Suzanne W. S., Wong, H. Y. (1998). Standards of laboratory practice, analgesic drug monitoring.<br />

Clin. Chem. 44(5), 1110–1123.<br />

Tapan, K. G., Dulal, K. T. (2010). <strong>Ph</strong>ysicochemical classification and formulation development<br />

of solid dispersion of poorly water soluble drugs, an updated review. J. <strong>Ph</strong>arm. Biol.<br />

Arch. 1(2), 189 – 192.<br />

Tattersall, J., Greenwood, R., Farrington, K. (1992). Membranous nephropathy associated with<br />

diclofenac. Postgrad. Med. J. 68, 392–3.<br />

Thun, M. J., Henley, S. J., Patrono, C. (2002). Nonsteroidal Anti-inflammatory Drugs as<br />

Anticancer Agents, Mechanistic, <strong>Ph</strong>armacologic, and Clinical Issues. J. Natl. Cancer<br />

Inst. 94(4), 252-266.<br />

Torriero, A. A. J., Luco, J. M., Sereno, L., Raba, J. (2004). Voltammetric determination of<br />

salicylic acid in pharmaceuticals formulations of acetylsalicylic acid. Talanta 62(2), 247-<br />

254.<br />

Townshend, A., Alan Wheatley, R. (1998). Oxidative chemiluminescence of some nitrogen<br />

nucleophiles in the presence of formic acid as an ancillary reductant. Analyst, 123(2),<br />

267-272.<br />

Trinder, P. (1954). Rapid determination of salicylate in biological fluids. Biochem. J. 57(2), 301-<br />

300.<br />

Tsikas, D., Tewes, K. S., Gutzki, F.-M., Schwedhelm, E., Greipel, J., Frölich, J. C. (1998). Gas<br />

chromatographic-tandem mass spectrometric determination of acetylsalicylic acid in<br />

human plasma after oral administration of low-dose aspirin and guaimesal. J.<br />

Chromatogr. 709(1), 79-88.<br />

Tubino, M., de Souza, R. L. (2006). Determination of diclofenac in pharmaceutical preparations<br />

by diffuse reflectance photometry. Talanta 68(3), 776-780.<br />

129


Utzinger, U., Richards-Kortum, R. R. (2003). Fiber optic probes for biomedical optical<br />

spectroscopy. J. Biomed. Opt. 8(1), 121-147.<br />

Valcárcel, M., Gallego, M., Ríos, A. (1998). Coupling continuous flow systems to instruments<br />

based on discrete sample introduction. Fresenius. J. Anal. Chem. 362(1), 58-66.<br />

Valderrama, P., Poppi, R. J. (2010) Second order standard addition method and fluorescence<br />

spectroscopy in the quantification of ibuprofen enantiomers in biological fluids.<br />

Chemometr. Intell. (Available online).<br />

Vane J. R, Flower R. J, Botting R.M. (1990). History of aspirin and its mechanism of action.<br />

Stroke. 21(12 Suppl), 12-23.<br />

Velasco, D., Danoux, C. B., Redondo, J. A., Elvira, C., San Román, J., Wray, P. S., Kazarian, S.<br />

G. (2011). pH-sensitive polymer hydrogels derived from morpholine to prevent the<br />

crystallization of ibuprofen. J. Controlled Release. 149(2), 140-145.<br />

Vidal, A. D. n., Reyes, J. F. G. a., Barrales, P. O., DÃaz, A. M. (2002). Uv Spectrophotometric<br />

flow-through multiparameter sensor for the simultaneous determination of<br />

acetaminophen, acetylsalicylic acid, and caffeine. Anal. Lett. 35(15), 2433 - 2447.<br />

Vila-Jato L.J., J. Blanco, M.J. Alonso, J. <strong>Ph</strong>arm. <strong>Ph</strong>armacol. (1986) The effect of the molecular<br />

weight of polyethylene glycol on the bioavailability of paracetamol-polyethylene glycol<br />

solid dispersions. 38, 126-128.<br />

Vilchez, J., Blanc, R., Avidad, R., Navalón, A. (1995). Spectrofluorimetric determination of<br />

paracetamol in pharmaceuticals and biological fluids. J. <strong>Ph</strong>arm. Biomed. Anal. 13(9),<br />

1119-1125.<br />

Vire, J. C., Kauffmann, J. M. (1994). Trends in electrochemistry in drug analysis. Curr. Top.<br />

Electrochem. 3, 493-515.<br />

Walily El, A. F. M., Abdel-Kader G. A., Belal, S. F., Khamis, E. F. (1999). Selective<br />

spectrofluorimetric determination of phenolic [beta]-lactam antibiotics through the<br />

formation of their coumarin derivatives. J. <strong>Ph</strong>arm. Biomed. Anal. 20(4), 643-653.<br />

Wang, B., Liu, G., Dou, Y., Liang, L., Zhang, H., Ren, Y. (2009). Quantitative analysis of<br />

diclofenac sodium powder via near-infrared spectroscopy combined with artificial neural<br />

network. J. <strong>Ph</strong>arm. Biomed. Anal. 50(2), 158-163.<br />

Wang, S. F., Xie, F., Hu, R.-F. (2007). Carbon-coated nickel magnetic nanoparticles modified<br />

electrodes as a sensor for determination of acetaminophen. Sens. Actuators B. 123(1),<br />

495-500.<br />

Wang, W. H., Wong, W. M., Dailidiene, D., Berg, D. E., Gu, Q., Lai, K. C., Lam, S. K., Wong,<br />

B. C. Y. (2003). Aspirin inhibits the growth of helicobacter pylori and enhances its<br />

susceptibility to antimicrobial agents. Gut 52(4), 490-495.<br />

Wangfuengkanagul, N., Chailapakul, O. (2002). Electrochemical analysis of acetaminophen<br />

using a boron-doped diamond thin film electrode applied to flow injection system. J.<br />

<strong>Ph</strong>arm. Biomed. Anal. 28(5), 841-847.<br />

130


Welch, R. M., Conney, A. H. (1965). A simple method for the quantitative determination of Nacetyl-p-aminophenol<br />

(APAP) in urine. Clin Chem, 11(12), 1064-1067.<br />

Whelton, A., Stout, R. L., Spilman, P. S., Klassen, D. K. (1990). Renal effects of ibuprofen,<br />

piroxicam, and sulindac in patients with asymptomatic renal failure. Ann. Internal Med.<br />

112(8), 568-576.<br />

Whittle, B. J. (2003). Gastrointestinal effects of nonsteroidal anti-inflammatory drugs. Fundam.<br />

clin. <strong>Ph</strong>armacol. 17(3), 301-313.<br />

Wong T. L, Solomonraj G, Thomas H. B. (1976). High-pressure liquid chromatographic<br />

determination of acetaminophen in biological fluids. J. <strong>Ph</strong>arm. Sci. 65, 1064–1066.<br />

Wong, S., Appleberg, M., Ward, C. M., Lewis, D. R. (2004). Aspirin resistance in cardiovascular<br />

disease, A Review. Off. J. Eur. Soc. Vasc. Surg. 27(5), 456-465.<br />

Wyszecka, K. E., Warowna, G. M., Fijalek, Z. (2003). Determination of 4-aminophenol<br />

impurities in multicomponent analgesic preparations by HPLC with amperometric<br />

detection. J. <strong>Ph</strong>arm. Biomed. Anal. 32(4-5), 1081-1086.<br />

Xianwen, K., Zhirong, G., Yao, Z., Zhilin, W., Jun, J. Z. (2009) Magnetic molecularly imprinted<br />

polymer for aspirin recognition and controlled release. Nanotechnology 20, 1- 7.<br />

Xu, C., Li, B. (2004). Spectrophotometric determination of paracetamol with microwave assisted<br />

alkaline hydrolysis. Spectrochim. Acta Part A. 60(8-9), 1861-1864.<br />

Xu, M., Chen, L., Song, J. (2004). Polarographic behaviors of diclofenac sodium in the presence<br />

of dissolved oxygen and its analytical application. Anal. Biochem. 329(1), 21-27.<br />

Xu, Z., Yue, Q., Zhuang, Z., Xiao, D. (2009). Flow injection amperometric determination of<br />

acetaminophen at a gold nanoparticle modified carbon paste electrode. Microchim. Acta<br />

164(3), 387-393.<br />

Yang, X., Wang, F., Hu, S. (2008). Enhanced oxidation of diclofenac sodium at a nano-structured<br />

electrochemical sensing film constructed by multi-wall carbon nanotubes-surfactant<br />

composite. Mater. Sci. Engineer. C 28(1), 188-194.<br />

Yilmaz, B. (2010) Simultaneous determination of estradiol valerate and medroxyprogesterone<br />

acetate in a tablet formulation by gas chromatography and mass spectrometry. Anal. Sci.<br />

26(3), 391-393.<br />

Yinnon, A. M., Moreb, J. S., Slotki, I. N. (1987). Drug points, Nephrotic syndrome associated<br />

with diclofenac sodium. Brit. Med. J. 295(6597), 556.<br />

Yousef, E. M., Heli, H., Bathaie, S. Z., Mousavi, M. F. (2007). Electrocatalytic oxidation of<br />

glucose at a Ni-curcumin modified glassy carbon electrode. J. Solid State Electrochem.<br />

11(2), 273-282.<br />

131


Zen, J. M., Ting, Y. S. (1997). Simultaneous determination of caffeine and acetaminophen in<br />

drug formulations by square-wave voltammetry using a chemically modified electrode.<br />

Anal. Chim. Acta 342(2-3), 175-180.<br />

Zhao, S., Bai, W., Yuan, H., Xiao, D. (2006). Detection of paracetamol by capillary<br />

electrophoresis with chemiluminescence detection. Anal. Chim. Acta 559(2), 195-199.<br />

132


List of Publications<br />

1. Simpler Spectrophotometric Assay of Paracetamol in Tablets and Urine Samples. Sirajuddin,<br />

Abdul Rauf Khaskheli, Afzal Shah, Muhammad Iqbal Bhanger, Abdul Niaz and Sarfaraz<br />

Mahesar; Spectrochimica Acta Part A, 2007, 68, 747-751.<br />

2. Simpler and faster spectrophotometric determination of diclofenac sodium in tablets, serum<br />

and urine samples. Abdul Rauf Khaskheli, Sirajuddin, Kamran Abro, S.T.H Sherazi, H.I<br />

Afridi, S.A. Mahesar, Munawar Saeed; Pakistan Journal of Environmental and Analytical<br />

Chemistry, 2009, 10, 53-58.<br />

3. Highly sensitive spectrometric method for determination of hydroquinone in skin lightening<br />

creams: application in cosmetics. S. Uddin, A. Rauf, T.G. Kazi, H. I. Afridi and G.<br />

Lutfullah; International Journal of Cosmetic Science, 2010, 1- 8.<br />

4. Differntial pulse voltammetric determination of paracetamol in tablet and urine samples at a<br />

carbon film electrode. Abdul Rauf Khaskheli, Jan Fischer, Jiří Barek,Vlastimil Vyskočil,<br />

Sirajuddin and Muhammad Iqbal Bhanger. (Submitted in Electroanalysis).<br />

5. Evaluation of ibuprofen in tablet formulation and urine samples by transmission FTIR<br />

spectroscopy in conjunction with partial least squares (PLS). A. R. Khaskheli, S. T. H.<br />

Sherazi, Sirajuddin, S. A. Mahesar, M. Ali, Nazar Kalwar and Aftab Kandhro. (Submitted in<br />

Talanta).<br />

6. Differntial pulse voltammetric determination of salicylic acid and acetyl salicylic acid in<br />

tablet and urine samples at carbon film electrode. Abdul Rauf Khaskheli, Jan Fischer, Jiří<br />

Barek,Vlastimil Vyskočil, Sirajuddin, Muhammad Iqbal Bhanger and Munawar Saeed.<br />

(Submitted in Electroanalysis).<br />

Other Publications<br />

1. Ultra-trace level determination of hydroquinone in waste photographic solutions by UV–Vis<br />

spectrophotometry. Sirajuddin , M. Iqbal Bhanger, Abdul Niaz, Afzal Shah and Abdul<br />

Rauf; Talanta, 2007.<br />

2. Abdul Niaz, Sirajuddin, Afzal Shah, S. A. Mahesar, Abdul Rauf. Adsorptive stripping<br />

voltammetric determination of hydroquinone using an electrochemically pretreated glassy<br />

carbon electrode. Pakistan Journal of Environmental and Analytical Chemistry, 2008, 9,<br />

110-117.<br />

3. GC-MS Quantification of Fatty Acid Profile including trans FA in the Locally Manufactured<br />

Margarines of Pakistan. Aftab Kandhro, S. T. H Sherazi, S. A. Mahesar, M. I. Bhanger, M.<br />

Younis Talpur and Abdul Rauf Khaskheli; Food Chemistry, 2008, 109, 1, 207-211.<br />

133


4. Simultaneous assessment of zinc, lead, cadmium and copper in poultry feeds by differential<br />

pulse anodic stripping voltammetry. S. A. Mahesar, S. T. H Sherazi, A. Niaz, M. I. Bhanger,<br />

Sirajuudin and Abdul Rauf Khaskheli; Food and Chemical Toxicology, 2010, 48, 2357-<br />

2360.<br />

5. Fast voltammetric assay of water soluble phthalates in bottled and coolers water. Munawar<br />

Saeed, Sirajuddin, Abdul Niaz, Afzal Shah, H.I Afridi and Abdul Rauf; Analytical<br />

Methods, 2010, 2, 844-850.<br />

6. Assessment of Azithromycin in pharmaceutical formulation by FTIR transmission<br />

spectroscopy. M. Ali, S.T.H Sherazi, S.A. Mahesar and Abdul Rauf. (Submitted in Journal<br />

of Brazillian chemical Society).<br />

7. Appraisal of Erythromycin in pharmaceutical formulation by FTIR transmission<br />

spectroscopy. M. Ali, S.T.H Sherazi, S.A. Mahesar and Abdul Rauf. (Submitted in Pakistan<br />

Journal of <strong>Ph</strong>armaceutical Sciences).<br />

134

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!